88 results on '"Kwok-Ming Yao"'
Search Results
2. Involvement of FSP1-CoQ10-NADH and GSH-GPx-4 pathways in retinal pigment epithelium ferroptosis
- Author
-
Ming Yang, Michelle Grace Tsui, Jessica Kwan Wun Tsang, Rajesh Kumar Goit, Kwok-Ming Yao, Kwok-Fai So, Wai-Ching Lam, and Amy Cheuk Yin Lo
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Retinal pigment epithelium (RPE) degeneration plays an important role in a group of retinal disorders such as retinal degeneration (RD) and age-related macular degeneration (AMD). The mechanism of RPE cell death is not yet fully elucidated. Ferroptosis, a novel regulated cell death pathway, participates in cancer and several neurodegenerative diseases. Glutathione peroxidase 4 (GPx-4) and ferroptosis suppressor protein 1 (FSP1) have been proposed to be two main regulators of ferroptosis in these diseases; yet, their roles in RPE degeneration remain elusive. Here, we report that both FSP1-CoQ10-NADH and GSH-GPx-4 pathways inhibit retinal ferroptosis in sodium iodate (SIO)-induced retinal degeneration pathologies in human primary RPE cells (HRPEpiC), ARPE-19 cell line, and mice. GSH-GPx-4 signaling was compromised after a toxic injury caused by SIO, which was aggravated by silencing GPx-4, and ferroptosis inhibitors robustly protected RPE cells from the challenge. Interestingly, while inhibition of FSP1 caused RPE cell death, which was aggravated by SIO exposure, overexpression of FSP1 effectively protected RPE cells from SIO-induced injury, accompanied by a significant down-regulation of CoQ10/NADH and lipid peroxidation. Most importantly, in vivo results showed that Ferrostatin-1 not only remarkably alleviated SIO-induced RPE cell loss, photoreceptor death, and retinal dysfunction but also significantly ameliorated the compromised GSH-GPx-4 and FSP1-CoQ10-NADH signaling in RPE cells isolated from SIO-induced RPE degeneration. These data describe a distinct role for ferroptosis in controlling RPE cell death in vitro and in vivo and may provide a new avenue for identifying treatment targets for RPE degeneration.
- Published
- 2022
- Full Text
- View/download PDF
3. Novel Nuclear Partnering Role of EPS8 With FOXM1 in Regulating Cell Proliferation
- Author
-
Adaline Wan Ling Ngan, Michelle Grace Tsui, Danny Hon Fai So, Wai Ying Leung, David W. Chan, and Kwok-Ming Yao
- Subjects
FOXM1 ,EPS8 ,nuclear export signal ,CCNB1 ,cell proliferation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
One hallmark of cancer cells is sustaining proliferative signaling that leads to uncontrolled cell proliferation. Both the Forkhead box (FOX) M1 transcription factor and the Epidermal Growth Factor (EGF) receptor Pathway Substrate 8 (EPS8) are known to be activated by mitogenic signaling and their levels upregulated in cancer. Well-known to regulate Rac-mediated actin remodeling at the cell cortex, EPS8 carries a nuclear localization signal but its possible nuclear role remains unclear. Here, we demonstrated interaction of FOXM1 with EPS8 in yeast two-hybrid and immunoprecipitation assays. Immunostaining revealed co-localization of the two proteins during G2/M phase of the cell cycle. EPS8 became nuclear localized when CRM1/Exportin 1-dependent nuclear export was inhibited by Leptomycin B, and a functional nuclear export signal could be identified within EPS8 using EGFP-tagging and site-directed mutagenesis. Downregulation of EPS8 using shRNAs suppressed expression of FOXM1 and the FOXM1-target CCNB1, and slowed down G2/M transition in cervical cancer cells. Chromatin immunoprecipitation analysis indicated recruitment of EPS8 to the CCNB1 and CDC25B promoters. Taken together, our findings support a novel partnering role of EPS8 with FOXM1 in the regulation of cancer cell proliferation and provides interesting insight into future design of therapeutic strategy to inhibit cancer cell proliferation.
- Published
- 2019
- Full Text
- View/download PDF
4. Melatonin Inhibits Androgen Receptor Splice Variant-7 (AR-V7)-Induced Nuclear Factor-Kappa B (NF-κB) Activation and NF-κB Activator-Induced AR-V7 Expression in Prostate Cancer Cells: Potential Implications for the Use of Melatonin in Castration-Resistant Prostate Cancer (CRPC) Therapy
- Author
-
Vincent Wing Sun Liu, Wing Lung Yau, Chun Wai Tam, Kwok-Ming Yao, and Stephen Yuen Wing Shiu
- Subjects
melatonin ,androgen receptor splice variant-7 ,nuclear factor-kappa B ,castration-resistant prostate cancer ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
A major current challenge in the treatment of advanced prostate cancer, which can be initially controlled by medical or surgical castration, is the development of effective, safe, and affordable therapies against progression of the disease to the stage of castration resistance. Here, we showed that in LNCaP and 22Rv1 prostate cancer cells transiently overexpressing androgen receptor splice variant-7 (AR-V7), nuclear factor-kappa B (NF-κB) was activated and could result in up-regulated interleukin (IL)-6 gene expression, indicating a positive interaction between AR-V7 expression and activated NF-κB/IL-6 signaling in castration-resistant prostate cancer (CRPC) pathogenesis. Importantly, both AR-V7-induced NF-κB activation and IL-6 gene transcription in LNCaP and 22Rv1 cells could be inhibited by melatonin. Furthermore, stimulation of AR-V7 mRNA expression in LNCaP cells by betulinic acid, a pharmacological NF-κB activator, was reduced by melatonin treatment. Our data support the presence of bi-directional positive interactions between AR-V7 expression and NF-κB activation in CRPC pathogenesis. Of note, melatonin, by inhibiting NF-κB activation via the previously-reported MT1 receptor-mediated antiproliferative pathway, can disrupt these bi-directional positive interactions between AR-V7 and NF-κB and thereby delay the development of castration resistance in advanced prostate cancer. Apparently, this therapeutic potential of melatonin in advanced prostate cancer/CRPC management is worth translation in the clinic via combined androgen depletion and melatonin repletion.
- Published
- 2017
- Full Text
- View/download PDF
5. Involvement of FSP1-CoQ
- Author
-
Ming, Yang, Michelle Grace, Tsui, Jessica Kwan Wun, Tsang, Rajesh Kumar, Goit, Kwok-Ming, Yao, Kwok-Fai, So, Wai-Ching, Lam, and Amy Cheuk Yin, Lo
- Subjects
Mice ,Oxidative Stress ,Ubiquinone ,Retinal Degeneration ,Animals ,Ferroptosis ,S100 Calcium-Binding Protein A4 ,Retinal Pigment Epithelium ,NAD ,Phospholipid Hydroperoxide Glutathione Peroxidase ,Glutathione ,Signal Transduction - Abstract
Retinal pigment epithelium (RPE) degeneration plays an important role in a group of retinal disorders such as retinal degeneration (RD) and age-related macular degeneration (AMD). The mechanism of RPE cell death is not yet fully elucidated. Ferroptosis, a novel regulated cell death pathway, participates in cancer and several neurodegenerative diseases. Glutathione peroxidase 4 (GPx-4) and ferroptosis suppressor protein 1 (FSP1) have been proposed to be two main regulators of ferroptosis in these diseases; yet, their roles in RPE degeneration remain elusive. Here, we report that both FSP1-CoQ
- Published
- 2021
6. PDZD2 and Primary Open‐Angle Glaucoma: Functional Study of the Silencer‐Like Region in NT2 Neuronal Progenitor Cells
- Author
-
Michelle Grace Tsui, Amy C. Y. Lo, Kwok-Ming Yao, and Yan Yi Lam
- Subjects
Open angle glaucoma ,Genetics ,Progenitor cell ,Biology ,Silencer ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2021
7. Novel Nuclear Partnering Role of EPS8 With FOXM1 in Regulating Cell Proliferation
- Author
-
Kwok-Ming Yao, Michelle Grace Tsui, Danny Hon Fai So, Wai Ying Leung, Adaline Wan Ling Ngan, and David W. Chan
- Subjects
0301 basic medicine ,Cancer Research ,Chemistry ,Cell growth ,FOXM1 ,nuclear export signal ,CCNB1 ,EPS8 ,Cell cycle ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Cell biology ,03 medical and health sciences ,cell proliferation ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Epidermal growth factor ,030220 oncology & carcinogenesis ,Cancer cell ,Nuclear export signal ,Chromatin immunoprecipitation ,Transcription factor ,Nuclear localization sequence ,Original Research - Abstract
One hallmark of cancer cells is sustaining proliferative signaling that leads to uncontrolled cell proliferation. Both the Forkhead box (FOX) M1 transcription factor and the Epidermal Growth Factor (EGF) receptor Pathway Substrate 8 (EPS8) are known to be activated by mitogenic signaling and their levels upregulated in cancer. Well-known to regulate Rac-mediated actin remodeling at the cell cortex, EPS8 carries a nuclear localization signal but its possible nuclear role remains unclear. Here, we demonstrated interaction of FOXM1 with EPS8 in yeast two-hybrid and immunoprecipitation assays. Immunostaining revealed co-localization of the two proteins during G2/M phase of the cell cycle. EPS8 became nuclear localized when CRM1/Exportin 1-dependent nuclear export was inhibited by Leptomycin B, and a functional nuclear export signal could be identified within EPS8 using EGFP-tagging and site-directed mutagenesis. Downregulation of EPS8 using shRNAs suppressed expression of FOXM1 and the FOXM1-target CCNB1, and slowed down G2/M transition in cervical cancer cells. Chromatin immunoprecipitation analysis indicated recruitment of EPS8 to the CCNB1 and CDC25B promoters. Taken together, our findings support a novel partnering role of EPS8 with FOXM1 in the regulation of cancer cell proliferation and provides interesting insight into future design of therapeutic strategy to inhibit cancer cell proliferation.
- Published
- 2019
8. Secreted PDZD2 exerts an insulinotropic effect on INS-1E cells by a PKA-dependent mechanism
- Author
-
Joe Cho Yan Chan, Kwok-Ming Yao, Michelle Grace Tsui, Danny Hon Fai So, and Phyllis Siu Wai Tsang
- Subjects
0301 basic medicine ,endocrine system ,030209 endocrinology & metabolism ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,RNA interference ,Insulin-Secreting Cells ,Insulin Secretion ,Gene expression ,Animals ,Insulin ,RNA, Small Interfering ,Molecular Biology ,Gene ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Glucose Transporter Type 2 ,Secretory Pathway ,biology ,Chemistry ,Cyclic AMP-Dependent Protein Kinases ,Rats ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,biology.protein ,GLUT2 ,Phosphorylation ,PDX1 ,RNA Interference ,Cell Adhesion Molecules ,Nuclear localization sequence ,Intracellular ,Signal Transduction - Abstract
Secreted PDZD2 (sPDZD2) is a signaling molecule generated upon proteolytic processing of the multi-PDZ-containing protein PDZD2. Previous analysis of gene-trap mice deficient in the synthesis of full-length PDZD2, but not the secreted form, revealed a role of PDZD2 in the regulation of glucose-stimulated insulin secretion. Here, using the pancreatic INS-1E β cells as in vitro model, we showed that depletion of PDZD2/sPDZD2 by RNA interference suppressed the expression of β-cell genes Ins1, Glut2 and MafA whereas treatment with recombinant sPDZD2 rescued the suppressive effect. Similar to GLP-1, sPDZD2 stimulated intracellular cAMP levels, activated β-cell gene expression in a PKA-dependent manner and induced the phosphorylation and nuclear localization of PDX1. Depletion of PDX1 inhibited the sPDZD2 insulinotropic effect, which could also be demonstrated in mouse islets. In summary, our findings are consistent with sPDZD2 serving a signaling function in regulating β-cell gene expression.
- Published
- 2020
9. Injectable cell-encapsulating composite alginate-collagen platform with inducible termination switch for safer ocular drug delivery
- Author
-
Amy Man Wah Chu, Francisca S. Y. Wong, Kwok-Ming Yao, Barbara P. Chan, Amy C. Y. Lo, and KK Tsang
- Subjects
Male ,Alginates ,Cell Survival ,Biophysics ,Bioengineering ,Apoptosis ,02 engineering and technology ,Pharmacology ,Biomaterials ,03 medical and health sciences ,Drug Delivery Systems ,Neurotrophic factors ,In vivo ,Glial cell line-derived neurotrophic factor ,medicine ,Animals ,Humans ,Viability assay ,Glial Cell Line-Derived Neurotrophic Factor ,030304 developmental biology ,Cell Proliferation ,Doxycycline ,Caspase 7 ,0303 health sciences ,biology ,Chemistry ,Caspase 3 ,Retinal Degeneration ,021001 nanoscience & nanotechnology ,In vitro ,Rats ,HEK293 Cells ,Mechanics of Materials ,Drug delivery ,Ceramics and Composites ,biology.protein ,Microscopy, Electron, Scanning ,Collagen ,0210 nano-technology ,Retinitis Pigmentosa ,medicine.drug - Abstract
Effective retinal drug delivery remains a challenge for treating vision-threatening diseases. Encapsulated-cell therapy (ECT) can provide local drug delivery without repeated invasive injections but is plagued by unsteady performance and biosafety issues. Here, an injectable composite alginate-collagen (CAC) ECT gel with a Tet-on inducible pro-caspase 8 mechanism that acted as an orally-inducible biosafety switch was developed for safer drug delivery. The optimised gels (2 mg/ml collagen, 1.5% high molecular weight alginate and 50,000 cells/gel) could be effectively terminated in vitro (≥20 pg/ml Doxycycline) and in vivo (1 mg/ml oral Doxycycline after 48 h). Also, they displayed effective proliferation control and continuous delivery of bioactive glial-cell derived neurotrophic factor (GDNF) with no significant gel degradation in vitro and in rat vitreous. Most importantly, intravitreally injected gels demonstrated therapeutic efficacy in Royal College of Surgeons rats with degenerating retina in reducing photoreceptor apoptosis and retina function loss. Furthermore, double gel injections into the same eye yielded better outcomes without compromising gel viability. Retrieved gels showed no host-tissue attachment or cell-protrusion 6 months post-implantation. The CAC ECT system exhibited mechanical stability, good encapsulation power, cell viability support, multiplexed GDNF dosage, and compatibility with different cell types (HEK293 and ARPE-19) without immunosuppressant, making it an attractive, safe and well-controlled platform for treating various eye diseases.
- Published
- 2018
10. Modification of N staging systems for penile cancer: a more precise prediction of prognosis
- Author
-
Q. W. Mi, Fangjian Zhou, H. Han, Jian Chen, P. Chen, Zhiwei Liu, Z. K. Qin, Y. Li, Kwok-Ming Yao, Z. S. Li, and B. Wang
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Penile Neoplasm ,lymph node excision ,Prostate cancer ,Breast cancer ,penile neoplasms ,Medicine ,Penile cancer ,Humans ,Lung cancer ,neoplasm staging ,Molecular Diagnostics ,Aged ,Proportional Hazards Models ,Cervical cancer ,Aged, 80 and over ,lymph node metastasis ,business.industry ,Neoplasms, Second Primary ,Middle Aged ,medicine.disease ,Surgery ,Treatment Outcome ,Oncology ,Lymphatic Metastasis ,Radiology ,Lymph Nodes ,prognosis ,Skin cancer ,business ,Corrigendum - Abstract
Background: The tumour-node-metastasis (TNM) classification is the most widely used tool for penile cancer. However, the current system is based on few studies and has been unchanged since 2009. We determined whether a modified pathological N staging system that incorporates the laterality and number of lymph node metastases (LNMs) increases the accuracy of the results in predicting survival compared with the 7th edition of the pathological N staging system of the American Joint Committee on Cancer (AJCC) for penile cancer. Methods: The clinical and histopathologic data from 111 patients with penile cancer with LNMs were analysed. Univariate and multivariate Cox proportional hazard regression analyses were used to determine the impact of the clinical and pathological factors on disease-specific survival of these patients. The predictive accuracy was further assessed using the concordance index. Results: According to the 7th edition of the pathological N classification, the 3-year disease-specific survival (DSS) rates for patients with pN1, pN2, and pN3 disease are 89.6%, 65.9%, and 33.6%, respectively (PN1–N2=0.030, PN2–N3
- Published
- 2015
11. Elevated TAK1 augments tumor growth and metastatic capacities of ovarian cancer cells through activation of NF-κB signaling
- Author
-
Hermit Wai-Man Tang, Vincent W.S. Liu, Kwok-Ming Yao, Iris J. Liu, Hextan Y.S. Ngan, Lei Shi, David W. Chan, Thomas Ho-Yin Leung, Karen K. L. Chan, Patty C. H. Cai, and Judy Wai Ping Yam
- Subjects
TAK1 ,Blotting, Western ,Mice, Nude ,Biology ,Transfection ,medicine.disease_cause ,Metastasis ,Mice ,RNA interference ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Ovarian Neoplasms ,Mice, Inbred BALB C ,MAP kinase kinase kinase ,Reverse Transcriptase Polymerase Chain Reaction ,Kinase ,NF-kappa B ,high-grade tumor ,MAP Kinase Kinase Kinases ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,ovarian cancer ,Oncology ,NF-κB signaling ,Cancer research ,Female ,Signal transduction ,Ovarian cancer ,Carcinogenesis ,Research Paper ,Signal Transduction ,Transforming growth factor - Abstract
// Patty C.H. Cai 1 , Lei Shi 2 , Vincent W.S. Liu 1 , Hermit W.M. Tang 1 , Iris J. Liu 1 , Thomas H.Y. Leung 1 , Karen K.L. Chan 1 , Judy W.P. Yam 3 , Kwok-Ming Yao 2 , Hextan Y.S. Ngan 1 and David W. Chan 1 1 Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R.China 2 Department of Biochemistry, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R.China 3 Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R.China Correspondence: Hextan YS Ngan, email: // David W Chan, email: // Keywords : TAK1, NF-κB signaling, ovarian cancer, high-grade tumor Received : May 07, 2014 Accepted : July 26, 2014 Published : July 27, 2014 Abstract Transforming growth factor (TGF)-β-activating kinase 1 (TAK1) is a serine/threonine kinase which is frequently associated with human cancer progression. However, its functional role in tumorigenesis is still controversial. Here, we report that TAK1 enhances the oncogenic capacity of ovarian cancer cells through the activation of NF-κB signaling. We found that TAK1 is frequently upregulated and significantly associated with high-grade and metastatic ovarian cancers. Mechanistic studies showed that Ser412 phosphorylation is required for TAK1 in activating NF-κB signaling and promotes aggressiveness of ovarian cancer cells. Conversely, suppression of TAK1 activity by point mutation at Ser412, RNAi mediated gene knockdown or TAK1 specific inhibitor ((5Z) -7-Oxozeaenol) remarkably impairs tumor growth and metastasis in ovarian cancer in vitro and in vivo . Our study underscores the importance of targeting TAK1 as a promising therapeutic approach to counteract the ovarian cancer progression.
- Published
- 2014
12. Melatonin MT1 receptor-induced transcriptional up-regulation of p27Kip1 in prostate cancer antiproliferation is mediated via inhibition of constitutively active nuclear factor kappa B (NF-κB): potential implications on prostate cancer chemoprevention and
- Author
-
Kwok-Ming Yao, Chun W. Tam, Vincent W.S. Liu, Stephen Y. W. Shiu, and Wai Y. Leung
- Subjects
Male ,Down-Regulation ,Biology ,Melatonin ,chemistry.chemical_compound ,Endocrinology ,Cell Line, Tumor ,LNCaP ,medicine ,Humans ,Protein kinase A ,Transcription factor ,Protein kinase C ,Activator (genetics) ,Receptor, Melatonin, MT1 ,NF-kappa B ,Prostatic Neoplasms ,NF-κB ,Molecular biology ,Up-Regulation ,Androgen receptor ,chemistry ,Cancer research ,Cyclin-Dependent Kinase Inhibitor p27 ,medicine.drug - Abstract
Our laboratory has recently demonstrated a melatonin MT1 receptor-mediated antiproliferative signaling mechanism in androgen receptor (AR)-positive prostate epithelial cells which involves up-regulation of p27(Kip1) through dual activation of Gα(s)/protein kinase A (PKA) and Gα(q)/protein kinase C (PKC) in parallel, and down-regulation of activated AR signaling via PKC stimulation. The aim of the present investigation was to identify the transcription factor that mediates melatonin's up-regulatory effect on p27(Kip1) in LNCaP and 22Rv1 prostate cancer cells. Deletion mapping and reporter assays of the p27(Kip1) promoter revealed that the putative melatonin-responsive transcription factor binds to a 116 base-pair region of the promoter sequence, which contains a potential nuclear factor kappa B (NF-κB) binding site. When the NF-κB binding site was abolished by site-directed mutagenesis, the stimulatory effect of melatonin on p27(Kip1) promoter activity was mitigated. Notably, melatonin inhibited the DNA binding of activated NF-κB via MT1 receptor-induced PKA and PKC stimulation. Furthermore, melatonin's up-regulatory effect on p27(Kip1) transcription and consequent cell antiproliferation were abrogated by NF-κB activator but mimicked by NF-κB inhibitor. The results indicate that inhibition of constitutively active NF-κB via melatonin MT1 receptor-induced dual activation of (Gα(s)) PKA and (Gα(q)) PKC can de-repress the p27(Kip1) promoter leading to transcriptional up-regulation of p27(Kip1). MT1 receptor-mediated inhibition of activated NF-κB signaling provides a novel mechanism supporting the use of melatonin in prostate cancer chemoprevention and therapy.
- Published
- 2012
13. LY294002 and Metformin Cooperatively Enhance the Inhibition of Growth and the Induction of Apoptosis of Ovarian Cancer Cells
- Author
-
Hextan Y.S. Ngan, Cuilan Li, Vincent Liu, Kwok-Ming Yao, and David W. Chan
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Programmed cell death ,Morpholines ,Blotting, Western ,Antineoplastic Agents ,Apoptosis ,AMP-Activated Protein Kinases ,Biology ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,Proliferating Cell Nuclear Antigen ,Biomarkers, Tumor ,Humans ,Enzyme Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Ovarian Neoplasms ,Phosphoinositide 3-kinase ,Caspase 3 ,Cell growth ,TOR Serine-Threonine Kinases ,RPTOR ,Obstetrics and Gynecology ,AMPK ,Drug Synergism ,Cell cycle ,Flow Cytometry ,Metformin ,Oncology ,Chromones ,Cancer research ,biology.protein ,Female ,Poly(ADP-ribose) Polymerases ,Tumor Suppressor Protein p53 ,Proto-Oncogene Proteins c-akt ,Acetyl-CoA Carboxylase - Abstract
BackgroundThe phosphoinositide 3 kinase (PI3K)/v-akt murine thymoma viral oncogene homolog (AKT)/mammalian target of rapamycin (mTOR) pathway is frequently aberrantly activated in ovarian cancer and confers the chemoresistant phenotype of ovarian cancer cells. LY294002 (PI3K inhibitor) and metformin (5′-adenosine monophosphate [AMP]-activated protein kinase [AMPK] activator) are 2 drugs that were known to inhibit mTOR expression through the AKT-dependent and AKT-independent pathways, respectively. In this study, we explored the effectiveness of LY294002 and metformin in combination on inhibition of ovarian cancer cell growth.MethodsWestern blotting was used to detect the changes of PI3K/AKT/mTOR and AMPK/acetyl-CoA carboxylase (ACC) signaling activities, cell cycle control, and apoptosis. Cell growth was evaluated by cell proliferation, colony formation, and soft agar assays. Flow cytometry was used to study cell cycle distribution and cell death upon drug treatment.ResultsOur study showed that LY294002 and metformin in combination could simultaneously enhance the repression of the PI3K/AKT/mTOR pathway and the activation of the AMPK/ACC pathway. The downstream target of AKT and AMPK, mTOR, was cooperatively repressed when the drugs were used together. The cell cycle regulatory factors, p53, p27, and p21, were up-regulated. On the other hand, caspase 3 and poly (ADP-ribose) polymerase activities involved in apoptosis were also activated. Cell growth assays indicated that LY294002 and metformin could effectively inhibit ovarian cancer cell growth. Flow cytometry analysis showed that the treatment of the 2 drugs mentioned above induced cell cycle arrest at G1 phase and increased sub-G1 apoptotic cells.ConclusionThe combinational use of LY294002 and metformin can enhance inhibition of the growth and induction of the apoptosis of ovarian cancer cells. Our results may provide significant insight into the future therapeutic regimens in ovarian cancer.
- Published
- 2012
14. Correlated evolution of transcription factors and their binding sites
- Author
-
Shu Yang, Pak Chung Sham, Xinran Li, Michael Q. Zhang, Kwok-Ming Yao, Junwen Wang, and Hari Krishna Yalamanchili
- Subjects
Statistics and Probability ,Genetics ,Supplementary data ,Regulation of gene expression ,Mutation ,Binding Sites ,Base Sequence ,Protein family ,DNA ,Biology ,medicine.disease_cause ,Biochemistry ,DNA sequencing ,Computer Science Applications ,Evolution, Molecular ,DNA binding site ,Computational Mathematics ,Computational Theory and Mathematics ,medicine ,Humans ,Regulatory Elements, Transcriptional ,Binding site ,Molecular Biology ,Transcription factor ,Transcription Factors - Abstract
Motivation:The interaction between transcription factor (TF) and transcription factor binding site (TFBS) is essential for gene regulation. Mutation in either the TF or the TFBS may weaken their interaction and thus result in abnormalities. To maintain such vital interaction, a mutation in one of the interacting partners might be compensated by a corresponding mutation in its binding partner during the course of evolution. Confirming this co-evolutionary relationship will guide us in designing protein sequences to target a specific DNA sequence or in predicting TFBS for poorly studied proteins, or even correcting and rescuing disease mutations in clinical applications. Results: Based on six, publicly available, experimentally validated TF–TFBS binding datasets for the basic Helix–Loop–Helix (bHLH) family, Homeo family, High-Mobility Group (HMG) family and Transient Receptor Potential channels (TRP) family, we showed that the evolutions of the TFs and their TFBSs are significantly correlated across eukaryotes. We further developed a mutual information-based method to identify co-evolved protein residues and DNA bases. This research sheds light on the dynamic relationship between TF and TFBS during their evolution. The same principle and strategy can be applied to co-evolutionary studies on protein–DNA interactions in other protein families. Availability: All the datasets, scripts and other related files have been made freely available at: http://jjwanglab.org/co-evo. Contact: junwen@uw.edu Supplementary Information: Supplementary data are available at Bioinformatics online.
- Published
- 2011
15. Zic2 synergistically enhances Hedgehog signalling through nuclear retention of Gli1 in cervical cancer cells
- Author
-
Annie N.Y. Cheung, Kwok-Ming Yao, Ling Yang Leung, David W. Chan, Vincent W.S. Liu, Karen K. L. Chan, and Hextan Y.S. Ngan
- Subjects
Uterine Cervical Neoplasms ,Adenocarcinoma ,Zinc Finger Protein GLI1 ,Pathology and Forensic Medicine ,GLI1 ,Cell Line, Tumor ,medicine ,Humans ,Hedgehog Proteins ,RNA, Messenger ,Nuclear protein ,Transcription factor ,Hedgehog ,Cell Line, Transformed ,Cell Nucleus ,integumentary system ,biology ,Cell growth ,Nuclear Proteins ,Gene Expression Regulation, Neoplastic ,Cell nucleus ,medicine.anatomical_structure ,Cancer research ,biology.protein ,Female ,Signal transduction ,Signal Transduction ,Transcription Factors - Abstract
Aberrant activation of Hedgehog (Hh) signalling has been implicated in the pathogenesis of human cancers. However, the cognate molecular mechanisms contributing to this disregulated pathway are incompletely understood. In this study, we showed that Zic2 was frequently over-expressed and associated with high-grade cervical cancer (p = 0.032), high levels of Gli1 (p < 0.001) and CyclinD1 (p < 0.001) by immunohistochemical and quantitative RT-PCR analyses. Further biochemical studies using luciferase reporter, co-immunoprecipitation, subcellular fractionation and immunofluorescence analyses demonstrated that Zic2 can physically interact with Gli1 and retain it in the nucleus, which in turn increases Gli-mediated transcriptional activity. Gain- and loss-of-function analyses of Zic2 showed that Zic2 could increase Hh signalling activity, cell proliferation and anchorage-independent growth ability in cervical cancer cells. Conversely, deletion of the zinc finger domain at C-terminus of Zic2 significantly abrogated its interaction with Gli1, the retention of Gli1 in the nucleus, effects on Hh signalling activity and oncogenic properties in cervical cancer cells. Our findings suggest that Zic2 is a positive modulator increasing Gli1 transcriptional and oncogenic activity by retaining Gli1 in the nucleus of cervical cancer cells.
- Published
- 2011
16. Signal transduction of receptor-mediated antiproliferative action of melatonin on human prostate epithelial cells involves dual activation of Gαs and Gαq proteins
- Author
-
Chun W. Tam, Stephen Y. W. Shiu, Bo Pang, and Kwok-Ming Yao
- Subjects
medicine.medical_specialty ,medicine.drug_class ,G protein ,Biology ,Receptor antagonist ,Cell biology ,Melatonin ,Endocrinology ,Internal medicine ,medicine ,Signal transduction ,Luzindole ,Receptor ,Protein kinase A ,Protein kinase C ,medicine.drug - Abstract
Melatonin has been shown to inhibit the proliferation of malignant and transformed human prostate epithelial cells by transcriptional up-regulation of p27(Kip1) expression via MTNR1A receptor-mediated activation of protein kinase A (PKA) and protein kinase C (PKC) in parallel. Given that melatonin MTNR1A receptor is a G protein-coupled receptor, this study was conducted to identify the specific G proteins that mediate the antiproliferative action of melatonin on human prostate epithelial cells. In 22Rv1 and RWPE-1 cells, knockdown of either Gα(s) or Gα(q) , but not Gα(i2) expression by RNA interference, abrogated the effects of melatonin on p27(Kip1) and cell proliferation. Conversely, cellular overexpression of activated mutants of Gα(s) and Gα(q) in 22Rv1 and RWPE-1 cells mimicked the effects of melatonin on prostate epithelial cell antiproliferation by increasing p27(Kip1) expression through downstream activation of PKA and PKC in parallel. Moreover, melatonin or 2-iodomelatonin induced elevation of adenosine-3',5'-cyclic monophosphate (cAMP) in 22Rv1 and RWPE-1 cells. The effects of 2-iodomelatonin on cAMP were blocked by the nonselective MTNR1A/MTNR1B receptor antagonist luzindole but were not affected by the selective MTNR1B receptor antagonist 4-phenyl-2-propionamidotetraline (4-P-PDOT). Furthermore, knockdown of Gα(s) mitigated the stimulatory effects of 2-iodomelatonin on cAMP. Collectively, the data demonstrated, for the first time, functional coupling of MTNR1A receptor to Gα(s) in cancerous or transformed human cells expressing endogenous melatonin receptors. Our results also showed that dual activation of Gα(s) and Gα(q) proteins is involved in the signal transduction of MTNR1A receptor-mediated antiproliferative action of melatonin on human prostate epithelial cells.
- Published
- 2010
17. A multi PDZ-domain protein Pdzd2 contributes to functional expression of sensory neuron-specific sodium channel NaV1.8
- Author
-
Misbah Malik-Hall, John N. Wood, Francois Rugiero, Mark D. Baker, Dongmin Shao, Kwok-Ming Yao, Kathryn S.E. Cheah, Kenji Okuse, Bjarke Abrahamsen, and W.-Y.Louisa Poon
- Subjects
Sensory Receptor Cells ,Sodium Channels - genetics - metabolism ,Recombinant Fusion Proteins ,Nerve Tissue Proteins - genetics - metabolism ,Molecular Sequence Data ,PDZ domain ,Pain ,PDZ Domains ,Nerve Tissue Proteins ,Sensory system ,Biology ,Article ,Sodium Channels ,NAV1.8 Voltage-Gated Sodium Channel ,Mice ,Sensory Receptor Cells - cytology - metabolism ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Adaptor Proteins, Signal Transducing - genetics - metabolism ,0302 clinical medicine ,Downregulation and upregulation ,Ganglia, Spinal ,Two-Hybrid System Techniques ,Animals ,Amino Acid Sequence ,RNA, Small Interfering ,Rats, Wistar ,Molecular Biology ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,Pain Measurement ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Sodium channel ,NAV1.7 Voltage-Gated Sodium Channel ,Cell Biology ,Rats ,3. Good health ,Nociception ,NAV1 ,Cell Adhesion Molecules ,Sequence Alignment ,Neuroscience ,030217 neurology & neurosurgery ,Intracellular ,Protein Binding - Abstract
The voltage-gated sodium channel NaV1.8 is expressed exclusively in nociceptive sensory neurons and plays an important role in pain pathways. NaV1.8 cannot be functionally expressed in non-neuronal cells even in the presence of β-subunits. We have previously identified Pdzd2, a multi PDZ-domain protein, as a potential interactor for NaV1.8. Here we report that Pdzd2 binds directly to the intracellular loops of NaV1.8 and NaV1.7. The endogenous NaV1.8 current in sensory neurons is inhibited by antisense- and siRNA-mediated downregulation of Pdzd2. However, no marked change in pain behaviours is observed in Pdzd2-decificent mice. This may be due to compensatory upregulation of p11, another regulatory factor for NaV1.8, in dorsal root ganglia of Pdzd2-deficient mice. These findings reveal that Pdzd2 and p11 play collaborative roles in regulation of NaV1.8 expression in sensory neurons. © 2009 Elsevier Inc. All rights reserved., link_to_OA_fulltext
- Published
- 2009
18. PDZ-Domain Containing-2 (PDZD2) Drives the Maturity of Human Fetal Pancreatic Progenitor-Derived Islet-Like Cell Clusters With Functional Responsiveness Against Membrane Depolarization
- Author
-
Kwok-Ming Yao, Kwan Keung Leung, Po Sing Leung, Wing-Hung Ko, Tse Kin Lau, and Po Man Suen
- Subjects
medicine.medical_specialty ,Calcium Channels, L-Type ,medicine.medical_treatment ,Population ,Stimulation ,Biology ,Membrane Potentials ,Potassium Chloride ,Fetus ,Downregulation and upregulation ,Insulin-Secreting Cells ,Internal medicine ,Insulin Secretion ,medicine ,Humans ,Insulin ,Progenitor cell ,education ,Cells, Cultured ,Adaptor Proteins, Signal Transducing ,geography ,education.field_of_study ,Ion Transport ,geography.geographical_feature_category ,C-Peptide ,Voltage-dependent calcium channel ,Stem Cells ,Calcium channel ,Cell Differentiation ,Cell Biology ,Hematology ,Islet ,Antigens, Differentiation ,Neoplasm Proteins ,Up-Regulation ,Endocrinology ,Calcium ,Cell Adhesion Molecules ,Developmental Biology - Abstract
We recently reported the isolation and characterization of a population of pancreatic progenitor cells (PPCs) from early trimester human fetal pancreata. The PPCs, being the forerunners of adult pancreatic cell lineages, were amenable to growth and differentiation into insulin-secreting islet-like cell clusters (ICCs) upon stimulation by adequate morphogens. Of note, a novel morphogenic factor, PDZ-domain containing-2 (PDZD2) and its secreted form (sPDZD2) were ubiquitously expressed in the PPCs. Our goals for this study were to evaluate the potential role of sPDZD2 in stimulating PPC differentiation and to establish the optimal concentration for such stimulation. We found that 10(-9)M sPDZD2 promoted PPC differentiation, as evidenced by the upregulation of the pancreatic endocrine markers (PDX-1, NGN3, NEURO-D, ISL-1, NKX 2.2, NKX 6.1) and INSULIN mRNA. Inhibited endogenous production of sPDZD2 suppressed expression of these factors. Secreted PDZD2 treatment significantly elevated the C-peptide content of the ICCs and increased the basal rate of insulin secretion. However, they remained unresponsive to glucose stimulation, reflected by a minimal increase in GLUT-2 and GLUCOKINASE mRNA expression. Interestingly, sPDZD2 treatment induced increased expression of the L-type voltage-gated calcium channel (Ca(v)1.2) in the ICCs, triggering calcium ion influx under KCl stimulation and conferring an ability to secrete insulin in response to KCl. Pancreatic progenitor cells from 10- and 13-week fetal pancreata showed peak expression of endogenous sPDZD2, implying that sPDZD2 has a specific role in islet development during the first trimester. In conclusion, our data suggest that sPDZD2 promotes functional maturation of human fetal PPC-derived ICCs, thus enhancing its transplanting potentials.
- Published
- 2009
19. PDZ-domain containing-2 (PDZD2) is a novel factor that affects the growth and differentiation of human fetal pancreatic progenitor cells
- Author
-
Po Man Suen, T.K. Lau, Zhang Ying, Chunlin Zou, Juliana C.N. Chan, Kwok-Ming Yao, and Po Sing Leung
- Subjects
Blotting, Western ,Population ,Enteroendocrine cell ,Biology ,Biochemistry ,Cell Line ,Downregulation and upregulation ,medicine ,Humans ,Progenitor cell ,education ,Pancreas ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,education.field_of_study ,C-Peptide ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Differentiation ,Cell Biology ,Immunohistochemistry ,Molecular biology ,Recombinant Proteins ,Neoplasm Proteins ,Transplantation ,medicine.anatomical_structure ,Cell culture ,Cancer research ,Stem cell ,Cell Adhesion Molecules - Abstract
Early-trimester human fetal pancreas is a promising potential source of pancreatic progenitor cells. However, the ethical controversy associated with the source of these cells, and technical difficulties associated with their differentiation into insulin-producing cells have limited both their availability and utility. This study aimed to characterize a population of pancreatic progenitor cells (PPCs) isolated from human fetus and describe the effects of a novel factor, PDZ-domain containing-2 (PDZD2), and its secreted form (sPDZD2), on PPC proliferation and differentiation. In particular, we examined and characterized the expression of several stem cell (nestin, ABCG2, c-kit), growth and differentiation markers (GLP-1R, c-met, erbB1), and PDZD2 in PPCs by RT-PCR, Western blot, and immunocytochemistry. We also examined the effects of sPDZD2 on PPC proliferation and differentiation by examining BrdU incorporation, MTT, cell number, and real-time PCR as well as ELISA. PPCs were isolated, cultured and characterized from human fetal pancreas. PDZD2 and sPDZD2 were detected at high levels in both human fetal pancreas and in PPCs. sPDZD2 acted as a potent mitogen on PPCs, and inhibited the differentiation of PPC-derived islet-like cell-clusters (ICCs), evidenced by the downregulation of Isl-1, Pdx-1, and insulin mRNA levels. sPDZD2 treatment also reduced levels of C-peptide in ICCs. These results show that a novel pancreatic developmental factor, PDZD2, is sufficient to promote the proliferation of human fetal PPCs while limiting differentiation of ICCs into islet/endocrine cells. Findings from this study will contribute to the development of improved methods for islet transplantation therapy in the treatment of diabetes.
- Published
- 2008
20. Over-expression of FOXM1 transcription factor is associated with cervical cancer progression and pathogenesis
- Author
-
Hextan Y.S. Ngan, Kwok-Ming Yao, S. Y. M. Yu, David W. Chan, Annie N.Y. Cheung, PM Chiu, and Vws Liu
- Subjects
Pathology ,medicine.medical_specialty ,Blotting, Western ,Gene Expression ,Uterine Cervical Neoplasms ,Cell Cycle Proteins ,Cervix Uteri ,Biology ,Cervical intraepithelial neoplasia ,Statistics, Nonparametric ,Pathology and Forensic Medicine ,Biomarkers, Tumor ,medicine ,Carcinoma ,Humans ,Cyclin B1 ,Cell Proliferation ,Neoplasm Staging ,Cervical cancer ,Cell growth ,Forkhead Box Protein M1 ,Cancer ,Forkhead Transcription Factors ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Tumor progression ,Carcinoma, Squamous Cell ,Cancer research ,FOXM1 ,Female ,Biomarkers - Abstract
The Forkhead Box M1 (FOXM1) transcription factor plays a crucial role in regulating expression of cell cycle genes which are essentially involved in cell proliferation, differentiation and transformation. Recent studies have reported that aberrant expression of FOXM1 in a variety of human cancers is associated with their aggressive behaviour. However, the functional significance of FOXM1 in human cervical cancer is not known. We have shown that FOXM1 was significantly over-expressed in cervical squamous cell carcinoma (SCC) compared to normal cervical epithelium immunohistochemically (p < 0.001). In addition, intratumoural FOXM1 positivity was increased in cervical intraepithelial neoplasia (CIN) and carcinoma, compared with that in normal epithelium, indicating that FOXM1 is involved in tumour progression. Indeed, this is supported by clinicopathological analysis that the over-expression of FOXM1 was significantly associated with tumour late stage (p = 0.012) and cell proliferation marker, Ki67 (p < 0.001). Functionally, enforced expression of FOXM1c in FOXM1-deficient cervical cancer cells (C33A) remarkably enhanced cell proliferation and anchorage-independent growth ability. Conversely, depletion of FOXM1 by RNA interference in FOXM1-over-expressing cervical cancer cells (SiHa) caused significant inhibition on cell proliferation and anchorage-independent growth ability on soft agar. This inhibitory phenomenon was associated with the reduced expressions of cyclin B1, cyclinD1 and cdc25B but increased expression of p27Kip1 and p21Cip1. Our findings suggest a role for FOXM1 in the development and pathogenesis of human cervical SCC. Copyright © 2008 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2008
21. The Forkhead box transcription factor FOXM1 is required for the maintenance of cell proliferation and protection against oxidative stress in human embryonic stem cells
- Author
-
Jing Qin, Y. Qin, C.T.D. Kwok, M.H. Leung, Kwok-Ming Yao, Jin Wang, and Y.L. Lee
- Subjects
Pluripotency ,0301 basic medicine ,Homeobox protein NANOG ,Chromatin Immunoprecipitation ,Cellular differentiation ,Human Embryonic Stem Cells ,Biology ,Chromatin immunoprecipitation-sequencing ,Cell Line ,03 medical and health sciences ,Humans ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Transcription factor ,reproductive and urinary physiology ,Cell Proliferation ,Medicine(all) ,Cyclin-dependent kinase 1 ,Cell growth ,Forkhead Box Protein M1 ,FOXM1 ,Nanog Homeobox Protein ,Cell Differentiation ,Cell Biology ,General Medicine ,Hydrogen Peroxide ,Sequence Analysis, DNA ,Cell cycle ,Embryonic stem cell ,Cell biology ,Oxidative Stress ,030104 developmental biology ,lcsh:Biology (General) ,Microscopy, Fluorescence ,embryonic structures ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,Octamer Transcription Factor-3 ,Cell Division ,Developmental Biology - Abstract
Human embryonic stem cells (hESCs) exhibit unique cell cycle structure, self-renewal and pluripotency. The Forkhead box transcription factor M1 (FOXM1) is critically required for the maintenance of pluripotency in mouse embryonic stem cells and mouse embryonal carcinoma cells, but its role in hESCs remains unclear. Here, we show that FOXM1 expression was enriched in undifferentiated hESCs and was regulated in a cell cycle-dependent manner with peak levels detected at the G2/M phase. Expression of FOXM1 did not correlate with OCT4 and NANOG during in vitro differentiation of hESCs. Importantly, knockdown of FOXM1 expression led to aberrant cell cycle distribution with impairment in mitotic progression but showed no profound effect on the undifferentiated state. Interestingly, FOXM1 depletion sensitized hESCs to oxidative stress. Moreover, genome-wide analysis of FOXM1 targets by ChIP-seq identified genes important for M phase including CCNB1 and CDK1, which were subsequently confirmed by ChIP and RNA interference analyses. Further peak set comparison against a differentiating hESC line and a cancer cell line revealed a substantial difference in the genomic binding profile of FOXM1 in hESCs. Taken together, our findings provide the first evidence to support FOXM1 as an important regulator of cell cycle progression and defense against oxidative stress in hESCs.
- Published
- 2015
22. Inhibition of Prostate Cancer Cell Growth by Human Secreted PDZ Domain-Containing Protein 2, a Potential Autocrine Prostate Tumor Suppressor
- Author
-
Stephen Y. W. Shiu, R. Y. M. Ma, Chun W. Tam, A. S. Cheng, and Kwok-Ming Yao
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Male ,medicine.medical_specialty ,PDZ domain ,Apoptosis ,Biology ,urologic and male genital diseases ,medicine.disease_cause ,Autocrine Communication ,Endocrinology ,DU145 ,Cell Line, Tumor ,Internal medicine ,LNCaP ,medicine ,Humans ,Autocrine signalling ,Adaptor Proteins, Signal Transducing ,Cell growth ,Tumor Suppressor Proteins ,Cell Cycle ,Intracellular Signaling Peptides and Proteins ,Prostatic Neoplasms ,Dipeptides ,Ketones ,Caspase Inhibitors ,Recombinant Proteins ,Neoplasm Proteins ,Cancer cell ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Cell Adhesion Molecules ,Cyclin-Dependent Kinase Inhibitor p27 - Abstract
A possible role of the PDZ domain-containing protein 2 (PDZD2) in prostate tumorigenesis has been suggested. Besides, PDZD2 is posttranslationally cleaved by a caspasedependent mechanism to form a secreted PDZ domaincontaining protein 2 (sPDZD2) with unknown functions in humans. In this study, we demonstrate the endogenous expression of PDZD2 and secretion of sPDZD2 in cancerous DU145, PC-3, 22Rv1, LNCaP, and immortalized RWPE-1 prostate epithelial cells. Inhibition of endogenous sPDZD2 production and secretion by DU145, PC-3, 22Rv1, and RWPE-1 cells via the caspase-3 inhibitor Z-DEVD-FMK resulted in increased cell proliferation, which was abrogated by treatment with exogenous recombinant sPDZD2. Whereas sPDZD2induced antiproliferation in DU145, PC-3, and 22Rv1 cells, it induced apoptosis in LNCaP cells. The data suggest that endogenous sPDZD2, produced by caspase-3-mediated cleavage from PDZD2, may function as a novel autocrine growth suppressor for human prostate cancer cells. The antiproliferative effect of sPDZD2 was apparently mediated through slowing the entry of DU145, PC-3, and 22Rv1 cells into the S phase of the cell cycle. In DU145 cells, this can be attributed to stimulated p53 and p21 CIP1/WAF1 expression by sPDZD2. On the otherhand,theapoptoticeffectofsPDZD2onLNCaPcellswas apparently mediated via p53-independent Bad stimulation. Together our results indicate the presence of p53-dependent and p53-independent PDZD2/sPDZD2 autocrine growth suppressive signaling pathways in human prostate cancer cells and suggest a novel therapeutic approach of harnessing the latent tumor-suppressive potential of an endogenous autocrine signaling protein like sPDZD2 to inhibit prostate cancer growth. (Endocrinology 147: 5023–5033, 2006)
- Published
- 2006
23. Interactions with p300 enhance transcriptional activation by the PDZ-domain coactivator Bridge-1
- Author
-
Jamie L. Volinic, Constanze Banz, Melissa K. Thomas, Jee H. Lee, and Kwok-Ming Yao
- Subjects
Transcriptional Activation ,Proteasome Endopeptidase Complex ,medicine.medical_specialty ,Transcription, Genetic ,Endocrinology, Diabetes and Metabolism ,Blotting, Western ,Biology ,Hydroxamic Acids ,Endocrinology ,Yeasts ,Internal medicine ,medicine ,Animals ,Immunoprecipitation ,Insulin ,Point Mutation ,p300-CBP Transcription Factors ,Cells, Cultured ,Glutathione Transferase ,Nuclear receptor co-repressor 2 ,PELP-1 ,Dose-Response Relationship, Drug ,YY1 ,Helix-Loop-Helix Motifs ,Proteins ,HDAC4 ,Molecular biology ,Protein Structure, Tertiary ,Cell biology ,Histone Deacetylase Inhibitors ,Nuclear receptor coactivator 1 ,Diabetes Mellitus, Type 1 ,PCAF ,GATAD2B ,Mutagenesis, Site-Directed ,Nuclear receptor coactivator 2 ,Adenovirus E1A Proteins ,Gene Deletion - Abstract
Transcriptional coactivators are essential mediators of signal amplification in the regulation of gene expression in response to hormones and extracellular signals. We previously identified Bridge-1 (PSMD9) as a PDZ-domain coregulator that augments insulin gene transcription via interactions with the basic helix-loop-helix transcription factors E12 and E47, and that increases transcriptional activation by the homeodomain transcription factor PDX-1. In these studies, we find that transcriptional activation by Bridge-1 can be regulated via interactions with the histone acetyltransferase and nuclear receptor coactivator p300. In transfection assays, transcriptional activation by Bridge-1 is increased by the inhibition of endogenous histone deacetylase activity with trichostatin A, indicating that the transcriptional activation function of Bridge-1 can be regulated by histone modifications. The exogenous expression of p300 enhances the transcriptional activation by Bridge-1 in a dose-dependent manner. In contrast, the sequestration of p300 by the overexpression of the adenoviral protein E1A, but not by an E1A mutant protein that is unable to interact with p300, suppresses the transcriptional activation by Bridge-1. We demonstrate that p300 and Bridge-1 proteins interact in immunopre-cipitation and glutathione-S-transferase (GST) pull-down assays. Bridge-1 interacts directly with multiple regions within p300 that encompass C/H1 or C/H2 cysteine- and histidine-rich protein interaction domains and the histone acetyltransferase domain. Deletion or point mutagenesis of the Bridge-1 PDZ domain substantially reduces transcriptional activation by Bridge-1 and interrupts interactions with p300. We propose that p300 interactions with Bridge-1 can augment the transcriptional activation of regulatory target genes by Bridge-1.
- Published
- 2005
24. Identification of cis-elements for ethylene and circadian regulation of the Solanum melongena gene encoding cysteine proteinase
- Author
-
Mee-Len Chye, Kwok-Ming Yao, Zeng-Fu Xu, and Reetika Rawat
- Subjects
5' Flanking Region ,Recombinant Fusion Proteins ,Transgene ,Molecular Sequence Data ,Mutant ,DNA Footprinting ,Electrophoretic Mobility Shift Assay ,Plant Science ,Biology ,Response Elements ,Organophosphorus Compounds ,Plant Growth Regulators ,Tobacco ,Genetics ,Deoxyribonuclease I ,Solanum melongena ,Nuclear protein ,Luciferases ,Promoter Regions, Genetic ,Gene ,Glucuronidase ,Plant Proteins ,Binding Sites ,Base Sequence ,DNase-I Footprinting ,Nuclear Proteins ,Promoter ,General Medicine ,Ethylenes ,Plants, Genetically Modified ,Molecular biology ,In vitro ,Circadian Rhythm ,Cysteine Endopeptidases ,Biochemistry ,Agronomy and Crop Science ,Protein Binding ,Cysteine - Abstract
We have previously shown that the expression of SmCP which encodes Solanum melongena cysteine proteinase is ethylene-inducible and is under circadian control. To understand the regulation of SmCP, a 1.34-kb SmCP 5'-flanking region and its deletion derivatives were analyzed for cis-elements using GUS and luc fusions and by in vitro binding assays. Analysis of transgenic tobacco transformed with SmCP promoter-GUS constructs confirmed that the promoter region -415/+54 containing Ethylene Responsive Element ERE(-355/-348) conferred threefold ethylene-induction of GUS expression, while -827/+54 which also contains ERE(-683/-676), produced fivefold induction. Using gel mobility shift assays, we demonstrated that each ERE binds nuclear proteins from both ethephon-treated and untreated 5-week-old seedlings, suggesting that different transcriptions factors bind each ERE under varying physiological conditions. Binding was also observed in extracts from senescent, but not young, fruits. The variation in binding at the EREs in fruits and seedlings imply that organ-specific factors may participate in binding. Analysis of transgenic tobacco expressing various SmCP promoter-luc constructs containing wild-type or mutant Evening Elements (EEs) confirmed that both conserved EEs at -795/-787 and -785/-777 are important in circadian control. We confirmed the binding of total nuclear proteins to EEs in gel mobility shift assays and in DNase I footprinting. Our results suggest that multiple proteins bind the EEs which are conserved in plants other than Arabidopsis and that functional EEs and EREs are present in the 5'-flanking region of a gene encoding cysteine proteinase.
- Published
- 2005
25. Identification and Characterization of Human VCY2-Interacting Protein: VCY2IP-1, a Microtubule-Associated Protein-Like Protein
- Author
-
Jenny Y.M. Tse, Po-Chor Tam, Vincent Chi Hang Lui, Kwok-Ming Yao, William S.B. Yeung, and Elaine Wong
- Subjects
Male ,Microtubule-associated protein ,Molecular Sequence Data ,Biology ,Homology (biology) ,Exon ,Two-Hybrid System Techniques ,Yeasts ,Testis ,Humans ,Immunoprecipitation ,Amino Acid Sequence ,RNA, Messenger ,Northern blot ,Gene ,Peptide sequence ,Gene Library ,Genetics ,chemistry.chemical_classification ,Base Sequence ,Sequence Homology, Amino Acid ,Proteins ,Cell Biology ,General Medicine ,Spermatozoa ,Amino acid ,Open reading frame ,Reproductive Medicine ,chemistry ,Chromosomes, Human, Pair 19 ,Microtubule-Associated Proteins - Abstract
VCY2 is a testis-specific protein that locates in a frequently deleted azoospermia factor c region on chromosome Yq. Although its genomic structure has been characterized, the function of VCY2 is still unknown. To gain insight regarding the likely function of VCY2, we investigated the proteins that interact with VCY2 using the yeast two-hybrid system. We identified a novel VCY2 interaction partner, named VCY2IP-1, that encodes an open reading frame of 1059 amino acids. The amino acid sequence of VCY2IP-1 shows 59.3% and 41.9% homology to two human microtubule-associated proteins (MAPs), MAP1B and MAP1A, respectively. VCY2IP-1 has an extensive homology to the N-terminus and C-terminus regions of MAP1B and MAP1A, placing it within a large family of MAPs. We mapped VCY2IP-1 to chromosome 19p13.11. The VCY2IP-1 gene spans 15 kilobases (kb) and consists of seven exons. Northern blot analysis identified a single, intense band of approximately 3.2-kb VCY2IP-1 transcript, predominantly expressed in human testis. In situ hybridization of human testicular sections showed the localization of VCY2IP-1 transcripts in germ cells, and reverse transcription-polymerase chain reaction analysis demonstrated the presence of VCY2 and VCY2IP-1 transcripts in human ejaculated spermatozoa. Our expression data support the involvement of VCY2 and VCY2IP-1 in spermatogenesis. Based on the high homology of VCY2IP-1 with MAPs, we propose the involvement of VCY2 in the cytoskeletal network via interaction with VCY2IP-1.
- Published
- 2004
26. [Untitled]
- Author
-
Mee-Len Chye, Fang-Xiu Xu, Hong-Ye Li, Kwok-Ming Yao, and Zeng-Fu Xu
- Subjects
Regulation of gene expression ,Mutant ,Plant Science ,General Medicine ,Protein degradation ,Biology ,Molecular biology ,Transcription (biology) ,Genetics ,Circadian rhythm ,Northern blot ,Nuclear protein ,Agronomy and Crop Science ,Gene - Abstract
We have previously shown that SmCP, the gene encoding Solanum melongena cysteine proteinase, is expressed during developmental events associated with programmed cell death (PCD) suggesting its involvement in protein degradation during these events (Xu and Chye, Plant Journal 17 (1999) 321–327). Here, we investigated the regulation of SmCP expression and showed that it is ethylene-inducible and is under circadian control. This circadian rhythm is entrained by light/dark (LD) cycling with peak expression in the late light period, as opposed to that in early light for rbcS, suggesting that protein degradation and photosynthesis are temporally separated by circadian control. Northern blot analysis shows that the pattern of ethylene induction of SmCP is consistent with our previous observation of its significantly increased expression at leaf senescence and fruit ripening when endogenous ethylene is abundant. To further understand SmCP regulation, we have cloned the SmCP promoter and identified a G-box (CACGTG) at −85/−80 by DNase I footprinting analysis of the −221/+17 region. Its specific interaction with nuclear proteins in S. melongena leaves and fruits was confirmed by competitive electrophoretic mobility shift assays using oligonucleotides containing the G-box and mutant derivatives. G-box binding activity was stronger in senescent than young fruits. In circadian-regulated leaves, stronger binding activity coincided with peak circadian expression of SmCP. This correlation between binding activity and expression suggests that G-box binding factors enhance SmCP transcription and that the G-box likely plays a role in circadian regulation of genes affected by LD cycling.
- Published
- 2003
27. Reduced expression of AMPK-β1 during tumor progression enhances the oncogenic capacity of advanced ovarian cancer
- Author
-
Vincent W.S. Liu, Cuilan Li, Kwok-Ming Yao, Pui Man Chiu, David W. Chan, and Hextan Y.S. Ngan
- Subjects
Cancer Research ,medicine.medical_specialty ,Blotting, Western ,Fluorescent Antibody Technique ,AMPK-β1 ,AMP-Activated Protein Kinases ,Biology ,Transfection ,medicine.disease_cause ,AMP-activated protein kinase ,Ovarian cancer ,Cell Line, Tumor ,Internal medicine ,medicine ,Humans ,AMPK activation ,Protein kinase B ,Ovarian Neoplasms ,Microscopy, Confocal ,Cell growth ,Research ,AKT ,AMPK ,Cell migration ,Flow Cytometry ,medicine.disease ,Immunohistochemistry ,Enzyme Activation ,ERK ,Endocrinology ,Oncology ,Tissue Array Analysis ,Tumor progression ,Disease Progression ,biology.protein ,Cancer research ,Molecular Medicine ,Female ,JNK ,Carcinogenesis - Abstract
AMP-activated protein kinase (AMPK) is a key energy sensor that is involved in regulating cell metabolism. Our previous study revealed that the subunits of the heterotimeric AMPK enzyme are diversely expressed during ovarian cancer progression. However, the impact of the variable expression of these AMPK subunits in ovarian cancer oncogenesis remains obscure. Here, we provide evidence to show that reduced expression of the AMPK-beta1 subunit during tumor progression is associated with the increased oncogenic capacity of advanced ovarian cancer cells. Immunohistochemical analysis revealed that AMPK-beta1 levels were reduced in advanced-stage (P = 0.008), high-grade (P = 0.013) and metastatic ovarian cancers (P = 0.008). Intriguingly, down-regulation of AMPK-beta1 was progressively reduced from tumor stages 1 to 3 of ovarian cancer. Functionally, enforced expression of AMPK-beta1 inhibited ovarian-cancer-cell proliferation, anchorage-independent cell growth, cell migration and invasion. Conversely, depletion of AMPK-beta1 by siRNA enhanced the oncogenic capacities of ovarian cancer cells, suggesting that the loss of AMPK-beta1 favors the aggressiveness of ovarian cancer. Mechanistically, enforced expression of AMPK-beta1 increased AMPK activity, which, in turn, induced cell-cycle arrest via inhibition of AKT/ERK signaling activity as well as impaired cell migration/invasion through the suppression of JNK signaling in ovarian cancer cells. Taken together, these findings suggest that the reduced expression of AMPK-beta1 confers lower AMPK activity, which enhances the oncogenic capacity of advanced-stage ovarian cancer., published_or_final_version
- Published
- 2014
28. Inhibition of malignant trophoblastic cell proliferation in vitro and in vivo by melatonin
- Author
-
Kwok-Ming Yao, L. Mei, Jinjin Xu, Shiu-Fun Pang, Stephen Y. W. Shiu, S.C. Xi, and Joseph T.Y. Wong
- Subjects
Cell Survival ,Cyclin A ,Receptors, Melatonin ,Mice, Nude ,Receptors, Cytoplasmic and Nuclear ,Receptors, Cell Surface ,Melatonin receptor ,General Biochemistry, Genetics and Molecular Biology ,Melatonin ,Mice ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Choriocarcinoma ,General Pharmacology, Toxicology and Pharmaceutics ,Receptor ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Cell growth ,General Medicine ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Apoptosis ,Uterine Neoplasms ,embryonic structures ,biology.protein ,Female ,Cell Division ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Melatonin inhibited thymidine incorporation into human choriocarcinoma JEG-3 cells at physiological and pharmacological concentrations in the present study. Gene expression of MT2 receptor, but not that of mt1 receptor, was detected in JEG-3 cells by reverse transcription-polymerase chain reaction (RT-PCR). The gene expression profile of the two human melatonin receptor subtypes in JEG-3 cells was identical to that previously reported for JAr cells, whose proliferation had also been shown to be similarly inhibited by physiological and pharmacological concentrations of melatonin. In contrast, melatonin had no effect on thymidine incorporation into 3A-Sub-E cells (a transformed trophoblast cell line), in which gene expression of both receptor subtypes could not be detected. The data suggest that in human placental trophoblasts, a correlation may exist between MT2 receptor gene expression and the direct anti-proliferative action of melatonin. Although melatonin has been reported to induce G1/S delay in cell cycle progression of JAr cells, no significant changes in the percentages of JEG-3 cells in different cell cycle phases upon melatonin treatment was recorded by flow cytometric analysis. This indicates that G1/S transition delay is probably not an important cellular mechanism in the direct anti-proliferative action of melatonin on human JEG-3 cells in vitro. Furthermore, melatonin inhibited the growth of both JAr and JEG-3 xenograft tumors in athymic nude mice, and prolonged the survival of those animals that developed choriocarcinoma. While the number of apoptotic tumor cells was not increased by melatonin, the pineal hormone induced significant decreases in the numbers of JAr and JEG-3 cells expressing proliferating cell nuclear antigen (PCNA) and cyclin A in the tumors. Taking into account both the in vitro and in vivo findings, it is likely that the inhibitory effect of melatonin on choriocarcinoma JAr and JEG-3 cell proliferation in vivo is largely a direct action of the hormone on the tumor cells.
- Published
- 2000
29. Diversity, function, and transcriptional regulation of gut innate lymphocytes
- Author
-
Adaline Wan Ling Ngan, David W. Chan, Wai Ying Leung, Davis C T Kwok, Andy King Yin Lam, Kwok-Ming Yao, Vincent Liu, and Man-Hong Leung
- Subjects
Gene isoform ,Cancer Research ,business.industry ,medicine.disease_cause ,Inhibitory postsynaptic potential ,Bioinformatics ,Cell biology ,Exon ,Mapk signaling ,Oncology ,Cancer cell ,medicine ,FOXM1 ,Carcinogenesis ,business ,Transcription factor - Abstract
The forkhead box (FOX) M1 transcription factor is required to maintain the proliferation of cancer cells. Two transcriptionally active isoforms of FOXM1, FOXM1b and FOXM1c, have been identified, but their functional differences remain unclear. FOXM1c is distinguished from FOXM1b by an extra exon (exon Va) that contains an ERK1/2 target sequence. Based on a literature search and quantitative PCR analysis, we concluded that FOXM1b is the predominant isoform that is overexpressed in cancers. The further characterization of FOXM1b and FOXM1c revealed two interesting differences. First, FOXM1b exhibited a higher transforming ability than FOXM1c in a soft agar assay. Second, the transactivating activity of FOXM1c, but not that of FOXM1b, was sensitive to activation by RAF/MEK/MAPK signaling. Importantly, the MEK1 activation of FOXM1c was associated with proteolytic processing to generate short forms that might represent constitutively active forms missing the N-terminal inhibitory domain; in contrast, the proteolytic processing of FOXM1b did not require MEK1 activation. Our findings suggest that FOXM1b is functionally more active. These results provide novel insights into the regulation of FOXM1 activity and its role in tumorigenesis.
- Published
- 2013
30. Secreted PDZ domain-containing protein 2 (sPDZD2) exerts insulinotropic effects on INS-1E cells via a protein kinase A-dependent mechanism
- Author
-
Kwok-Ming Yao, CY Chan, and SW Tsang
- Subjects
Biochemistry ,Chemistry ,Mechanism (biology) ,Kinase ,PDZ domain ,A protein - Published
- 2012
31. Purification and characterization of ornithine decarbozylase fromtetrahymena thermophila
- Author
-
Kwok-ming. Yao
- Subjects
chemistry.chemical_compound ,Biochemistry ,Chemistry ,Ornithine - Published
- 2012
32. Targeted inactivation of kinesin-1 in pancreatic β-cells in vivo leads to insulin secretory deficiency
- Author
-
Zai Wang, Ju Cui, Neal G. Copeland, Nancy A. Jenkins, Kwok-Ming Yao, Po Sing Leung, Xin Mei Zhang, Raozhou Lin, Jian-Dong Huang, and Qianni Cheng
- Subjects
Male ,medicine.medical_specialty ,Genotype ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Blotting, Western ,Molecular Sequence Data ,Kinesins ,Enzyme-Linked Immunosorbent Assay ,Biology ,Islets of Langerhans ,Mice ,Insulin resistance ,Kinesin - antagonists and inhibitors - chemistry - deficiency - drug effects - genetics ,Internal medicine ,Insulin-Secreting Cells ,Conditional gene knockout ,Insulin Secretion ,Internal Medicine ,medicine ,Genetics ,Glucose homeostasis ,Animals ,Humans ,Insulin ,Amino Acid Sequence ,DNA Primers ,Mice, Knockout ,geography ,geography.geographical_feature_category ,Base Sequence ,Oligonucleotides, Antisense ,medicine.disease ,Islet ,Molecular biology ,Peptide Fragments ,Insulin oscillation ,Endocrinology ,medicine.anatomical_structure ,Insulin-Secreting Cells - drug effects - physiology ,Knockout mouse ,Pancreas ,Insulin - deficiency - secretion - Abstract
OBJECTIVE: Suppression of Kinesin-1 by antisense oligonucleotides, or overexpression of dominant-negative acting kinesin heavy chain, has been reported to affect the sustained phase of glucose-stimulated insulin secretion in β-cells in vitro. In this study, we examined the in vivo physiological role of Kinesin-1 in β-cell development and function. RESEARCH DESIGN AND METHODS: A Cre-LoxP strategy was used to generate conditional knockout mice in which the Kif5b gene is specifically inactivated in pancreatic β-cells. Physiological and histological analyses were carried out in Kif5b knockout mice as well as littermate controls. RESULTS: Mice with β-cell specific deletion of Kif5b (Kif5b fl/-: RIP2-Cre) displayed significantly retarded growth as well as slight hyperglycemia in both nonfasting and 16-h fasting conditions compared with control littermates. In addition, Kif5b fl/-: RIP2-Cre mice displayed significant glucose intolerance, which was not due to insulin resistance but was related to an insulin secretory defect in response to glucose challenge. These defects of β-cell function in mutant mice were not coupled with observable changes in islet morphology, islet cell composition, or β-cell size. However, compared with controls, pancreas of Kif5b fl/-: RIP2-Cre mice exhibited both reduced islet size and increased islet number, concomitant with an increased insulin vesicle density in β-cells. CONCLUSIONS: In addition to being essential for maintaining glucose homeostasis and regulating β-cell function, Kif5b may be involved in β-cell development by regulating β-cell proliferation and insulin vesicle synthesis. © 2011 by the American Diabetes Association., published_or_final_version
- Published
- 2010
33. Signal transduction of receptor-mediated antiproliferative action of melatonin on human prostate epithelial cells involves dual activation of Gα(s) and Gα(q) proteins
- Author
-
Stephen Y W, Shiu, Bo, Pang, Chun W, Tam, and Kwok-Ming, Yao
- Subjects
Human papillomavirus 18 ,Tetrahydronaphthalenes ,Reverse Transcriptase Polymerase Chain Reaction ,Radioimmunoassay ,Receptors, Melatonin ,Antioxidants ,Tryptamines ,Cell Line, Tumor ,GTP-Binding Protein alpha Subunits, Gs ,GTP-Binding Protein alpha Subunits, Gq-G11 ,Humans ,RNA Interference ,GTP-Binding Protein alpha Subunit, Gi2 ,Cyclin-Dependent Kinase Inhibitor p27 ,Cell Proliferation ,Melatonin ,Signal Transduction - Abstract
Melatonin has been shown to inhibit the proliferation of malignant and transformed human prostate epithelial cells by transcriptional up-regulation of p27(Kip1) expression via MTNR1A receptor-mediated activation of protein kinase A (PKA) and protein kinase C (PKC) in parallel. Given that melatonin MTNR1A receptor is a G protein-coupled receptor, this study was conducted to identify the specific G proteins that mediate the antiproliferative action of melatonin on human prostate epithelial cells. In 22Rv1 and RWPE-1 cells, knockdown of either Gα(s) or Gα(q) , but not Gα(i2) expression by RNA interference, abrogated the effects of melatonin on p27(Kip1) and cell proliferation. Conversely, cellular overexpression of activated mutants of Gα(s) and Gα(q) in 22Rv1 and RWPE-1 cells mimicked the effects of melatonin on prostate epithelial cell antiproliferation by increasing p27(Kip1) expression through downstream activation of PKA and PKC in parallel. Moreover, melatonin or 2-iodomelatonin induced elevation of adenosine-3',5'-cyclic monophosphate (cAMP) in 22Rv1 and RWPE-1 cells. The effects of 2-iodomelatonin on cAMP were blocked by the nonselective MTNR1A/MTNR1B receptor antagonist luzindole but were not affected by the selective MTNR1B receptor antagonist 4-phenyl-2-propionamidotetraline (4-P-PDOT). Furthermore, knockdown of Gα(s) mitigated the stimulatory effects of 2-iodomelatonin on cAMP. Collectively, the data demonstrated, for the first time, functional coupling of MTNR1A receptor to Gα(s) in cancerous or transformed human cells expressing endogenous melatonin receptors. Our results also showed that dual activation of Gα(s) and Gα(q) proteins is involved in the signal transduction of MTNR1A receptor-mediated antiproliferative action of melatonin on human prostate epithelial cells.
- Published
- 2010
34. Raf/MEK/MAPK signaling stimulates the nuclear translocation and transactivating activity of FOXM1
- Author
-
Richard Y M, Ma, Tommy H K, Tong, Wai Ying, Leung, and Kwok-Ming, Yao
- Subjects
Cell Nucleus ,Mitogen-Activated Protein Kinase Kinases ,Transcriptional Activation ,Mice ,Genes, Reporter ,MAP Kinase Signaling System ,Active Transport, Cell Nucleus ,Animals ,Humans ,Forkhead Transcription Factors ,raf Kinases ,Fibroblasts ,Cell Line - Abstract
The proliferation-associated transcription factor FOXM1 is essential for cell cycle progression into mitosis. Using synchronized human fibroblasts we detected, by immunostaining, that FOXM1 is localized predominantly in the cytoplasm in cells at late-G1 and S phases. Nuclear translocation occurs just before progression into the G2/M phase of the cell cycle and requires activity of the Raf/MEK/MAPK signaling pathway. Using pharmacological modulators, we demonstrated that activity of the Raf/MEK/MAPK pathway is both necessary and sufficient for the nuclear translocation of FOXM1. Consistent with FoxM1c being the major isoform expressed in proliferating fibroblasts, constitutively active MEK1 enhances the transactivating effect of FOXM1c, but not FOXM1b, on the cyclin B1 promoter in transient reporter assays. Here, we describe in detail the methods involved in generating these findings, which support the notion that FOXM1 is an effector of Raf/MEK/MAPK signaling in G2/M regulation.
- Published
- 2010
35. Raf/MEK/MAPK Signaling Stimulates the Nuclear Translocation and Transactivating Activity of FOXM1
- Author
-
Wai Ying Leung, Kwok-Ming Yao, Richard Y. M. Ma, and Tommy H. K. Tong
- Subjects
MAPK/ERK pathway ,Chemistry ,Cytoplasm ,Effector ,FOXM1 ,Cell cycle ,Cyclin B1 ,Transcription factor ,Mitosis ,Cell biology - Abstract
The proliferation-associated transcription factor FOXM1 is essential for cell cycle progression into mitosis. Using synchronized human fibroblasts we detected, by immunostaining, that FOXM1 is localized predominantly in the cytoplasm in cells at late-G1 and S phases. Nuclear translocation occurs just before progression into the G2/M phase of the cell cycle and requires activity of the Raf/MEK/MAPK signaling pathway. Using pharmacological modulators, we demonstrated that activity of the Raf/MEK/MAPK pathway is both necessary and sufficient for the nuclear translocation of FOXM1. Consistent with FoxM1c being the major isoform expressed in proliferating fibroblasts, constitutively active MEK1 enhances the transactivating effect of FOXM1c, but not FOXM1b, on the cyclin B1 promoter in transient reporter assays. Here, we describe in detail the methods involved in generating these findings, which support the notion that FOXM1 is an effector of Raf/MEK/MAPK signaling in G2/M regulation.
- Published
- 2010
36. Increased basal insulin secretion in Pdzd2-deficient mice
- Author
-
K. S.E. Cheah, D. Shao, Kenji Okuse, Kwok-Ming Yao, Siu Wai Tsang, and Po Sing Leung
- Subjects
Blood Glucose ,medicine.medical_specialty ,medicine.medical_treatment ,Nerve Tissue Proteins ,Biology ,Biochemistry ,Mice ,Endocrinology ,Diabetes mellitus ,Internal medicine ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Animals ,Insulin ,Secretion ,Gene Silencing ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,Glucose tolerance test ,geography ,geography.geographical_feature_category ,medicine.diagnostic_test ,Pancreatic islets ,Body Weight ,Glucose Tolerance Test ,medicine.disease ,Islet ,Insulin oscillation ,medicine.anatomical_structure ,Phenotype ,Pancreas ,Cell Adhesion Molecules - Abstract
Expression of the multi-PDZ protein Pdzd2 (PDZ domain-containing protein 2) is enriched in pancreatic islet beta cells, but not in exocrine or alpha cells, suggesting a role for Pdzd2 in the regulation of pancreatic beta-cell function. To explore the in vivo function of Pdzd2, Pdzd2-deficient mice were generated. Homozygous Pdzd2 mutant mice were viable and their gross morphology appeared normal. Interestingly, Pdzd2-deficient mice showed enhanced glucose tolerance in intraperitoneal glucose tolerance tests and their plasma insulin levels indicated increased basal insulin secretion after fasting. Moreover, insulin release from mutant pancreatic islets was found to be twofold higher than from normal islets. To verify the functional defect in vitro, Pdzd2 was depleted in INS-1E cells using two siRNA duplexes. Pdzd2-depleted INS-1E cells also displayed increased insulin secretion at low concentrations of glucose. Our results provide the first evidence that Pdzd2 is required for normal regulation of basal insulin secretion.
- Published
- 2009
37. Overexpression of FOXG1 contributes to TGF-β resistance through inhibition of p21 WAF1/CIP1 expression in ovarian cancer
- Author
-
W Y W Lee, Annie N.Y. Cheung, R M Y To, Kwok-Ming Yao, David W. Chan, PM Chiu, Vws Liu, and Hextan Ys Ngan
- Subjects
Adult ,Cyclin-Dependent Kinase Inhibitor p21 ,TGF-β ,Cancer Research ,Tumor suppressor gene ,Active Transport, Cell Nucleus ,Cyclin-Dependent Kinase Inhibitor p21 - antagonists and inhibitors - genetics ,Nerve Tissue Proteins ,Small hairpin RNA ,Transforming Growth Factor beta ,Ovarian cancer ,Cell Line, Tumor ,medicine ,Humans ,Promoter Regions, Genetic ,Molecular Diagnostics ,Aged ,Cell Proliferation ,Ovarian Neoplasms ,Gene knockdown ,biology ,Ovarian Neoplasms - drug therapy - pathology ,Cell growth ,Cancer ,Forkhead Transcription Factors - analysis - genetics - physiology ,Forkhead Transcription Factors ,Transforming growth factor beta ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Transforming Growth Factor beta - pharmacology ,Nerve Tissue Proteins - analysis - genetics - physiology ,Oncology ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Female ,FOXG1 ,P21WAF1/CIP1 ,Transforming growth factor ,Signal Transduction - Abstract
Background:Loss of growth inhibitory response to transforming growth factor-Β (TGF-Β) is a common feature of epithelial cancers. Recent studies have reported that genetic lesions and overexpression of oncoproteins in TGF-Β/Smads signalling cascade contribute to the TGF-Β resistance. Here, we showed that the overexpressed FOXG1 was involved in attenuating the anti-proliferative control of TGF-Β/Smads signalling in ovarian cancer.Methods:FOXG1 and p21 WAF1/CIP1 expressions were evaluated by real-time quantitative reverse-transcription polymerase chain reaction (RT-PCR), western blot and immunohistochemical analyses. The effect of FOXG1 on p21 WAF1/CIP1 transcriptional activity was examined by luciferase reporter assays. Cell lines stably expressing or short hairpin RNA interference-mediated knockdown FOXG1 were established for studying the gain-or-loss functional effects of FOXG1. XTT cell proliferation assay was used to measure cell growth of ovarian cancer cells.Results:Quantitative RT-PCR and western blot analyses showed that FOXG1 was upregulated and inversely associated with the expression levels of p21 WAF1/CIP1 in ovarian cancer. The overexpression of FOXG1 was significantly correlated with high-grade ovarian cancer (P0.025). Immunohistochemical analysis on ovarian cancer tissue array was further evidenced that FOXG1 was highly expressed and significantly correlated with high-grade ovarian cancer (P0.048). Functionally, enforced expression of FOXG1 selectively blocked the TGF-Β-induced p21 WAF1/CIP1 expressions and increased cell proliferation in ovarian cancer cells. Conversely, FOXG1 knockdown resulted in a 20-26% decrease in cell proliferation together with 16-33% increase in p21 WAF1/CIP1 expression. Notably, FOXG1 was able to inhibit the p21 WAF1/CIP1 promoter activity in a p53-independent manner by transient reporter assays.ConclusionOur results suggest that FOXG1 acts as an oncoprotein inhibiting TGF-Β-mediated anti-proliferative responses in ovarian cancer cells through suppressing p21 WAF1/CIP1 transcription. © 2009 Cancer Research UK All rights reserved., published_or_final_version
- Published
- 2009
38. Organizational analysis of elav gene and functional analysis of ELAV protein of Drosophila melanogaster and Drosophila virilis
- Author
-
Kalpana White and Kwok-Ming Yao
- Subjects
Sequence analysis ,Molecular Sequence Data ,Restriction Mapping ,Polymerase Chain Reaction ,Homology (biology) ,Conserved sequence ,Open Reading Frames ,Sequence Homology, Nucleic Acid ,Melanogaster ,Animals ,Amino Acid Sequence ,Molecular Biology ,Gene ,Genetics ,Base Sequence ,biology ,DNA ,Cell Biology ,biology.organism_classification ,Drosophila virilis ,Open reading frame ,Drosophila melanogaster ,ELAV Proteins ,Ribonucleoproteins ,Drosophila ,Genes, Lethal ,Oligonucleotide Probes ,Plasmids ,Research Article - Abstract
Drosophila virilis genomic DNA corresponding to the D. melanogaster embryonic lethal abnormal visual system (elav) locus was cloned. DNA sequence analysis of a 3.8-kb genomic piece allowed identification of (i) an open reading frame (ORF) with striking homology to the previously identified D. melanogaster ORF and (ii) conserved sequence elements of possible regulatory relevance within and flanking the second intron. Conceptual translation of the D. virilis ORF predicts a 519-amino-acid-long ribonucleoprotein consensus sequence-type protein. Similar to D. melanogaster ELAV protein, it contains three tandem RNA-binding domains and an alanine/glutamine-rich amino-terminal region. The sequence throughout the RNA-binding domains, comprising the carboxy-terminal 346 amino acids, shows an extraordinary 100% identity at the amino acid level, indicating a strong structural constraint for this functional domain. The amino-terminal region is 36 amino acids longer in D. virilis, and the conservation is 66%. In in vivo functional tests, the D. virilis ORF was indistinguishable from the D. melanogaster ORF. Furthermore, a D. melanogaster ORF encoding an ELAV protein with a 40-amino-acid deletion within the alanine/glutamine-rich region was also able to supply elav function in vivo. Thus, the divergence of the amino-terminal region of the ELAV protein reflects lowered functional constraint rather than species-specific functional specification.
- Published
- 1991
39. Melatonin as a negative mitogenic hormonal regulator of human prostate epithelial cell growth: potential mechanisms and clinical significance
- Author
-
Bo Pang, Stephen Y. W. Shiu, Kwok-Ming Yao, Chun W. Tam, Vincent W.S. Liu, and Kwok W. Chan
- Subjects
Male ,medicine.medical_specialty ,Tetrahydronaphthalenes ,Immunoblotting ,Receptors, Melatonin ,Biology ,Prostate cancer ,Endocrinology ,Prostate ,Cell surface receptor ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,RNA, Small Interfering ,Protein kinase A ,Receptor ,Growth Substances ,Protein kinase C ,Cells, Cultured ,Cell Line, Transformed ,Cell Proliferation ,Melatonin ,Analysis of Variance ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Prostatic Neoplasms ,Epithelial Cells ,medicine.disease ,Immunohistochemistry ,Tryptamines ,medicine.anatomical_structure ,Gene Expression Regulation ,Receptors, Androgen ,Cancer research ,Luzindole ,Protein Kinases ,Cyclin-Dependent Kinase Inhibitor p27 ,Signal Transduction - Abstract
Circannual variation in the human serum levels of prostate-specific antigen, a growth marker of the prostate gland, has been reported recently. The present study was conducted to investigate the role of the photoperiodic hormone melatonin (MLT) and its membrane receptors in the modulation of human prostate growth. Expression of MT(1) and MT(2) receptors was detected in benign human prostatic epithelial tissues and RWPE-1 cells. MLT and 2-iodomelatonin inhibited RWPE-1 cell proliferation and up-regulated p27(Kip1) gene and protein expression in the cells. The effects of MLT were blocked by the nonselective MT(1)/MT(2) receptor antagonist luzindole, but were not affected by the selective MT(2) receptor antagonist 4-phenyl-2-propionamidotetraline. Of note, the antiproliferative action of MLT on benign prostate epithelial RWPE-1 cells was effected via increased p27(Kip1) gene transcription through MT(1) receptor-mediated activation of protein kinase A (PKA) and protein kinase C (PKC) in parallel, a signaling process which has previously been demonstrated in 22Rv1 prostate cancer cells. Taken together, the demonstration of the MT(1)/PKA+PKC/p27(Kip1) antiproliferative pathway in benign and malignant prostate epithelial cell lines indicated the potential importance of this MLT receptor-mediated signaling mechanism in growth regulation of the human prostate gland in health and disease. Collectively, our data support the hypothesis that MLT may function as a negative mitogenic hormonal regulator of human prostate epithelial cell growth.
- Published
- 2008
40. Loss of MKP3 mediated by oxidative stress enhances tumorigenicity and chemoresistance of ovarian cancer cells
- Author
-
Hextan Y.S. Ngan, Toru Furukawa, George S.W. Tsao, David W. Chan, Karen K. L. Chan, Kwok-Ming Yao, and Vincent W.S. Liu
- Subjects
MAPK/ERK pathway ,Cancer Research ,medicine.medical_specialty ,Blotting, Western ,Apoptosis ,Biology ,Small hairpin RNA ,Mice ,Dual Specificity Phosphatase 6 ,Internal medicine ,medicine ,Cell Adhesion ,Tumor Cells, Cultured ,Animals ,Humans ,RNA, Messenger ,Phosphorylation ,Protein kinase A ,Promoter Regions, Genetic ,Cell Proliferation ,Mitogen-Activated Protein Kinase 1 ,Ovarian Neoplasms ,Mitogen-Activated Protein Kinase 3 ,Kinase ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Ubiquitin ,Cancer ,General Medicine ,Hydrogen Peroxide ,medicine.disease ,Oxidative Stress ,Endocrinology ,Drug Resistance, Neoplasm ,Cancer research ,Disease Progression ,Female ,Signal transduction ,Cisplatin ,Ovarian cancer ,Signal Transduction - Abstract
The RAS-RAF-MEK-extracellular signal-regulated kinase (ERK) pathway plays a pivotal role in various cellular responses, including cellular growth, differentiation, survival and motility. Constitutive activation of the ERK pathway has been linked to the development and progression of human cancers. Here, we reported that mitogen-activated protein kinase phosphatase (MKP)-3, a negative regulator of ERK1/2, lost its expression particularly in the protein level, was significantly correlated with high ERK1/2 activity in primary human ovarian cancer cells using quantitative reverse transcription-polymerase chain reaction and western blot analyses. Intriguingly, the loss of MKP3 protein was associated with ubiquitination/proteosome degradation mediated by high intracellular reactive oxygen species (ROS) accumulation such as hydrogen peroxide in ovarian cancer cells. Functionally, short hairpin RNA knock down of endogenous MKP3 resulted in increased ERK1/2 activity, cell proliferation rate, anchorage-independent growth ability and resistance to cisplatin in ovarian cancer cells. Conversely, enforced expression of MKP3 in MKP3-deficient ovarian cancer cells significantly reduced ERK1/2 activity and inhibited cell proliferation, anchorage-independent growth ability and tumor development in nude mice. Furthermore, the enforced expression of MKP3 succeeded to sensitize ovarian cancer cells to cisplatin-induced apoptosis in vitro and in vivo. These results suggest a molecular mechanism by which the accumulation of ROS during ovarian cancer progression may cause the degradation of MKP3, which in turn leads to aberrant ERK1/2 activation and contributes to tumorigenicity and chemoresistance of human ovarian cancer cells.
- Published
- 2008
41. The autocrine human secreted PDZ domain-containing protein 2 (sPDZD2) induces senescence or quiescence of prostate, breast and liver cancer cells via transcriptional activation of p53
- Author
-
Chun Wai Tam, Kwok-Ming Yao, Vincent W.S. Liu, Wai Ying Leung, and Stephen Y. W. Shiu
- Subjects
Senescence ,Male ,Transcriptional Activation ,Cancer Research ,PDZ domain ,Apoptosis ,Breast Neoplasms ,Biology ,Prostate cancer ,DU145 ,Cell Line, Tumor ,medicine ,Transcriptional regulation ,Humans ,Autocrine signalling ,Cellular Senescence ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,DNA Primers ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Liver Neoplasms ,Prostatic Neoplasms ,medicine.disease ,Neoplasm Proteins ,Oncology ,Cell culture ,Cancer research ,RNA Interference ,Tumor Suppressor Protein p53 ,Cell Adhesion Molecules - Abstract
Tumor suppressive actions of the autocrine human secreted PDZ domain-containing protein 2 (sPDZD2) have been reported, but the mechanisms remain enigmatic. Here, we showed that sPDZD2 induced senescence of prostate cancer DU145 cells, quiescence of breast cancer MCF-7 and liver cancer Hep-G2 cells, via transcriptional activation of mutant or wild-type p53. Furthermore, sPDZD2 sensitized mutant p53-positive DU145 cells and wild-type p53-positive MCF-7 cells to apoptosis induction through genotoxic stress imposed by sub-lethal concentration of hydrogen peroxide. Together, our findings suggest a potential autocrine pathway of p53 activation by transcriptional regulation, and a new approach to reactivate p53 for cancer therapy.
- Published
- 2008
42. FoxM1c counteracts oxidative stress-induced senescence and stimulates Bmi-1 expression
- Author
-
Wai Ying Leung, Goberdhan P. Dimri, Samuel K.M. Li, Alice M.S. Cheung, Eric Lam, Kwok-Ming Yao, and David K. Smith
- Subjects
Senescence ,Forkhead Transcription Factors - metabolism - physiology ,Repressor Proteins - metabolism - physiology ,Biology ,medicine.disease_cause ,Biochemistry ,Models, Biological ,Proto-Oncogene Proteins c-myc ,Mice ,Molecular Basis of Cell and Developmental Biology ,RNA interference ,Proto-Oncogene Proteins ,medicine ,Proto-Oncogene Proteins - metabolism - physiology ,Animals ,Molecular Biology ,Transcription factor ,Cellular Senescence ,Polycomb Repressive Complex 1 ,Binding Sites ,Cell growth ,Nuclear Proteins - metabolism - physiology ,Forkhead Box Protein M1 ,Nuclear Proteins ,Forkhead Transcription Factors ,Cell Biology ,Cell cycle ,Molecular biology ,Anti-Bacterial Agents ,Repressor Proteins ,Gene Expression Regulation, Neoplastic ,Oxidative Stress ,Cell culture ,Doxycycline ,FOXM1 ,NIH 3T3 Cells ,RNA Interference ,Tumor Suppressor Protein p53 ,Carcinogenesis - Abstract
The Forkhead box transcription factor FoxM1 is expressed in proliferating cells. When it was depleted in mice and cell lines, cell cycle defects and chromosomal instability resulted. Premature senescence was observed in embryonic fibroblasts derived from FoxM1 knock-out mice, but the underlying cause has remained unclear. To investigate whether FoxM1 can protect cells against stress-induced premature senescence, we established NIH3T3 lines with doxycycline-inducible overexpression of FoxM1c. Treatment of these lines with sublethal doses (20 and 100 μM) of H 2O 2 induced senescence with senescence-associated β-galactosidase expression and elevated levels of p53 and p21. Induction of FoxM1c expression markedly suppressed senescence and expression of p53 and p21. Consistent with down-regulation of the p19 Arf-p53 pathway, p19 Arf levels decreased while expression of the Polycomb group protein Bmi-1 was induced. That Bmi-1 is a downstream target of FoxM1c was further supported by the dose-dependent induction of Bmi-1 by FoxM1c at both the protein and mRNA levels, and FoxM1 and Bmi-1 reached maximal levels in cells at the G 2/M phase. Depletion of FoxM1 by RNA interference decreased Bmi-1 expression. Using Bmi-1 promoter reporters with wild-type and mutated c-Myc binding sites and short hairpin RNAs targeting c-Myc, we further demonstrated that FoxM1c activated Bmi-1 expression via c-Myc, which was recently reported to be regulated by FoxM1c. Our results reveal a functional link between FoxM1c, c-Myc, and Bmi-1, which are major regulators of tumorigenesis. This link has important implications for the regulation of cell proliferation and senescence by FoxM1 and Bmi-1. © 2008 by The American Society for Biochemistry and Molecular Biology, Inc., link_to_subscribed_fulltext
- Published
- 2008
43. A 3D collagen microsphere culture system for GDNF-secreting HEK293 cells with enhanced protein productivity
- Author
-
Ming-Xi Wang, Hoi-Ling Wong, Barbara P. Chan, Pik-To Cheung, and Kwok-Ming Yao
- Subjects
Biophysics ,Cell Culture Techniques ,Bioengineering ,Kidney ,PC12 Cells ,Collagen Type I ,Cell Line ,Biomaterials ,Mice ,Neurotrophic factors ,Glial cell line-derived neurotrophic factor ,Animals ,Humans ,Glial Cell Line-Derived Neurotrophic Factor ,biology ,Cell growth ,HEK 293 cells ,Cell cycle ,Embryonic stem cell ,Microspheres ,Recombinant Proteins ,Cell biology ,Rats ,Mechanics of Materials ,Apoptosis ,Cell culture ,Ceramics and Composites ,biology.protein ,Biomedical engineering - Abstract
Mammalian cell culture technology has been used for decades in mass production of therapeutic proteins. However, unrestricted cell proliferation usually results in low-protein productivity. Controlled proliferation technologies such as metabolism intervention and genetic manipulation are therefore applied to enhance the productivity. Nevertheless, these strategies induced growth arrest with reduced viability and increased apoptosis. In this study, we report a new controlled proliferation technology by encapsulating human embryonic kidney (HEK) 293 cells over-expressing glial-derived neurotrophic factor (GDNF) in 3D collagen microspheres for extended culture. We investigated the viability, proliferation, cell cycle and GDNF productivity of HEK293 cells in microspheres as compared to monolayer culture. This system provides a physiologically relevant tissue-like environment for cells to grow and exerts proliferation control throughout the culture period without compromising the viability. A significant increase in the production rate of GDNF was found in the 3D microsphere system comparing with the monolayer culture. GDNF productivity was also significantly affected by the initial cell number and the serum concentration. The secreted GDNF was still bioactive as it induced neurite extension in PC12 cells. In summary, the 3D collagen microsphere system presents a cost-effective controlled growth technology for protein production in pharmaceutical manufacturing.
- Published
- 2007
44. The anti‐proliferative effects of sPDZD2 on prostate and non‐prostate cancer cells
- Author
-
Chun-Wai Tam, Kwok-Ming Yao, Stephen Y. W. Shiu, and Amy Shan Cheng
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Anti proliferative ,medicine.disease ,Biochemistry ,Prostate cancer ,medicine.anatomical_structure ,Prostate ,Internal medicine ,Genetics ,medicine ,business ,Molecular Biology ,Biotechnology - Published
- 2007
45. Signaling mechanisms of melatonin in antiproliferation of hormone-refractory 22Rv1 human prostate cancer cells: implications for prostate cancer chemoprevention
- Author
-
Kwok-Ming Yao, Chun W. Tam, Stephen Y. W. Shiu, and Chi W. Mo
- Subjects
Male ,medicine.medical_specialty ,medicine.drug_class ,Receptor expression ,Biology ,Melatonin receptor ,Melatonin ,Prostate cancer ,Endocrinology ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Cell Proliferation ,Prostatic Neoplasms ,Receptor antagonist ,medicine.disease ,Growth Inhibitors ,Cancer cell ,Cancer research ,Signal transduction ,Luzindole ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug ,Signal Transduction - Abstract
There is an unmet clinical demand for safe and effective pharmaceuticals/nutraceuticals for prostate cancer prevention and hormone-refractory prostate cancer treatment. Previous laboratory and human studies of our laboratory demonstrated an association between the antiproliferative action of melatonin and melatonin MT(1) receptor expression in prostate cancer. The aim of this study was to determine, using a pharmacological approach, the signaling mechanisms of melatonin in hormone-refractory 22Rv1 human prostate cancer cell antiproliferation. Both immunoreactive MT(1) and MT(2) subtypes of G protein-coupled melatonin receptor were expressed in 22Rv1 cells. Melatonin inhibited, concentration dependently, cell proliferation, upregulated p27(Kip1) gene transcription and protein expression, and downregulated activated androgen signaling in 22Rv1 cells. While the effects of melatonin were mimicked by 2-iodomelatonin, a high-affinity nonselective MT(1) and MT(2) receptor agonist, melatonin effects were blocked by luzindole, a nonselective MT(1) and MT(2) receptor antagonist, but were unaffected by 4-phenyl-2-propionamidotetraline, a selective MT(2) receptor antagonist. Importantly, we discovered that the antiproliferative effect of melatonin exerted via MT(1) receptor on p27(Kip1) gene and protein upregulation is mediated by a novel signaling mechanism involving co-activation of protein kinase C (PKC) and PKA in parallel. Moreover, we also showed that a melatonin/MT(1)/PKC mechanism is involved in melatonin-induced downregulation of activated androgen signal transduction in 22Rv1 cells. Taken together with the known molecular mechanisms of prostate cancer progression and transition to androgen independence, our data provide strong support for melatonin to be a promising small-molecule useful for prostate cancer primary prevention and secondary prevention of the development and progression of hormone refractoriness.
- Published
- 2007
46. The Forkhead box M1 protein regulates the transcription of the estrogen receptor alpha in breast cancer cells
- Author
-
Kwok-Ming Yao, R. Charles Coombes, Demetra Constantinidou, Rana Varshochi, Patricia A. Madureira, Eric Lam, and Richard E. Francis
- Subjects
Transcription, Genetic ,Oligonucleotides ,Breast Neoplasms ,Biology ,Biochemistry ,Cell Line, Tumor ,Humans ,Biotinylation ,RNA, Messenger ,Molecular Biology ,Transcription factor ,Estrogen receptor beta ,Regulation of gene expression ,Models, Genetic ,Cell Cycle ,Forkhead Box Protein M1 ,Forkhead Box Protein O3 ,Estrogen Receptor alpha ,Forkhead Transcription Factors ,Cell Biology ,Molecular biology ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Ectopic expression ,FOXA1 ,Chromatin immunoprecipitation ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists - Abstract
In this study, we have identified the Forkhead transcription factor FoxM1 as a physiological regulator of estrogen receptor alpha (ERalpha) expression in breast carcinoma cells. Our survey of a panel of 16 different breast cell lines showed a good correlation (13/16) between FoxM1 expression and expression of ERalpha at both protein and mRNA levels. We have also demonstrated that ectopic expression of FoxM1 in two different estrogen receptor-positive breast cancer cell lines, MCF-7 and ZR-75-30, led to up-regulation of ERalpha expression at protein and transcript levels. Furthermore, treatment of MCF-7 cells with the MEK inhibitor U0126, which blocks ERK1/2-dependent activation of FoxM1, also repressed ERalpha expression. Consistent with this, silencing of FoxM1 expression in MCF-7 cells using small interfering RNA resulted in the almost complete abrogation of ERalpha expression. We also went on to show that FoxM1 can activate the transcriptional activity of human ERalpha promoter primarily through two closely located Forkhead response elements located at the proximal region of the ERalpha promoter. Chromatin immunoprecipitation and biotinylated oligonucleotide pulldown assays have allowed us to confirm these Forkhead response elements as important for FoxM1 binding. Further co-immunoprecipitation experiments showed that FoxO3a and FoxM1 interact in vivo. Together with the chromatin immunoprecipitation and biotinylated oligonucleotide pulldown data, the co-immunoprecipitation results also suggest the possibility that FoxM1 and FoxO3a cooperate to regulate ERalpha gene transcription.
- Published
- 2006
47. Secreted PDZD2 exerts concentration-dependent effects on the proliferation of INS-1E cells
- Author
-
Sookja K. Chung, Melissa K. Thomas, Tammy Siu Man Tam, Po Sing Leung, Siu Wai Tsang, Kwok-Ming Yao, Richard Y. M. Ma, Ada P.M. Suen, and Philip Man Lung Yeung
- Subjects
Adult ,PDZ domain ,Biochemistry ,Cell Line, Tumor ,Insulin-Secreting Cells ,medicine ,Humans ,Insulinoma ,Pancreas ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Antiserum ,biology ,Endoplasmic reticulum ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,medicine.disease ,Molecular biology ,In vitro ,Neoplasm Proteins ,medicine.anatomical_structure ,Cell culture ,Mitogen-activated protein kinase ,biology.protein ,Mitogens ,Cell Adhesion Molecules - Abstract
PDZD2 (PDZ domain containing 2) is a multi-PDZ protein expressed in pancreas and many other tissues. PDZD2 shows extensive homology to pro-interleukin-16 (pro-IL-16) and is localized mainly to the endoplasmic reticulum. We have recently demonstrated that PDZD2, like pro-IL-16, is proteolytically cleaved at its C-terminus to generate a secreted protein, sPDZD2 (for secreted PDZD2). To understand the possible functional role of PDZD2 in pancreas, we investigated the cellular distribution of PDZD2 in adult pancreas using an antiserum that recognizes both the full-length and secreted forms of PDZD2. Immunohistochemical analysis revealed a strong expression of PDZD2 in pancreatic islet beta cells but not alpha cells. Consistent with the beta-cell-enriched expression of PDZD2, immunoblot analysis indicated expression of both full-length PDZD2 and sPDZD2 in the insulinoma cell line INS-1E. A recombinant sPDZD2 protein was synthesized for study of its functional effect on INS-1E cells. In culture media with limiting serum, co-incubation with sPDZD2 stimulated the proliferation of INS-1E cells. The mitogenic effect of sPDZD2 was concentration-dependent, and was associated with a slight inhibition of the insulin promoter activity at high sPDZD2 concentrations. As a potential mitogen of beta-like cells, sPDZD2 may be useful for the optimization of beta-cell growth and differentiation in vitro.
- Published
- 2005
48. Raf/MEK/MAPK signaling stimulates the nuclear translocation and transactivating activity of FOXM1c
- Author
-
Anthony C. C. Tsang, Kwok-Ming Yao, Alice M.S. Cheung, Tommy H. K. Tong, Wai Ying Leung, and Richard Y. M. Ma
- Subjects
MAPK/ERK pathway ,Transcriptional Activation ,MAP Kinase Signaling System ,Molecular Sequence Data ,Active Transport, Cell Nucleus ,Biology ,chemistry.chemical_compound ,Mice ,Aurintricarboxylic acid ,Animals ,Humans ,Protein Isoforms ,Amino Acid Sequence ,Phosphorylation ,Cyclin B1 ,Extracellular Signal-Regulated MAP Kinases ,Mitosis ,Transcription factor ,Cell Nucleus ,Mitogen-Activated Protein Kinase Kinases ,Forkhead Box Protein M1 ,Forkhead Transcription Factors ,Cell Biology ,Cell cycle ,Molecular biology ,Cell biology ,chemistry ,FOXM1 ,Trans-Activators ,raf Kinases ,Transcription Factors - Abstract
The forkhead box (FOX) transcription factor FOXM1 is ubiquitously expressed in proliferating cells. FOXM1 expression peaks at the G2/M phase of the cell cycle and its functional deficiency in mice leads to defects in mitosis. To investigate the role of FOXM1 in the cell cycle, we used synchronized hTERT-BJ1 fibroblasts to examine the cell cycle-dependent regulation of FOXM1 function. We observed that FOXM1 is localized mainly in the cytoplasm in cells at late-G1 and S phases. Nuclear translocation occurs just before entry into the G2/M phase and is associated with phosphorylation of FOXM1. Consistent with the dependency of FOXM1 function on mitogenic signals, nuclear translocation of FOXM1 requires activity of the Raf/MEK/MAPK signaling pathway and is enhanced by the MAPK activator aurintricarboxylic acid. This activating effect was suppressed by the MEK1/2 inhibitor U0126. In transient reporter assays, constitutively active MEK1 enhances the transactivating effect of FOXM1c, but not FOXM1b, on the cyclin B1 promoter. RT-PCR analysis confirmed that different cell lines and tissues predominantly express the FOXM1c transcript. Mutations of two ERK1/2 target sequences within FOXM1c completely abolish the MEK1 enhancing effect, suggesting a direct link between Raf/MEK/MAPK signaling and FOXM1 function. Importantly, inhibition of Raf/MEK/MAPK signaling by U0126 led to suppression of FOXM1 target gene expression and delayed progression through G2/M, verifying the functional relevance of FOXM1 activation by MEK1. In summary, we provide the first evidence that Raf/MEK/MAPK signaling exerts its G2/M regulatory effect via FOXM1c.
- Published
- 2005
49. G-box binding coincides with increased Solanum melongena cysteine proteinase expression in senescent fruits and circadian-regulated leaves
- Author
-
Zeng-Fu, Xu, Mee-Len, Chye, Hong-Ye, Li, Fang-Xiu, Xu, and Kwok-Ming, Yao
- Subjects
Aging ,Genomic Library ,Base Sequence ,Molecular Sequence Data ,Solanum ,Gene Expression Regulation, Enzymologic ,Circadian Rhythm ,Plant Leaves ,Cysteine Endopeptidases ,Gene Expression Regulation, Plant ,Sequence Homology, Nucleic Acid ,Seeds ,RNA, Messenger ,Promoter Regions, Genetic ,Sequence Alignment ,DNA Primers - Abstract
We have previously shown that SmCP, the gene encoding Solanum melongena cysteine proteinase, is expressed during developmental events associated with programmed cell death (PCD) suggesting its involvement in protein degradation during these events (Xu and Chye, Plant Journal 17 (1999) 321-327). Here, we investigated the regulation of SmCP expression and showed that it is ethylene-inducible and is under circadian control. This circadian rhythm is entrained by light/dark (LD) cycling with peak expression in the late light period, as opposed to that in early light for rbcS, suggesting that protein degradation and photosynthesis are temporally separated by circadian control. Northern blot analysis shows that the pattern of ethylene induction of SmCP is consistent with our previous observation of its significantly increased expression at leaf senescence and fruit ripening when endogenous ethylene is abundant. To further understand SmCP regulation, we have cloned the SmCP promoter and identified a G-box (CACGTG) at -85/-80 by DNase I footprinting analysis of the -221/+17 region. Its specific interaction with nuclear proteins in S. melongena leaves and fruits was confirmed by competitive electrophoretic mobility shift assays using oligonucleotides containing the G-box and mutant derivatives. G-box binding activity was stronger in senescent than young fruits. In circadian-regulated leaves, stronger binding activity coincided with peak circadian expression of SmCP. This correlation between binding activity and expression suggests that G-box binding factors enhance SmCP transcription and that the G-box likely plays a role in circadian regulation of genes affected by LD cycling.
- Published
- 2003
50. Proteolytic cleavage of PDZD2 generates a secreted peptide containing two PDZ domains
- Author
-
Man Lung Yeung, Kwok-Ming Yao, Tammy Siu Man Tam, and Anthony C. C. Tsang
- Subjects
Male ,PDZ domain ,Molecular Sequence Data ,Scientific Report ,Sequence alignment ,Peptide ,Biology ,Cleavage (embryo) ,Endoplasmic Reticulum ,Kidney ,Transfection ,Biochemistry ,Cell Line ,Mice ,Genetics ,Animals ,Humans ,Secretion ,Amino Acid Sequence ,Binding site ,Molecular Biology ,Peptide sequence ,Adaptor Proteins, Signal Transducing ,chemistry.chemical_classification ,Carrier Proteins - chemistry - genetics - metabolism ,Binding Sites ,Sequence Homology, Amino Acid ,Endoplasmic reticulum ,Hydrolysis ,Prostatic Neoplasms - chemistry - metabolism ,Prostatic Neoplasms ,Molecular biology ,Recombinant Proteins ,Cell biology ,Neoplasm Proteins ,Rats ,chemistry ,Culture Media, Conditioned ,Neoplasm Proteins - chemistry - metabolism ,Carrier Proteins ,Cell Adhesion Molecules ,Sequence Alignment - Abstract
PDZD2 (PDZ-domain-containing 2; also known as PAPIN, AIPC and PIN1) is a ubiquitously expressed multi-PDZ-domain protein. We have shown that PDZD2, which shows extensive homology to pro-interleukin-16 (pro-IL-16), is localized mainly to the endoplasmic reticulum (ER). Pro-IL-16 is cleaved in a caspase-3-dependent mechanism to generate the secreted cytokine IL-16. The abundant expression of PDZD2 in the ER, and its sequence similarity to pro-IL-16, suggests that similar post-translational processing of PDZD2 may occur. Indeed, western blotting and mass spectrometry analysis of conditioned medium from cells transfected with epitope-tagged PDZD2 show that there is secretion of a PDZD2 peptide of approximately 37 kDa (sPDZD2, for secreted PDZD2) that contains two PDZ domains. Expression of PDZD2 was detected in several tissues. Furthermore, sPDZD2 secretion is suppressed by the mutation of a sequence that shows similarity to caspase recognition motifs or by treatment with a caspase inhibitor. In summary, PDZD2 is the first reported multi-PDZ protein that is processed by proteolytic cleavage to generate a secreted peptide containing two PDZ domains., published_or_final_version
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.