51
|
Qian X, Zhao P, Li W, Shi ZM, Wang L, Xu Q, Wang M, Liu N, Liu LZ, Jiang BH. MicroRNA-26a promotes tumor growth and angiogenesis in glioma by directly targeting prohibitin. CNS Neurosci Ther 2013; 19:804-12. [PMID: 23870455 DOI: 10.1111/cns.12149] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 05/23/2013] [Accepted: 06/10/2013] [Indexed: 11/29/2022] Open
Abstract
BACKGROUNDS AND AIMS Glioma accounts for the majority of primary malignant brain tumors in adults. Upregulation of microRNA-26a (miR-26a) has been observed in glioma. However, the biological function and molecular mechanism of miR-26a in glioma remain to be elucidated. METHODS Glioma cells stably overexpressing or down-expressing miR-26a were analyzed for both in vitro and in vivo biological functions. Novel target of miR-26a was identified by bioinformatics searching and molecular biological assays. Glioma specimens and normal brain tissues were analyzed for both expression levels of miR-26a and its target. RESULTS Forced expression of miR-26a in glioma cells significantly increased both growth rate and colony formation in vitro and tumor growth and angiogenesis in vivo, while reduced expression of miR-26a played opposite roles. MiR-26a directly targeted prohibitin (PHB) whose expression levels were downregulated in glioma specimens. The levels of miR-26a were inversely correlated with PHB expression levels in glioma samples and strongly correlated with clinical WHO grades of glioma. CONCLUSION These results reveal that miR-26a regulates PHB and promotes glioma progression both in vitro and in vivo and that miR-26a and its target PHB are associated with glioma development, which can be helpful in developing microRNA-based treatment for glioma in the future.
Collapse
Affiliation(s)
- Xu Qian
- Department of Pathology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Strong N, Millena AC, Walker L, Chaudhary J, Khan SA. Inhibitor of differentiation 1 (Id1) and Id3 proteins play different roles in TGFβ effects on cell proliferation and migration in prostate cancer cells. Prostate 2013; 73:624-33. [PMID: 23060149 PMCID: PMC4018743 DOI: 10.1002/pros.22603] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 09/17/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND In prostate cancer cells, transforming growth factor β (TGFβ) inhibits proliferation in earlier stages of the disease; however, the cancer cells become refractory to growth inhibitory effects in advanced stages where TGFβ promotes cancer progression and metastasis. Inhibitor of differentiation (Id) family of closely related proteins (Id1-Id4) are dominant negative regulators and basic helix loop helix (bHLH) transcription factors and in general promote proliferation, and inhibit differentiation. In the present study, we have investigated the role of Id1 and Id3 proteins in the growth inhibitory effects of TGFβ on prostate cancer cells. METHODS The effect of TGF β on proliferation and Id1 and Id3 expression were investigated in PZ-HPV7, DU145, and PC3 cells. Id1 silencing through siRNA was also used in DU145 and PC3 cells to examine its role in anti-proliferative and migratory effects of TGFβ. RESULTS TGFβ increased expression of Id1 and Id3 in all cell lines followed by a later down regulation of Id1 in PZ-HPV7 expression and DU145 cells but not in PC3 cells. Id3 expression remained elevated in all three cell lines. This loss of Id1 protein correlated with an increase of CDKNI p21. Id1 knockdown in both DU145 and PC3 cells resulted in decreased proliferation. However, while TGFβ caused a further decrease in proliferation of DU145, but had no further effects in PC3 cells. Knockdown of Id1 or Id3 inhibited TGFβ1induced migration in PC3 cells. CONCLUSIONS These findings suggest an essential role of Id1 and Id3 in TGFβ1 effects on proliferation and migration in prostate cancer cells.
Collapse
Affiliation(s)
| | | | | | | | - Shafiq A. Khan
- Correspondence to: Shafiq A. Khan, PhD, Center for Cancer Research and Therapeutic Development, Clark Atlanta University, 223 James P. Brawley Dr, SW, Atlanta, GA 30314.
| |
Collapse
|
53
|
Zhou TB, Qin YH. Signaling pathways of prohibitin and its role in diseases. J Recept Signal Transduct Res 2013; 33:28-36. [PMID: 23327602 DOI: 10.3109/10799893.2012.752006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Prohibitin (PHB), appearing to be a negative regulator of cell proliferation and to be a tumor suppressor, has been connected to diverse cellular functions including cell cycle control, senescence, apoptosis and the regulation of mitochondrial activities. It is a growth regulatory gene that has pleiotropic functions in the nucleus, mitochondria and cytoplasmic compartments. However, in different tissues/cells, the expression of PHB was different, such as that it was increased in most of the cancers, but its expression was reduced in kidney diseases. Signaling pathways might be very important in the pathogenesis of diseases. This review was performed to provide a relatively complete signaling pathways flowchart for PHB to the investigators who were interested in the roles of PHB in the pathogenesis of diseases. Here, we review the signal transduction pathways of PHB and its role in the pathogenesis of diseases.
Collapse
Affiliation(s)
- Tian-Biao Zhou
- Department of Pediatric Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | | |
Collapse
|
54
|
|
55
|
Stacey MM, Cuddihy SL, Hampton MB, Winterbourn CC. Protein thiol oxidation and formation of S-glutathionylated cyclophilin A in cells exposed to chloramines and hypochlorous acid. Arch Biochem Biophys 2012; 527:45-54. [DOI: 10.1016/j.abb.2012.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/21/2012] [Accepted: 07/23/2012] [Indexed: 12/31/2022]
|
56
|
Zhu JS, Ouyang DY, Shi ZJ, Xu LH, Zhang YT, He XH. Cucurbitacin B induces cell cycle arrest, apoptosis and autophagy associated with G actin reduction and persistent activation of cofilin in Jurkat cells. Pharmacology 2012; 89:348-6. [PMID: 22699368 DOI: 10.1159/000338757] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 03/21/2012] [Indexed: 01/11/2023]
Abstract
AIM The present study aimed to explore the antitumor effect and action mechanism of cucurbitacin B (CuB) on human T-cell leukemia Jurkat cells. METHODS Cell proliferation was measured by the MTS assay. Cell cycle distribution, mitochondrial membrane potential and annexin V staining were analyzed using flow cytometry. Western blotting was used to determine the levels of apoptosis- and autophagy-related proteins. RESULTS CuB inhibited the proliferation of Jurkat cells in a dose-dependent manner and induced G 2 /M phase arrest as well as formation of tetraploid cells. Accompanied with these effects, the actin dynamics was disrupted, and cofilin, a key regulator of actin dynamics, was persistently activated (dephosphorylated). Although CuB induced around 10% cells undergoing apoptosis, most of the cells were alive after CuB treatment for 24 h. Induction of autophagy was also evident by accumulation of LC3-II. CuB-induced autophagy seemed to be a prosurvival response, since suppression of CuB-induced autophagy significantly increased the activation of caspase-3. CONCLUSION Our results demonstrated that CuB exhibited antitumor activity in Jurkat cells through induction of cell cycle arrest and apoptosis which was at least partly due to the disruption of actin dynamics.
Collapse
Affiliation(s)
- Jun-Shan Zhu
- Department of Immunobiology, Jinan University, Guangzhou, PR China
| | | | | | | | | | | |
Collapse
|
57
|
Pérez-Martínez FC, Carrión B, Lucío MI, Rubio N, Herrero MA, Vázquez E, Ceña V. Enhanced docetaxel-mediated cytotoxicity in human prostate cancer cells through knockdown of cofilin-1 by carbon nanohorn delivered siRNA. Biomaterials 2012; 33:8152-9. [PMID: 22858003 DOI: 10.1016/j.biomaterials.2012.07.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 07/20/2012] [Indexed: 12/20/2022]
Abstract
We synthesized a non-viral delivery system (f-CNH3) for small interfering RNA (siRNA) by anchoring a fourth-generation polyamidoamine dendrimer (G4-PAMAM) to carbon nanohorns (CNHs). Using this new compound, we delivered a specific siRNA designed to knockdown cofilin-1, a key protein in the regulation of cellular cytoskeleton, to human prostate cancer (PCa) cells. The carbon nanohorn (CNH) derivative was able to bind siRNA and release it in the presence of an excess of the polyanion heparin. Moreover, this hybrid nanomaterial protected the siRNA from RNAse-mediated degradation. Synthetic siRNA delivered to PCa cells by f-CNH3 decreased the cofilin-1 mRNA and protein levels to about 20% of control values. Docetaxel, the drug of choice for the treatment of PCa, produced a concentration-dependent activation of caspase-3, an increase in cell death assessed by lactate dehydrogenase release to the culture medium, cell cycle arrest and inhibition of tumor cell proliferation. All of these toxic effects were potentiated when cofilin-1 was down regulated in these cells by a siRNA delivered by the nanoparticle. This suggests that knocking down certain proteins involved in cancer cell survival and/or proliferation may potentiate the cytotoxic actions of anticancer drugs and it might be a new therapeutic approach to treat tumors.
Collapse
|
58
|
Qiu QC, Hu B, He XP, Luo Q, Tang GH, Long ZF, Chen ZC, He XS. STGC3 inhibits xenograft tumor growth of nasopharyngeal carcinoma cells by altering the expression of proteins associated with apoptosis. Genet Mol Biol 2012; 35:18-26. [PMID: 22481869 PMCID: PMC3313509 DOI: 10.1590/s1415-47572012005000009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 08/12/2011] [Indexed: 01/03/2023] Open
Abstract
STGC3 is a potential tumor suppressor that inhibits the growth of the nasopharyngeal carcinoma cell line CNE2; the expression of this protein is reduced in nasopharyngeal carcinoma compared with normal nasopharyngeal tissue. In this study, we investigated the tumor-suppressing activity of STGC3 in nude mice injected subcutaneously with Tet/pTRE-STGC3/CNE2 cells. STGC3 expression was induced by the intraperitoneal injection of doxycycline (Dox). The volume mean of Tet/pTRE-STGC3/CNE2+Dox xenografts was smaller than that of Tet/pTRE/CNE2+Dox xenografts. In addition, Tet/pTRE-STGC3/CNE2+Dox xenografts showed an increase in the percentage of apoptotic cells, a decrease in Bcl-2 protein expression and an increase in Bax protein expression. A proteomic approach was used to assess the protein expression profile associated with STGC3-mediated apoptosis. Western blotting confirmed the differential up-regulation of prohibitin seen in proteomic analysis. These results indicate that overexpression of STGC3 inhibits xenograft growth in nude mice by enhancing apoptotic cell death through altered expression of apoptosis-related proteins such as Bcl-2, Bax and prohibitin. These data contribute to our understanding of the function of STGC3 in human nasopharyngeal carcinoma and provide new clues for investigating other STGC3-associated tumors.
Collapse
Affiliation(s)
- Qing-Chao Qiu
- Cancer Research Institute, University of South China, Hengyang, China
| | | | | | | | | | | | | | | |
Collapse
|
59
|
ADF/cofilin proteins translocate to mitochondria during apoptosis but are not generally required for cell death signaling. Cell Death Differ 2011; 19:958-67. [PMID: 22139132 DOI: 10.1038/cdd.2011.180] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Non-muscle cofilin (n-cofilin) is a member of the ADF/cofilin family of actin depolymerizing proteins. Recent studies reported a mitochondrial translocation of n-cofilin during apoptosis. As these studies also revealed impaired cytochrome c release and a block in apoptosis upon small interfering RNA-mediated n-cofilin knockdown, n-cofilin was postulated to be essential for apoptosis induction. To elucidate the general importance of ADF/cofilin activity for apoptosis, we exposed mouse embryonic fibroblasts deficient for n-cofilin, ADF (actin depolymerizing factor), or all ADF/cofilin isoforms to well-characterized apoptosis inducers. Cytochrome c release, caspase-3 activation, and apoptotic chromatin condensation were unchanged in all mutant fibroblasts. Thus, we conclude that ADF/cofilin activity is not generally required for induction or progression of apoptosis in mammalian cells. Interestingly, mitochondrial association of ADF and n-cofilin during apoptosis was preceded by, and dependent on, actin that translocated by a yet unknown mechanism to mitochondria during cell death.
Collapse
|
60
|
Proteomic analysis of cancer stem cells in human prostate cancer cells. Biochem Biophys Res Commun 2011; 412:279-85. [PMID: 21820414 DOI: 10.1016/j.bbrc.2011.07.083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 07/20/2011] [Indexed: 01/06/2023]
Abstract
Results from recent studies support the hypothesis that cancer stem cells (CSCs) are responsible for tumor initiation and formation. Here, we applied a proteome profiling approach to investigate the mechanisms of CSCs and to identify potential biomarkers in the prostate cancer cell line DU145. Using MACS, the DU145 prostate cancer cell line was isolated into CD44+ or CD44- cells. In sphere culture, CD44+ cells possessed stem cell characteristics and highly expressed genes known to be important in stem cell maintenance. In addition, they showed strong tumorigenic potential in the clonogenic assay and soft agar colony formation assay. We then analyzed and identified proteins that were differentially expressed between CD44+ and CD44- using two-dimensional gel electrophoresis and LC-MS/MS. Cofilin and Annexin A5, which are associated with proliferation or metastasis in cancer, were found to be positively correlated with CD44 expression. These results provide information that will be important to the development of new cancer diagnostic tools and understanding the mechanisms of CSCs although a more detailed study is necessary to investigate the roles of Cofilin and Annexin A5 in CSCs.
Collapse
|
61
|
Kuželová K, Pluskalová M, Grebeňová D, Pavlásková K, Halada P, Hrkal Z. Changes in cell adhesivity and cytoskeleton-related proteins during imatinib-induced apoptosis of leukemic JURL-MK1 cells. J Cell Biochem 2011; 111:1413-25. [PMID: 20830748 DOI: 10.1002/jcb.22868] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The fusion protein Bcr-Abl, which is the molecular cause of chronic myelogenous leukemia (CML) interacts in multiple points with signaling pathways regulating the cellular adhesivity and cytoskeleton architecture and dynamics. We explored the effects of imatinib mesylate, an inhibitor of Bcr-Abl protein used in front-line CML therapy, on the adhesivity of JURL-MK1 cells to fibronectin and searched for underlying changes in the cell proteome. As imatinib induces apoptosis of JURL-MK1 cells, we used three different caspase inhibitors to discriminate between direct consequences of Bcr-Abl inhibition and secondary changes related to the apoptosis. Imatinib treatment caused a transient increase in JURL-MK1 cell adhesivity to fibronectin, possibly due to the switch off of Bcr-Abl activity. Subsequently, we observed a number of changes including a decrease in cell adhesivity, F-actin decomposition, reduction of integrin β1, CD44, and paxillin expression levels and a marked increase in cofilin phophorylation at Ser3. These events were generally related to the proceeding apoptosis but they differed in their sensitivity to the individual caspase inhibitors.
Collapse
Affiliation(s)
- K Kuželová
- Department of Cellular Biochemistry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
62
|
Hirota M, Motoyama A, Suzuki M, Yanagi M, Kitagaki M, Kouzuki H, Hagino S, Itagaki H, Sasa H, Kagatani S, Aiba S. Changes of cell-surface thiols and intracellular signaling in human monocytic cell line THP-1 treated with diphenylcyclopropenone. J Toxicol Sci 2011; 35:871-9. [PMID: 21139337 DOI: 10.2131/jts.35.871] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Changes of cell-surface thiols induced by chemical treatment may affect the conformations of membrane proteins and intracellular signaling mechanisms. In our previous study, we found that a non-toxic dose of diphenylcyclopropene (DPCP), which is a potent skin sensitizer, induced an increase of cell-surface thiols in cells of a human monocytic cell line, THP-1. Here, we examined the influence of DPCP on intracellular signaling. First, we confirmed that DPCP induced an increase of cell-surface thiols not only in THP-1 cells, but also in primary monocytes. The intracellular reduced-form glutathione/oxidized-form glutathione ratio (GSH/GSSG ratio) was not affected by DPCP treatment. By means of labeling with a membrane-impermeable thiol-reactive compound, Alexa Fluor 488 C5 maleimide (AFM), followed by two-dimensional gel electrophoresis and analysis by liquid chromatography coupled with electrospray tandem mass spectrometry (LC/MS/MS), we identified several proteins whose thiol contents were modified in response to DPCP. These proteins included cell membrane components, such as actin and β-tubulin, molecular chaperones, such as heat shock protein 27A and 70, and endoplasmic reticulum (ER) stress-inducible proteins. Next, we confirmed the expression in DPCP-treated cells of spliced XBP1, a known marker of ER stress. We also detected the phosphorylation of SAPK/JNK and p38 MAPK, which are downstream signaling molecules in the IRE1α-ASK1 pathway, which is activated by ER stress. These data suggested that increase of cell-surface thiols might be associated with activation of ER stress-mediated signaling.
Collapse
|
63
|
Overexpression of cofilin 1 can predict progression-free survival in patients with epithelial ovarian cancer receiving standard therapy. Hum Pathol 2011; 42:516-21. [PMID: 21237490 DOI: 10.1016/j.humpath.2010.07.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 07/12/2010] [Accepted: 07/16/2010] [Indexed: 11/23/2022]
Abstract
The aim of this study was to examine the relation between cofilin 1 expression and progression-free survival in advanced epithelial ovarian cancer. We performed quantitative reverse transcriptase polymerase chain reaction and immunohistochemical analysis in 78 patients with advanced epithelial ovarian cancer (excluding those with mucinous and clear-cell types). All patients received the standard therapy, including staging laparotomy and adjuvant chemotherapy consisting of carboplatin and paclitaxel. Of 78 samples, RNA from 62 samples was available for reverse transcriptase polymerase chain reaction analysis. We defined cofilin 1 high expression as relative gene expression equal to or higher than the median and low expression as gene expression lower than median. The progression-free survival was longer in cofilin 1 low-expression cases than in high-expression cases (P = .039). Multivariate analysis demonstrated that stage and cofilin 1 expression were significant predictors of progression-free survival. Of the 78 samples, 54 were appropriate for immunohistochemical study. In 35 of those 54 cases, cofilin 1 protein expression was detected. The progression-free survival was longer in cofilin 1 protein-negative cases than in protein-positive cases (P = .042). Expression of cofilin 1 may predict the progression-free survival of patients with advanced epithelial ovarian cancer receiving standard therapy.
Collapse
|
64
|
Abstract
Prohibitin-1 (PHB, also known as PHB1), a member of the Band-7 family of proteins, is highly conserved evolutionarily, widely expressed, and present in different cellular compartments. Genetic studies with different organism models have provided strong evidence for an important biological role of PHB in mitochondrial function, cell proliferation, and development. Recent discoveries regarding the involvement of PHB in phophatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) and transforming growth factor-β (TGF-β)/signal transducers and activators of transcription signaling pathways, and earlier reports on the interaction of PHB with Raf and its critical role in Ras/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling opened up the possibility that PHB has functions outside of the mitochondria (extramitochondrial) and may be a multifunctional protein. The PI3K/Akt and Ras/MAPK/ERK signaling cascades are versatile signaling processes that diverge from the same receptor tyrosine kinase root, and are involved in cell metabolism, proliferation, and development. Here, we review the emerging role of PHB and its post-translational modifications in signal transduction pathways, especially in PI3K/Akt and Ras/MAPK/ERK signaling. A recent discovery of opposing effects of PHB on longevity under different metabolic states and its potential connection with insulin/insulin-like growth factor-I signaling is also discussed.
Collapse
Affiliation(s)
- Suresh Mishra
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| | | | | |
Collapse
|
65
|
Wabnitz GH, Goursot C, Jahraus B, Kirchgessner H, Hellwig A, Klemke M, Konstandin MH, Samstag Y. Mitochondrial translocation of oxidized cofilin induces caspase-independent necrotic-like programmed cell death of T cells. Cell Death Dis 2010; 1:e58. [PMID: 21364663 PMCID: PMC3032559 DOI: 10.1038/cddis.2010.36] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Oxidative stress leads to T-cell hyporesponsiveness or death. The actin-binding protein cofilin is oxidized during oxidative stress, which provokes a stiff actin cytoskeleton and T-cell hyporesponsiveness. Here, we show that long-term oxidative stress leads to translocation of cofilin into the mitochondria and necrotic-like programmed cell death (PCD) in human T cells. Notably, cofilin mutants that functionally mimic oxidation by a single mutation at oxidation-sensitive cysteins (Cys-39 or Cys-80) predominately localize within the mitochondria. The expression of these mutants alone ultimately leads to necrotic-like PCD in T cells. Accordingly, cofilin knockdown partially protects T cells from the fatal effects of long-term oxidative stress. Thus, we introduce the oxidation and mitochondrial localization of cofilin as the checkpoint for necrotic-like PCD upon oxidative stress as it occurs, for example, in tumor environments.
Collapse
Affiliation(s)
- G H Wabnitz
- Institute for Immunology, Ruprecht-Karls-University, Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Wang XJ, Zhu YJ, Cui JG, Huang X, Gu J, Xu H, Wen H. Proteomic analysis of human umbilical vein endothelial cells incubated with Cryptococcus neoformans var. neoformans. Mycoses 2010; 54:e336-43. [PMID: 21910755 DOI: 10.1111/j.1439-0507.2010.01920.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cryptococcus neoformans is a medically important fungus and can infect all the organs of the body. As vascular endothelial cell is an important target for C. neoformans to penetrate any organs, the differential protein expression of human umbilical vascular endothelial cell (HUVEC) after incubating with C. neoformans may be the key to penetration. The proteins of HUVECs incubated with C. neoformans and normal HUVECs were collected and purified. After two-dimensional electrophoresis, the differential protein expression was identified by matrix-assisted laser desorption/ionisation mass spectrometry. The mRNA levels of some proteins were measured by real-time PCR. Three proteins were found significantly overexpressed in HUVECs incubated with C. neoformans, and nine other proteins were downregulated. The mRNA levels of S100A10 and peroxiredoxin I fluctuated with the protein levels. These results suggested that the expressions of peroxiredoxin I and S100A10 were regulated during the process of invasion of HUVECs by C. neoformans. We hypothesise that these proteins take part in the modifications of HUVEC cytoskeleton and the tolerance to oxidative stress, which may affect the process of invasion by C. neoformans.
Collapse
Affiliation(s)
- Xiao-Jun Wang
- Mycology Center and Department of Dermatology, Changzheng Hospital, Secondary Military Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
67
|
Hsieh YC, Rao YK, Wu CC, Huang CYF, Geethangili M, Hsu SL, Tzeng YM. Methyl Antcinate A from Antrodia camphorata Induces Apoptosis in Human Liver Cancer Cells through Oxidant-Mediated Cofilin- and Bax-Triggered Mitochondrial Pathway. Chem Res Toxicol 2010; 23:1256-67. [PMID: 20557081 DOI: 10.1021/tx100116a] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yun-Chih Hsieh
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China, Institute of Biochemical Sciences and Technology, Chaoyang University of Technology, Wufeng, Taiwan, Republic of China, Institute of Medical and Molecular Toxicology, Chung Shan Medical University, Taichung, Taiwan, Republic of China, and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Yerra Koteswara Rao
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China, Institute of Biochemical Sciences and Technology, Chaoyang University of Technology, Wufeng, Taiwan, Republic of China, Institute of Medical and Molecular Toxicology, Chung Shan Medical University, Taichung, Taiwan, Republic of China, and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Chun-Chi Wu
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China, Institute of Biochemical Sciences and Technology, Chaoyang University of Technology, Wufeng, Taiwan, Republic of China, Institute of Medical and Molecular Toxicology, Chung Shan Medical University, Taichung, Taiwan, Republic of China, and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Chi-Ying F. Huang
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China, Institute of Biochemical Sciences and Technology, Chaoyang University of Technology, Wufeng, Taiwan, Republic of China, Institute of Medical and Molecular Toxicology, Chung Shan Medical University, Taichung, Taiwan, Republic of China, and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Madamanchi Geethangili
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China, Institute of Biochemical Sciences and Technology, Chaoyang University of Technology, Wufeng, Taiwan, Republic of China, Institute of Medical and Molecular Toxicology, Chung Shan Medical University, Taichung, Taiwan, Republic of China, and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Shih-Lan Hsu
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China, Institute of Biochemical Sciences and Technology, Chaoyang University of Technology, Wufeng, Taiwan, Republic of China, Institute of Medical and Molecular Toxicology, Chung Shan Medical University, Taichung, Taiwan, Republic of China, and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| | - Yew-Min Tzeng
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China, Institute of Biochemical Sciences and Technology, Chaoyang University of Technology, Wufeng, Taiwan, Republic of China, Institute of Medical and Molecular Toxicology, Chung Shan Medical University, Taichung, Taiwan, Republic of China, and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Republic of China
| |
Collapse
|
68
|
Zhu B, Zhai J, Zhu H, Kyprianou N. Prohibitin regulates TGF-beta induced apoptosis as a downstream effector of Smad-dependent and -independent signaling. Prostate 2010; 70:17-26. [PMID: 19725029 PMCID: PMC3762596 DOI: 10.1002/pros.21033] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Prohibitin (PHB), a protein located on the inner mitochondrial membrane and nuclei, is an intracellular effector of transforming growth factor-beta (TGF-beta) signaling in prostate cancer cells. This study investigated the involvement of PHB in the apoptosis and survival outcomes of human prostate cancer cell to TGF-beta. shRNA PHB loss of function in prostate cancer cells led to enhanced apoptotic response to TGF-beta via Smad-dependent mechanism. METHOD TGF-beta activation of Raf-Erk intracellular signaling, led to PHB phosphorylation, decreased inner mitochondrial permeability, and increased cell survival. Calcein-based immunofluorescence studies revealed the functional involvement of PHB in maintaining inner mitochondrial membrane permeability as an integral component of TGF-beta induced apoptosis in prostate cancer cells. RESULTS These finding indicates that induction of TGF-beta apoptosis is mediated by Smad-dependent and Smad-independent signaling (MAPK) converging at PHB as a downstream effector regulating inner mitochondrial permeability. Putative PHB associated proteins were identified by subjecting TGF-beta treated cells to immunoprecipitation with anti-PHB, and mass spectrometry. A screen for the kinase specific phosphorylation sites of PHB revealed three protein kinase (PKC) binding sites. CONCLUSION Our results demonstrate that TGF-beta led to upregulation of the PKC inhibitor 14-3-3 protein and promoted its association with PHB, while PHB association with PKC-delta, was inhibited by the MEK1 inhibitor, documenting a critical interdependence between the MEK-ERK signaling and prohibitin phosphorylation. These findings suggest a dual role for PHB as a downstream determinant of the cellular response to TGF-beta via Smad-dependent pathway (apoptosis) and MAPK intracellular signaling (survival).
Collapse
Affiliation(s)
- Brian Zhu
- Department of Surgery, Division of Urology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | | | | | | |
Collapse
|
69
|
Chen S, Gao X, Sun Y, Xu C, Wang L, Zhou T. Analysis of HK-2 cells exposed to oxalate and calcium oxalate crystals: proteomic insights into the molecular mechanisms of renal injury and stone formation. ACTA ACUST UNITED AC 2009; 38:7-15. [DOI: 10.1007/s00240-009-0226-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 10/06/2009] [Indexed: 11/30/2022]
|
70
|
Klamt F, Zdanov S, Levine RL, Pariser A, Zhang Y, Zhang B, Yu LR, Veenstra TD, Shacter E. Oxidant-induced apoptosis is mediated by oxidation of the actin-regulatory protein cofilin. Nat Cell Biol 2009; 11:1241-6. [PMID: 19734890 DOI: 10.1038/ncb1968] [Citation(s) in RCA: 183] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 07/13/2009] [Indexed: 12/26/2022]
Abstract
Physiological oxidants that are generated by activated phagocytes comprise the main source of oxidative stress during inflammation. Oxidants such as taurine chloramine (TnCl) and hydrogen peroxide (H(2)O(2)) can damage proteins and induce apoptosis, but the role of specific protein oxidation in this process has not been defined. We found that the actin-binding protein cofilin is a key target of oxidation. When oxidation of this single regulatory protein is prevented, oxidant-induced apoptosis is inhibited. Oxidation of cofilin causes it to lose its affinity for actin and to translocate to the mitochondria, where it induces swelling and cytochrome c release by mediating opening of the permeability transition pore (PTP). This occurs independently of Bax activation and requires both oxidation of cofilin Cys residues and dephosphorylation at Ser 3. Knockdown of endogenous cofilin using targeted siRNA inhibits oxidant-induced apoptosis, which is restored by re-expression of wild-type cofilin but not by cofilin containing Cys to Ala mutations. Exposure of cofilin to TnCl results in intramolecular disulphide bonding and oxidation of Met residues to Met sulphoxide, but only Cys oxidation causes cofilin to induce mitochondrial damage.
Collapse
Affiliation(s)
- Fábio Klamt
- Laboratory of Biochemistry, Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Nedd4L expression is downregulated in prostate cancer compared to benign prostatic hyperplasia. Eur J Surg Oncol 2009; 35:527-31. [DOI: 10.1016/j.ejso.2008.09.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Revised: 09/06/2008] [Accepted: 09/30/2008] [Indexed: 11/24/2022] Open
|
72
|
Jones E, Pu H, Kyprianou N. Targeting TGF-beta in prostate cancer: therapeutic possibilities during tumor progression. Expert Opin Ther Targets 2009; 13:227-34. [PMID: 19236240 DOI: 10.1517/14728220802705696] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND TGF-beta regulates prostate growth by inhibiting epithelial cell proliferation and inducing apoptosis through eliciting a dynamic signaling pathway. In metastatic prostate cancer, however, TGF-beta serves as a tumor promoter. TGF-beta engages Smad-dependent and Smad-independent mechanisms to exert its action. During prostate tumorigenesis, prostate cells exhibit loss or mutation of TGF-beta transmembrane receptors. Increased production of TGF-beta causes immunosuppression, extracellular matrix degradation, epithelia to mesenchymal transition and angiogenesis that promotes tumor cell invasion and metastasis. OBJECTIVE The molecular basis for effective therapeutic targeting of TGF-beta must be directed towards the double-edge-sword nature of the cytokine: inhibiting the TGF-beta tumor promoter capabilities in advanced metastatic prostate cancer, although retaining the growth-inhibitory abilities exhibited in early stages of prostate tumorigenesis. RESULTS/CONCLUSION The current understanding of the therapeutic possibilities of targeting TGF-beta signaling during prostate tumor progression is built on preclinical studies. Studies targeting TGF-beta signaling pathway for the treatment of several human malignancies include the use of neutralizing antibodies, antisense oligonucelotides and small molecule inhibitors of kinase activity of the receptor complex. This review focuses on exploiting the therapeutic potential of targeting TGF-beta signaling in the context of its contribution to prostate cancer initiation and progression to metastasis.
Collapse
Affiliation(s)
- Elisabeth Jones
- University of Kentucky, Surgery/Urology and Markey Cancer Center, 306 Combs Building, 800 Rose Street, Lexington, KY 40536, USA
| | | | | |
Collapse
|
73
|
Keenan J, Murphy L, Henry M, Meleady P, Clynes M. Proteomic analysis of multidrug-resistance mechanisms in adriamycin-resistant variants of DLKP, a squamous lung cancer cell line. Proteomics 2009; 9:1556-66. [DOI: 10.1002/pmic.200800633] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
74
|
Abstract
Androgens promote the growth and differentiation of prostate cells through ligand activation of the androgen receptor (AR). Sensitization of the androgenic response by multifunctional growth factor signaling pathways is one of the mechanisms via which AR contributes to the emergence of androgen-independent prostate tumors. The ability of AR to cross-talk with key growth factor signaling events toward the regulation of cell cycle, apoptosis, and differentiation outcomes in prostate cancer cells is established. In this paper, we review the functional interaction between AR and an array of growth factor signal transduction events (including epidermal growth factor; fibroblast growth factor; IGF1; vascular endothelial growth factor; transforming growth factor-beta) in prostate tumors. The significance of this derailed cross-talk between androgens and key signaling networks in prostate cancer progression and its value as a therapeutic forum targeting androgen-independent metastatic prostate cancer is discussed.
Collapse
Affiliation(s)
- Meng-Lei Zhu
- Departments of Urology and Toxicology, University of Kentucky College of Medicine, University of Kentucky Medical Center, Combs Research Building Room 306, Lexington, Kentucky 40536, USA
| | | |
Collapse
|
75
|
Cofilin immunolabelling correlates with depth of invasion in gastrointestinal endocrine cell tumors. Acta Histochem 2008; 112:101-6. [PMID: 19004480 DOI: 10.1016/j.acthis.2008.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Revised: 07/22/2008] [Accepted: 07/24/2008] [Indexed: 11/22/2022]
Abstract
Gastrointestinal endocrine cell tumors are a heterogeneous population of lesions believed to arise from neuroendocrine cells of the gastrointestinal mucosa. The current classification of these tumors is based on tumor size, microscopic features and clinical evidence of metastasis. Although diagnostic categories generally correlate with prognosis, molecular prognostic markers will be clinically useful adjuncts. Cofilin has been implicated in tumor invasion, and its immunolocalisation was studied in gastrointestinal endocrine cell tumors. The immunolocalisation of cofilin was studied by immunohistochemistry in 34 formalin-fixed, paraffin wax-embedded gastrointestinal endocrine cell tumors using a tissue microarray platform. A significant correlation was found between high cofilin immunolabelling and the depth of invasion (p<0.05). Our findings suggest that cofilin might be useful clinically as a molecular prognostic adjunct in the evaluation of gastrointestinal endocrine cell tumors.
Collapse
|
76
|
Ummanni R, Junker H, Zimmermann U, Venz S, Teller S, Giebel J, Scharf C, Woenckhaus C, Dombrowski F, Walther R. Prohibitin identified by proteomic analysis of prostate biopsies distinguishes hyperplasia and cancer. Cancer Lett 2008; 266:171-85. [DOI: 10.1016/j.canlet.2008.02.047] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 02/18/2008] [Accepted: 02/19/2008] [Indexed: 10/22/2022]
|
77
|
Zhu ML, Partin JV, Bruckheimer EM, Strup SE, Kyprianou N. TGF-beta signaling and androgen receptor status determine apoptotic cross-talk in human prostate cancer cells. Prostate 2008; 68:287-95. [PMID: 18163430 DOI: 10.1002/pros.20698] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND A signaling interaction between transforming growth factor-beta (TGF-beta) and androgens promotes apoptosis in human prostate cancer cells LNCaP-TbetaRII (androgen-sensitive and TGF-beta responsive). This study investigated the contribution of androgen receptor (AR) in the combined effect of TGF-beta and dihydrotestosterone (DHT), on regulation of apoptosis and AR- and TGF-beta mediated transcriptional activity in human prostate cancer cells. METHODS Transcriptional activation in response to TGF-beta (5 ng/ml) and DHT (1 nM) was evaluated using transient transfections and luciferase assays in human prostate cancer cells, LNCaP-TbetaRII and PC-3, overexpressing the wild type AR. The apoptotic response to DHT/TGFbeta treatment was correlated with AR cellular distribution and the AR interaction with TGF-beta intracellular effector Smad4. RESULTS The results revealed that TGF-beta signaling induced AR-mediated transcriptional activation in two androgen-responsive promoters [probasin and prostate specific antigen (PSA)]. TGF-beta1 induced transcriptional activity enhanced by DHT in both cell lines (LNCaP-TbetaRII and PC-3-AR) via AR-Smad4 interaction. This interaction however does not exclusively drive TGF-beta mediated apoptosis as DHT failed to enhance such an effect in PC-3 AR (wt) cells. CONCLUSIONS These results demonstrate that the AR status determines the sensitivity of prostate cancer cells to the apoptotic effects of TGF-beta1, thus providing a new insight into the mechanism via which TGF-beta cross-sections the AR axis toward the functional convergence of the two pathways in the development of androgen-independent prostate cancer. This study is potentially significant in defining the contribution of AR status to the emergence of androgen-independent prostate tumors.
Collapse
MESH Headings
- Androgen-Binding Protein/metabolism
- Apoptosis/drug effects
- Apoptosis/physiology
- Blotting, Western
- Cell Line, Tumor
- Dihydrotestosterone/pharmacology
- Drug Synergism
- Humans
- Luciferases/genetics
- Luciferases/metabolism
- Male
- Microscopy, Fluorescence
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Protein Serine-Threonine Kinases/metabolism
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Androgen/biosynthesis
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction
- Smad4 Protein/metabolism
- Transcriptional Activation/drug effects
- Transfection
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- Meng-Lei Zhu
- Division of Urology, Department of Surgery, University of Kentucky Medical Center, Lexington, Kentucky 40536, USA
| | | | | | | | | |
Collapse
|
78
|
|
79
|
Cheng CJ, Wu YC, Shu JA, Ling TY, Kuo HC, Wu JY, Chang EE, Chang SC, Huang YH. Aberrant expression and distribution of the OCT-4 transcription factor in seminomas. J Biomed Sci 2007; 14:797-807. [PMID: 17682839 DOI: 10.1007/s11373-007-9198-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Accepted: 07/19/2007] [Indexed: 10/23/2022] Open
Abstract
Testicular germ cell tumors (TGCTs), comprised of seminomas and non-seminomas, are derived from premalignant and noninvasive intracellular germ cell neoplasias. Among TGCTs, seminomas are believed to resemble a transformed state of primordial germ cells (PGCs) and are known to exhibit a gene expression profile similar to that of embryonic stem (ES) cells, such as transcription factor OCT-4. OCT-4 has recently been recognized as a diagnostic marker for clinical aspects of seminomas. However, the role of the OCT-4 protein in seminomas has not been clarified. To determine a possible role of the OCT-4 protein in seminomas, in this paper, we studied a series of 41 testicular tumor tissues and four cell lines by immunohistochemistry, Western blotting, and reverse-transcriptase polymerase chain reaction (RT-PCR) to examine the expression and distribution of the OCT-4 transcription factor in seminomas. By utilizing immunohistochemical staining and Western blotting, we demonstrated that the OCT-4 transcription factor was aberrantly localized in the cytoplasm and nuclei of cells in the collected seminoma tissues. This observation was further confirmed using immunocytochemical staining of NCCIT (seminoma-embryonal carcinoma) and NT2 (embryonal carcinoma) cells. In addition, the RT-PCR results indicated that Oct-4 mRNA was relatively highly expressed in NCCIT, NT2 cells, and seminoma tissues when compared with human embryonic stem cells. The aberrant expression and distribution of the OCT-4 transcription factor in seminomas may provide some important clues concerning the cell transformation between germ line stem cells (like PGC) and testicular germ cell tumors.
Collapse
Affiliation(s)
- Chien-Jui Cheng
- Department of Pathology, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Browman DT, Hoegg MB, Robbins SM. The SPFH domain-containing proteins: more than lipid raft markers. Trends Cell Biol 2007; 17:394-402. [PMID: 17766116 DOI: 10.1016/j.tcb.2007.06.005] [Citation(s) in RCA: 265] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 06/06/2007] [Accepted: 06/06/2007] [Indexed: 01/03/2023]
Abstract
Membrane microdomains with distinct lipid compositions, called lipid rafts, represent a potential mechanism for compartmentalizing cellular functions within the plane of biological membranes. SPFH domain-containing proteins are found in lipid raft microdomains in diverse cellular membranes. The functions of these proteins are just beginning to be elucidated. Recent advances in the understanding of structural features and their roles within lipid rafts include a potential function for SPFH proteins in the formation of membrane microdomains and lipid raft-associated processes, such as endocytosis and mechanosensation.
Collapse
Affiliation(s)
- Duncan T Browman
- Southern Alberta Cancer Research Institute, Department of Oncology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | | | | |
Collapse
|
81
|
Rojas-Cartagena C, Ortíz-Pineda P, Ramírez-Gómez F, Suárez-Castillo EC, Matos-Cruz V, Rodríguez C, Ortíz-Zuazaga H, García-Arrarás JE. Distinct profiles of expressed sequence tags during intestinal regeneration in the sea cucumber Holothuria glaberrima. Physiol Genomics 2007; 31:203-15. [PMID: 17579180 PMCID: PMC2866185 DOI: 10.1152/physiolgenomics.00228.2006] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Repair and regeneration are key processes for tissue maintenance, and their disruption may lead to disease states. Little is known about the molecular mechanisms that underline the repair and regeneration of the digestive tract. The sea cucumber Holothuria glaberrima represents an excellent model to dissect and characterize the molecular events during intestinal regeneration. To study the gene expression profile, cDNA libraries were constructed from normal, 3-day, and 7-day regenerating intestines of H. glaberrima. Clones were randomly sequenced and queried against the nonredundant protein database at the National Center for Biotechnology Information. RT-PCR analyses were made of several genes to determine their expression profile during intestinal regeneration. A total of 5,173 sequences from three cDNA libraries were obtained. About 46.2, 35.6, and 26.2% of the sequences for the normal, 3-days, and 7-days cDNA libraries, respectively, shared significant similarity with known sequences in the protein database of GenBank but only present 10% of similarity among them. Analysis of the libraries in terms of functional processes, protein domains, and most common sequences suggests that a differential expression profile is taking place during the regeneration process. Further examination of the expressed sequence tag dataset revealed that 12 putative genes are differentially expressed at significant level (R > 6). Experimental validation by RT-PCR analysis reveals that at least three genes (unknown C-4677-1, melanotransferrin, and centaurin) present a differential expression during regeneration. These findings strongly suggest that the gene expression profile varies among regeneration stages and provide evidence for the existence of differential gene expression.
Collapse
|
82
|
Mostaghel EA, Montgomery RB, Lin DW. The basic biochemistry and molecular events of hormone therapy. Curr Urol Rep 2007; 8:224-32. [PMID: 17459272 DOI: 10.1007/s11934-007-0010-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Data regarding the molecular response of prostate cancer to hormone therapy continue to emerge, identifying a complex network of autocrine and paracrine signaling events mediating the tumor response to androgen suppression. Emerging data provide insight into cellular pathways important in the apoptotic response to therapy, including the transforming growth factor-beta, insulin-like growth factor-1, and vascular endothelial growth factor signaling axes. They also reveal mechanisms of direct antitumor cytotoxicity mediated by various hormonal agents and highlight the importance of developing antiandrogens capable of irreversibly inhibiting the androgen receptor. Accumulated data emphasize the presence of residual androgens and persistent activation of androgen receptor signaling in advanced prostate tumors despite castration. These factors suggest that a multitargeted treatment approach designed to ablate all contributions to the androgen receptor signaling axis within the prostate tumor microenvironment will be required in order for hormonal therapy to achieve optimal antitumor efficacy.
Collapse
Affiliation(s)
- Elahe A Mostaghel
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98109, USA.
| | | | | |
Collapse
|