1
|
Kang YE, Kim JM, Lim MA, Lee SE, Yi S, Kim JT, Oh C, Liu L, Jin Y, Jung SN, Won HR, Chang JW, Lee JH, Kim HJ, Koh HY, Jun S, Cho SW, Shong M, Koo BS. Growth Differentiation Factor 15 is a Cancer Cell-Induced Mitokine That Primes Thyroid Cancer Cells for Invasiveness. Thyroid 2021; 31:772-786. [PMID: 33256569 DOI: 10.1089/thy.2020.0034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background: Mitochondrial stress is known to activate the mitochondrial unfolded protein response (UPRmt). The UPRmt results in the secretion of mitochondrial cytokines (mitokines), which can promote a hormetic response cell nonautonomously, and has been reported to be protumorigenic. Growth differentiation factor 15 (GDF15) is a well-characterized mitokine, which is reported to have a mitohormetic effect. Thus, we investigated whether GDF15 induction could prime a subpopulation of thyroid cancer cells to provide invasive advantages. Methods: The UPRmt, including mitokine expression, was assessed in the context of thyroid cancer in vitro and in vivo. GDF15 expression in 266 patients with papillary thyroid carcinoma (PTC) was determined by immunohistochemistry. The serum levels of GDF15 were measured in healthy subjects and PTC patients. In addition, our own and The Cancer Genome Atlas data were analyzed to determine the expression level of GDF15 in thyroid cancers. The role of GDF15 in tumor aggressiveness was investigated by observing the effects of GDF15 knockdown in BCPAP, TPC-1, 8505C, and FRO cells. Results: Pharmacological inhibition of mitochondrial oxidative phosphorylation function in thyroid cancer cells robustly increased GDF15 expression. The expression of GDF15 was associated with activation of the mitochondrial integrated stress response pathway in PTC patients. Circulating GDF15 levels were significantly higher in PTC patients than in the controls, and tumor expression of GDF15 was related to tumor aggressiveness. In vitro and in vivo knockdown of GDF15 in a thyroid cancer model showed decreased viability, migration, and invasion compared with the control cells via regulation of STAT3. Conclusions: In this study, we demonstrated that GDF15 is a mitokine induced in thyroid cancer cells upon mitochondrial stress. GDF15-induced STAT3 activation determined tumor progression in thyroid cancer. The GDF15-STAT3 signaling axis may be a target in aggressiveness of thyroid cancer.
Collapse
MESH Headings
- Adenoma, Oxyphilic/genetics
- Adenoma, Oxyphilic/metabolism
- Adenoma, Oxyphilic/pathology
- Cell Line, Tumor
- Gene Knockdown Techniques
- Growth Differentiation Factor 15/genetics
- Growth Differentiation Factor 15/metabolism
- Humans
- Mitochondria
- Neoplasm Invasiveness
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Thyroid Cancer, Papillary/genetics
- Thyroid Cancer, Papillary/metabolism
- Thyroid Cancer, Papillary/pathology
- Thyroid Carcinoma, Anaplastic/genetics
- Thyroid Carcinoma, Anaplastic/metabolism
- Thyroid Carcinoma, Anaplastic/pathology
- Thyroid Epithelial Cells/metabolism
- Thyroid Neoplasms/genetics
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Unfolded Protein Response
Collapse
Affiliation(s)
- Yea Eun Kang
- Department of Endocrinology and Metabolism, College of Medicine, Chungnam National University, Daejeon, South Korea
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Jin Man Kim
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Mi Ae Lim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Seong Eun Lee
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Shinae Yi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Jung Tae Kim
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chan Oh
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Lihua Liu
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yanli Jin
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Seung-Nam Jung
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Ho-Ryun Won
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jae Won Chang
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering; Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyun Jung Kim
- Graduate School of Medical Science and Engineering; Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyun Yong Koh
- Graduate School of Medical Science and Engineering; Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sangmi Jun
- Drug & Disease Target Group, Korea Basic Science Institute, Cheongju, South Korea
- Convergent Research Center for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Sun Wook Cho
- Department of Endocrinology and Metabolism, College of Medicine, Seoul National University, Seoul, South Korea
| | - Minho Shong
- Department of Endocrinology and Metabolism, College of Medicine, Chungnam National University, Daejeon, South Korea
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Bon Seok Koo
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
2
|
Debeleç Bütüner B, Öztürk MB. Use of Non-steroidal Anti-inflammatory Drugs for Chemoprevention of Inflammation-induced Prostate Cancer. Turk J Pharm Sci 2017; 14:274-279. [PMID: 32454624 DOI: 10.4274/tjps.41636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/26/2017] [Indexed: 12/01/2022]
Abstract
Objectives Chronic inflammation has been known as one of the major causes of cancer progression and 25% of cancer cases initiate due to chronic inflammation according to epidemiologic data. It has been determined that chronic inflammation induces carcinogenesis through the abrogation of cell proliferation, apoptosis, and angiogenesis mechanisms. Therefore, it is believed that inhibition of inflammation-induced carcinogenic mechanisms is an efficient therapeutic strategy in drug development studies of cancer chemoprevention. It has also been observed that use of anti-inflammatory drugs reduces the incidence of cancer, and the risk of developing prostate cancer decreases 15-20% with regular use of aspirin and non-steroidal anti-inflammatory drugs (NSAID). Materials and Methods In this study, we investigated the effects of some clinically used NSAIDs on cellular mechanisms that play a role in inflammation-induced prostate carcinogenesis. Inhibition activities on the nuclear factor kappa-B signaling pathway, which activates tumorigenic mechanisms, as well as alterations on androgen receptor signaling, which regulates the proliferation of prostate cells, were investigated. In addition, protein kinase B (Akt) activation, which is stimulated a the inflammatory microenvironment, was examined. Results The results showed that anti-inflammatory agents alter the protein levels of androgen receptors as well as tumor suppressor NKX3.1, and might trigger an unexpected increase in Akt(S473) level, which induces tumorigenesis. Conclusion It is suggested that inflammatory pathways and prostate carcinogenesis-specific mechanisms should be taken into account for the use of anti-inflammatory drugs for chemoprevention of inflammation-induced prostate cancer.
Collapse
Affiliation(s)
- Bilge Debeleç Bütüner
- Ege University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, İzmir, Turkey
| | - Mert Burak Öztürk
- Ege University, Faculty of Engineering, Department of Bioengineering, Cancer Biology Laboratory, İzmir, Turkey
| |
Collapse
|
3
|
Gitau SC, Li X, Zhao D, Guo Z, Liang H, Qian M, Lv L, Li T, Xu B, Wang Z, Zhang Y, Xu C, Lu Y, Du Z, Shan H, Yang B. Acetyl salicylic acid attenuates cardiac hypertrophy through Wnt signaling. Front Med 2015; 9:444-56. [PMID: 26626190 DOI: 10.1007/s11684-015-0421-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/04/2015] [Indexed: 12/25/2022]
Abstract
Ventricular hypertrophy is a powerful and independent predictor of cardiovascular morbid events. The vascular properties of low-dose acetyl salicylic acid (aspirin) provide cardiovascular benefits through the irreversible inhibition of platelet cyclooxygenase 1; however, the possible anti-hypertrophic properties and potential mechanism of aspirin have not been investigated in detail. In this study, healthy wild-type male mice were randomly divided into three groups and subjected to transverse aortic constriction (TAC) or sham operation. The TAC-operated mice were treated with the human equivalent of low-dose aspirin (10 mg·kg(-1)·d(-1)); the remaining mice received an equal amount of phosphate buffered saline with 0.65% ethanol, which was used as a vehicle. A cardiomyocyte hypertrophy model induced by angiotensin II (10 nmol·L(-1)) was treated with the human equivalent of low (10 or 100 μmol·L(-1)) and high (1000 μmol·L(-1)) aspirin concentrations in plasma. Changes in the cardiac structure and function were assessed through echocardiography and transmission electron microscopy. Gene expression was determined through RT-PCR and western blot analysis. Results indicated that aspirin treatment abrogated the increased thickness of the left ventricular anterior and posterior walls, the swelling of mitochondria, and the increased surface area in in vivo and in vitro hypertrophy models. Aspirin also normalized the upregulated hypertrophic biomarkers, β-myosin heavy chain (β-MHC), atrial natriuretic peptide (ANP), and b-type natriuretic peptide (BNP). Aspirin efficiently reversed the upregulation of β-catenin and P-Akt expression and the TAC- or ANG II-induced downregulation of GSK-3β. Therefore, low-dose aspirin possesses significant anti-hypertrophic properties at clinically relevant concentrations for anti-thrombotic therapy. The downregulation of β-catenin and Akt may be the underlying signaling mechanism of the effects of aspirin.
Collapse
Affiliation(s)
- Samuel Chege Gitau
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China.,Department of Pharmacy and Complementary Medicine, School of Health Sciences, Kenyatta University, P.O. BOX 43844-00100, Nairobi, Kenya
| | - Xuelian Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Dandan Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Zhenfeng Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Ming Qian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Lifang Lv
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Tianshi Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Bozhi Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Zhiguo Wang
- Institute of Cardiovascular Research, Harbin Medical University, Harbin, 150081, China
| | - Yong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Chaoqian Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China
| | - Yanjie Lu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China.,Institute of Cardiovascular Research, Harbin Medical University, Harbin, 150081, China
| | - Zhiming Du
- Institute of Clinical Pharmacy, the Second Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Hongli Shan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China.
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150081, China. .,Institute of Cardiovascular Research, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
4
|
Akrami H, Aminzadeh S, Fallahi H. Inhibitory effect of ibuprofen on tumor survival and angiogenesis in gastric cancer cell. Tumour Biol 2014; 36:3237-43. [PMID: 25542229 DOI: 10.1007/s13277-014-2952-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/04/2014] [Indexed: 12/19/2022] Open
Abstract
Numerous epidemiological studies have suggested effectiveness of long-term and regular use of non-steroidal anti-inflammatory drugs (NSAIDs), such as ibuprofen and aspirin, in preventing and treatment of certain cancers including prostate, colon, breast, lung, and gastric cancers. We have studied the potential anti-turmeric effect of ibuprofen in adenocarcinoma gastric cell line (AGS). The effects of ibuprofen were investigated on cell proliferation, apoptosis, angiogenesis, and expression of stemness marker genes using real-time RT-PCR, DNA laddering, and tube formation assays via ECM gel and human umbilical vein endothelial cells (HUVECs). Annexin-V-FLUOS and propidium iodide (PI) were used to stain the apoptotic cells. Our findings indicate that ibuprofen at the concentrations of 100, 200, 300, 400, and 500 μM is able to reduce the cancerous characteristics of the AGS cells by inducing apoptosis, inhibition of cell proliferation, and angiogenesis. Real-time RT-PCR showed that ibuprofen altered the expression of several genes including Akt, P53, PCNA, Bax, and Bcl2 in the AGS cells. In addition, reduction in CD44 and OCT3/4 transcript levels revealed that ibuprofen reduces the stemness of the AGS cells and therefore it could be used as a potential anti-tumor drug.
Collapse
Affiliation(s)
- Hassan Akrami
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran,
| | | | | |
Collapse
|
5
|
Pernicová Z, Slabáková E, Fedr R, Šimečková Š, Jaroš J, Suchánková T, Bouchal J, Kharaishvili G, Král M, Kozubík A, Souček K. The role of high cell density in the promotion of neuroendocrine transdifferentiation of prostate cancer cells. Mol Cancer 2014; 13:113. [PMID: 24884804 PMCID: PMC4229954 DOI: 10.1186/1476-4598-13-113] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 05/13/2014] [Indexed: 11/13/2022] Open
Abstract
Background Tumor heterogeneity and the plasticity of cancer cells present challenges for effective clinical diagnosis and therapy. Such challenges are epitomized by neuroendocrine transdifferentiation (NED) and the emergence of neuroendocrine-like cancer cells in prostate tumors. This phenomenon frequently arises from androgen-depleted prostate adenocarcinoma and is associated with the development of castration-resistant prostate cancer and poor prognosis. Results In this study, we showed that NED was evoked in both androgen receptor (AR)-positive and AR-negative prostate epithelial cell lines by growing the cells to a high density. Androgen depletion and high-density cultivation were both associated with cell cycle arrest and deregulated expression of several cell cycle regulators, such as p27Kip1, members of the cyclin D protein family, and Cdk2. Dual inhibition of Cdk1 and Cdk2 using pharmacological inhibitor or RNAi led to modulation of the cell cycle and promotion of NED. We further demonstrated that the cyclic adenosine 3′, 5′-monophosphate (cAMP)-mediated pathway is activated in the high-density conditions. Importantly, inhibition of cAMP signaling using a specific inhibitor of adenylate cyclase, MDL-12330A, abolished the promotion of NED by high cell density. Conclusions Taken together, our results imply a new relationship between cell cycle attenuation and promotion of NED and suggest high cell density as a trigger for cAMP signaling that can mediate reversible NED in prostate cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, v,v,i, Královopolská 135, CZ-612 65 Brno, Czech Republic.
| |
Collapse
|
6
|
Zheng Q, Zhang Y, Ren Y, Wu Y, Yang S, Zhang Y, Chen H, Li W, Zhu Y. Antiproliferative and apoptotic effects of indomethacin on human retinoblastoma cell line Y79 and the involvement of β-catenin, nuclear factor-κB and Akt signaling pathways. Ophthalmic Res 2013; 51:109-15. [PMID: 24355977 DOI: 10.1159/000355844] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 08/23/2013] [Indexed: 11/19/2022]
Abstract
BACKGROUND To determine in vitro if indomethacin inhibits proliferation and induces apoptosis in human retinoblastoma cell line Y79, and to explore possibly involved signaling pathways. METHODS The human retinoblastoma cell line Y79 was cultured with indomethacin at various concentrations (0, 25, 50, 100, 200 and 400 µmol/l). The effect of indomethacin on cell proliferation and apoptosis was examined by the Cell Counting Kit-8 and TUNEL test, respectively. The mRNA level of survivin, β-catenin and Bcl-2 was detected by RT-PCR. The protein level of survivin was measured by ELISA. Western blot was used to analyze β-catenin, nuclear factor (NF)-κB/p65, phosphorylated Akt (pAkt) and total Akt (tAkt) expression in cultured cells. RESULTS Indomethacin treatment inhibits proliferation (at concentrations from 25 to 400 µmol/l) and induces apoptosis (at concentrations from 100 to 400 µmol/l) of human retinoblastoma cell line Y79 in a dose-dependent manner. RT-PCR showed that the mRNA expression of Bcl-2 (F = 20.497; p < 0.001) and of β-catenin (F = 14.835; p < 0.001) was significantly different among the treated groups. Survivin mRNA levels remained steady, but its protein levels decreased significantly as measured by ELISA (F = 67.633; p < 0.001). Western blot analysis showed a dose-dependent downregulation of β-catenin (F = 37.411; p < 0.001), NF-κB/p65 (F = 16.302; p < 0.001) and of pAkt (F = 27.700; p < 0.001) after indomethacin treatment, while tAkt protein expression was steady among the groups. CONCLUSIONS Treatment with indomethacin can potently suppress proliferation and induce apoptosis in the retinoblastoma Y79 cell line. Wnt/β-catenin, NF-κB and Akt/PKB pathways might be implicated in the process.
Collapse
|
7
|
Sankpal UT, Abdelrahim M, Connelly SF, Lee CM, Madero-Visbal R, Colon J, Smith J, Safe S, Maliakal P, Basha R. Small molecule tolfenamic acid inhibits PC-3 cell proliferation and invasion in vitro, and tumor growth in orthotopic mouse model for prostate cancer. Prostate 2012; 72:1648-58. [PMID: 22473873 DOI: 10.1002/pros.22518] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 02/24/2012] [Indexed: 02/02/2023]
Abstract
BACKGROUND Specificity protein (Sp) transcription factors are implicated in critical cellular and molecular processes associated with cancer that impact tumor growth and metastasis. The non-steroidal anti-inflammatory drug, tolfenamic acid (TA) is known to inhibit Sp proteins in some human cancer cells and laboratory animal models. We evaluated the anti-cancer activity of TA using in vitro and in vivo models for prostate cancer. METHODS The anti-proliferative efficacy of TA was evaluated using DU-145, PC-3, and LNCaP cells. PC-3 cells were treated with DMSO or 50 µM TA for 48 hr. Whole cell lysates were evaluated for the expression of Sp1, survivin, c-PARP, Akt/p-Akt, c-Met, cdk4, cdc2, cyclin D3, and E2F1 by Western blot analysis. Cell invasion was assessed by Boyden-chamber assay and flow cytometry analysis was used to study apoptosis and cell cycle distribution. An orthotopic mouse model for prostate cancer with PC-3-Luc cells was used to study the in vivo effect of TA. RESULTS TA inhibited the expression of Sp1, c-Met, p-Akt, and survivin; increased c-PARP expression and caspases activity in PC-3 cells. TA caused cell arrest at G(0) /G(1) phase accompanied by a decrease in cdk4, cdc2, cyclin D3, and E2F1 and an increase in critical apoptotic markers. TA augmented annexin-V staining, caspase activity, and c-PARP expression indicating the activation of apoptotic pathways. TA also decreased PC-3 cell invasion. TA significantly decreased the tumor weight and volume which was associated with low expression of Sp1 and survivin in tumor sections. CONCLUSION TA targets critical pathways associated with tumorigenesis and invasion. These pre-clinical data strongly demonstrated the anti-cancer activity of TA in prostate cancer.
Collapse
Affiliation(s)
- Umesh T Sankpal
- Cancer Research Institute, MD Anderson Cancer Center Orlando, Orlando, Florida 32827, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Sample preparation and analytical strategies for large-scale phosphoproteomics experiments. Semin Cell Dev Biol 2012; 23:843-53. [DOI: 10.1016/j.semcdb.2012.05.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 05/29/2012] [Indexed: 12/28/2022]
|
9
|
Growth/differentiation factor-15: prostate cancer suppressor or promoter? Prostate Cancer Prostatic Dis 2012; 15:320-8. [PMID: 22370725 DOI: 10.1038/pcan.2012.6] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Deregulation of expression and function of cytokines belonging to the transforming growth factor-β (TGF-β) family is often associated with various pathologies. For example, this cytokine family has been considered a promising target for cancer therapy. However, the detailed functions of several cytokines from the TGF-β family that could have a role in cancer progression and therapy remain unclear. One of these molecules is growth/differentiation factor-15 (GDF-15), a divergent member of the TGF-β family. This stress-induced cytokine has been proposed to possess immunomodulatory functions and its high expression is often associated with cancer progression, including prostate cancer (PCa). However, studies clearly demonstrating the mechanisms for signal transduction and functions in cell interaction, cancer progression and therapy are still lacking. New GDF-15 roles have recently been identified for modulating osteoclast differentiation and for therapy for PCa bone metastases. Moreover, GDF-15 is as an abundant cytokine in seminal plasma with immunosuppressive properties. We discuss studies that focus on the regulation of GDF-15 expression and its role in tissue homeostasis, repair and the immune response with an emphasis on the role in PCa development.
Collapse
|
10
|
Pernicová Z, Slabáková E, Kharaishvili G, Bouchal J, Král M, Kunická Z, Machala M, Kozubík A, Souček K. Androgen depletion induces senescence in prostate cancer cells through down-regulation of Skp2. Neoplasia 2011; 13:526-36. [PMID: 21677876 PMCID: PMC3114246 DOI: 10.1593/neo.11182] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 03/09/2011] [Accepted: 03/10/2011] [Indexed: 01/22/2023]
Abstract
Although the induction of senescence in cancer cells is a potent mechanism of tumor suppression, senescent cells remain metabolically active and may secrete a broad spectrum of factors that promote tumorigenicity in neighboring malignant cells. Here we show that androgen deprivation therapy (ADT), a widely used treatment for advanced prostate cancer, induces a senescence-associated secretory phenotype in prostate cancer epithelial cells, indicated by increases in senescence-associated β-galactosidase activity, heterochromatin protein 1β foci, and expression of cathepsin B and insulin-like growth factor binding protein 3. Interestingly, ADT also induced high levels of vimentin expression in prostate cancer cell lines in vitro and in human prostate tumors in vivo. The induction of the senescence-associated secretory phenotype by androgen depletion was mediated, at least in part, by down-regulation of S-phase kinase-associated protein 2, whereas the neuroendocrine differentiation of prostate cancer cells was under separate control. These data demonstrate a previously unrecognized link between inhibition of androgen receptor signaling, down-regulation of S-phase kinase-associated protein 2, and the appearance of secretory, tumor-promoting senescent cells in prostate tumors. We propose that ADT may contribute to the development of androgen-independent prostate cancer through modulation of the tissue microenvironment by senescent cells.
Collapse
Affiliation(s)
- Zuzana Pernicová
- Department of Cytokinetics, Institute of Biophysics, AS CR, Brno, Czech Republic
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Soucek K, Slabáková E, Ovesná P, Malenovská A, Kozubík A, Hampl A. Growth/differentiation factor-15 is an abundant cytokine in human seminal plasma. Hum Reprod 2010; 25:2962-71. [PMID: 20884666 DOI: 10.1093/humrep/deq264] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Transforming growth factor-β cytokines have various biological effects in female reproductive tissue, including modulation of inflammatory response and induction of immune tolerance to seminal antigens in the reproductive tract. However, no studies have analyzed the presence of growth/differentiation factor-15 (GDF-15/macrophage inhibitory cytokine-1) in seminal fluid or demonstrated the quantity and form of GDF-15, its possible role or the relationship between its concentration and semen quality. METHODS The form and the concentration of GDF-15 were determined in 53 seminal plasma samples of both fertile and infertile men by ELISA and western blot. The sperm cells of three volunteers were treated with recombinant GDF-15, and cell viability and apoptosis were assessed by flow cytometry. The effect of GDF-15 on vaginal epithelial cells and peripheral blood mononuclear cells (PBMCs) was analyzed by quantitative RT-PCR. RESULTS The GDF-15 concentration in seminal plasma ranged from 0.2 to 6.6 μg/ml as determined by ELISA. Western blot analysis revealed that GDF-15 is present in the active form. In vitro cultivation of sperm cells with GDF-15 did not affect their viability or rates of apoptosis; however, it did inhibit proliferation of PBMCs and induce expression of FOXP3 in CD4+CD25+ cells. CONCLUSIONS To the best of our knowledge, this is the first demonstration that GDF-15 is an abundant cytokine in seminal plasma, although its concentration is not associated with semen quality or the fertility/infertility status of the donors. Moreover, our data show that GDF-15 displays immunosuppressive characteristics.
Collapse
Affiliation(s)
- Karel Soucek
- Department of Cytokinetics, Institute of Biophysics, AS CR, Brno, Czech Republic.
| | | | | | | | | | | |
Collapse
|
12
|
Current World Literature. Curr Opin Support Palliat Care 2010; 4:111-20. [DOI: 10.1097/spc.0b013e32833a1dfc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Suzuki Y, Inoue T, Ra C. NSAIDs, Mitochondria and Calcium Signaling: Special Focus on Aspirin/Salicylates. Pharmaceuticals (Basel) 2010; 3:1594-1613. [PMID: 27713319 PMCID: PMC4033999 DOI: 10.3390/ph3051594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 04/26/2010] [Accepted: 05/14/2010] [Indexed: 12/21/2022] Open
Abstract
Aspirin (acetylsalicylic acid) is a well-known nonsteroidal anti-inflammatory drug (NSAID) that has long been used as an anti-pyretic and analgesic drug. Recently, much attention has been paid to the chemopreventive and apoptosis-inducing effects of NSAIDs in cancer cells. These effects have been thought to be primarily attributed to the inhibition of cyclooxygenase activity and prostaglandin synthesis. However, recent studies have demonstrated unequivocally that certain NSAIDs, including aspirin and its metabolite salicylic acid, exert their anti-inflammatory and chemopreventive effects independently of cyclooxygenase activity and prostaglandin synthesis inhibition. It is becoming increasingly evident that two potential common targets of NSAIDs are mitochondria and the Ca2+ signaling pathway. In this review, we provide an overview of the current knowledge regarding the roles of mitochondria and Ca2+ in the apoptosis-inducing effects as well as some side effects of aspirin, salicylates and other NSAIDs, and introducing the emerging role of L-type Ca2+ channels, a new Ca2+ entry pathway in non-excitable cells that is up-regulated in human cancer cells.
Collapse
Affiliation(s)
- Yoshihiro Suzuki
- Division of Molecular Cell Immunology and Allergology, Nihon University Graduate School of Medical Science, Tokyo, Japan.
| | - Toshio Inoue
- Division of Molecular Cell Immunology and Allergology, Nihon University Graduate School of Medical Science, Tokyo, Japan
| | - Chisei Ra
- Division of Molecular Cell Immunology and Allergology, Nihon University Graduate School of Medical Science, Tokyo, Japan
| |
Collapse
|