1
|
Colussi DM, Stathopulos PB. The mitochondrial calcium uniporter: Balancing tumourigenic and anti-tumourigenic responses. J Physiol 2024; 602:3315-3339. [PMID: 38857425 DOI: 10.1113/jp285515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
Increased malignancy and poor treatability associated with solid tumour cancers have commonly been attributed to mitochondrial calcium (Ca2+) dysregulation. The mitochondrial Ca2+ uniporter complex (mtCU) is the predominant mode of Ca2+ uptake into the mitochondrial matrix. The main components of mtCU are the pore-forming mitochondrial Ca2+ uniporter (MCU) subunit, MCU dominant-negative beta (MCUb) subunit, essential MCU regulator (EMRE) and the gatekeeping mitochondrial Ca2+ uptake 1 and 2 (MICU1 and MICU2) proteins. In this review, we describe mtCU-mediated mitochondrial Ca2+ dysregulation in solid tumour cancer types, finding enhanced mtCU activity observed in colorectal cancer, breast cancer, oral squamous cell carcinoma, pancreatic cancer, hepatocellular carcinoma and embryonal rhabdomyosarcoma. By contrast, decreased mtCU activity is associated with melanoma, whereas the nature of mtCU dysregulation remains unclear in glioblastoma. Furthermore, we show that numerous polymorphisms associated with cancer may alter phosphorylation sites on the pore forming MCU and MCUb subunits, which cluster at interfaces with EMRE. We highlight downstream/upstream biomolecular modulators of MCU and MCUb that alter mtCU-mediated mitochondrial Ca2+ uptake and may be used as biomarkers or to aid in the development of novel cancer therapeutics. Additionally, we provide an overview of the current small molecule inhibitors of mtCU that interact with the Asp residue of the critical Asp-Ile-Met-Glu motif or through other allosteric regulatory mechanisms to block Ca2+ permeation. Finally, we describe the relationship between MCU- and MCUb-mediating microRNAs and mitochondrial Ca2+ uptake that should be considered in the discovery of new treatment approaches for cancer.
Collapse
Affiliation(s)
- Danielle M Colussi
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
2
|
Giri P, Camarillo I, Sundararajan R. Synergistic Enhancement of TNBC Treatment in African American Women: Integrating Resveratrol with Electrical Pulse Therapy. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039456 DOI: 10.1109/embc53108.2024.10782423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
African American women face a disproportionately big challenge from Triple Negative Breast Cancer (TNBC), the deadliest form of breast cancer, marking a critical issue in the fields of women's health and oncology in the United States. This group experiences a 40% higher mortality rate compared to women of other ethnicities, highlighting a significant disparity, and underscoring the urgent need for the development of targeted, effective treatments, specifically tailored to address these demographic women. Towards this, the efficacy of high intensity, short duration electrical pulses combined with Resveratrol (Resv), a natural polyphenol, as a novel therapeutic agent against TNBC is investigated. Electrical pulses facilitate enhanced drug transport through the opening of pores in cell membrane for those molecules that are normally non-permeable. This approach addresses the challenge of Resv's limited bioavailability and enhances its apoptotic potential. Employing MDA-MB-468 cell line, derived from an African American woman, this study investigates the effects of Resv and electrical pulses (EP) on TNBC cells. The TNBC cells were treated with Resv and subjected to eight pulses of multiple electric fields ranging from 500V/cm to 1200V/cm at 100μs, and at 1Hz, followed by assessments monitoring viability, colony formation, and intracellular reactive oxygen species (ROS) levels. The results indicate an 84% reduction in live cancer cell viability and a 4-fold increase in ROS levels with the combined treatment, suggesting a promising approach to alternative TNBC therapy. This research evaluates a natural bio compound, along with an advanced drug delivery system, targeting a reduction in mortality rates and improved quality of life for TNBC patients.
Collapse
|
3
|
Lei Y, Li X, Zhu L. Matrine regulates miR-495-3p/miR-543/PDK1 axis to repress the progression of acute myeloid leukemia via the Wnt/β-catenin pathway. Chem Biol Drug Des 2024; 103:e14441. [PMID: 38230785 DOI: 10.1111/cbdd.14441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/22/2023] [Accepted: 12/20/2023] [Indexed: 01/18/2024]
Abstract
Acute myeloid leukemia (AML) is a commonly hematological malignancy with feature of rapidly increased immature myeloid cells in bone marrow. The anti-tumor activity of matrine has been reported in various cancers. However, the functional role of matrine in AML progression still needs to be studied. Cell growth, apoptosis and cell cycle arrest in AML cells were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, 5-ethynyl-2'-deoxyuridine (EdU) assay and flow cytometry, respectively. The levels of adenosine triphosphate (ATP)/adenosine diphosphate (ADP) ratio, lactate production and glucose consumption were detected to evaluate glycolysis. Dual-luciferase reporter assay was conducted to determine the relationships between phosphoinositide-dependent kinase 1 (PDK1) and microRNA-495-3p (miR-495-3p)/microRNA-543 (miR-543) in AML cells. The results showed that matrine inhibited cell proliferation, glycolysis, and accelerated cell apoptosis and cell cycle arrest in AML cells. MiR-495-3p/miR-543 was lowly expressed, and PDK1 was highly expressed in AML. Functionally, both miR-495-3p and miR-543 could reverse the effects of matrine on cell proliferation, glycolysis, apoptosis and cell cycle arrest in AML cells. Mechanistically, miR-495-3p/miR-543 directly targeted PDK1, and the inhibition impacts of miR-495-3p/miR-543 on AML progression could be rescued by PDK1 overexpression. Moreover, matrine also could regulate PDK1 expression to suppress AML progression. Besides, matrine modulated miR-495-3p/miR-543/PDK1 axis to inhibit the Wnt/β-catenin pathway. In summary, matrine hampered the progression of AML through targeting miR-495-3p and miR-543 to attenuate PDK1 expression, thereby repressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yanping Lei
- Department of Pharmacy, Weinan Maternal and Child Health Hospital (Weinan People's Hospital), Weinan, China
| | - Xiao Li
- Department of Gynecology, The Second Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - Liping Zhu
- Department of Hematology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Mohan Viswanathan T, Krishnakumar V, Senthilkumar D, Chitradevi K, Vijayabhaskar R, Rajesh Kannan V, Senthil Kumar N, Sundar K, Kunjiappan S, Babkiewicz E, Maszczyk P, Kathiresan T. Combinatorial Delivery of Gallium (III) Nitrate and Curcumin Complex-Loaded Hollow Mesoporous Silica Nanoparticles for Breast Cancer Treatment. NANOMATERIALS 2022; 12:nano12091472. [PMID: 35564180 PMCID: PMC9105406 DOI: 10.3390/nano12091472] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022]
Abstract
The main aims in the development of a novel drug delivery vehicle is to efficiently carry therapeutic drugs in the body's circulatory system and successfully deliver them to the targeted site as needed to safely achieve the desired therapeutic effect. In the present study, a passive targeted functionalised nanocarrier was fabricated or wrapped the hollow mesoporous silica nanoparticles with 3-aminopropyl triethoxysilane (APTES) to prepare APTES-coated hollow mesoporous silica nanoparticles (HMSNAP). A nitrogen sorption analysis confirmed that the shape of hysteresis loops is altered, and subsequently the pore volume and pore diameters of GaC-HMSNAP was reduced by around 56 and 37%, respectively, when compared with HMSNAP. The physico-chemical characterisation studies of fabricated HMSNAP, Ga-HMSNAP and GaC-HMSNAP have confirmed their stability. The drug release capacity of the fabricated Ga-HMSNAP and GaC-HMSNAP for delivery of gallium and curcumin was evaluated in the phosphate buffered saline (pH 3.0, 6.0 and 7.4). In an in silico molecular docking study of the gallium-curcumin complex in PDI, calnexin, HSP60, PDK, caspase 9, Akt1 and PTEN were found to be strong binding. In vitro antitumor activity of both Ga-HMSNAP and GaC-HMSNAP treated MCF-7 cells was investigated in a dose and time-dependent manner. The IC50 values of GaC-HMSNAP (25 µM) were significantly reduced when compared with free gallium concentration (40 µM). The mechanism of gallium-mediated apoptosis was analyzed through western blotting and GaC-HMSNAP has increased caspases 9, 6, cleaved caspase 6, PARP, and GSK 3β(S9) in MCF-7 cells. Similarly, GaC-HMSNAP is reduced mitochondrial proteins such as prohibitin1, HSP60, and SOD1. The phosphorylation of oncogenic proteins such as Akt (S473), c-Raf (S249) PDK1 (S241) and induced cell death in MCF-7 cells. Furthermore, the findings revealed that Ga-HMSNAP and GaC-HMSNAP provide a controlled release of loaded gallium, curcumin and their complex. Altogether, our results depicted that GaC-HMNSAP induced cell death through the mitochondrial intrinsic cell death pathway, which could lead to novel therapeutic strategies for breast adenocarcinoma therapy.
Collapse
Affiliation(s)
- Thimma Mohan Viswanathan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India; (T.M.V.); (D.S.); (K.C.); (K.S.); (S.K.)
| | - Vaithilingam Krishnakumar
- Department of Microbiology, Bharathidasan University, Tiruchirappalli 620024, India; (V.K.); (V.R.K.)
| | - Dharmaraj Senthilkumar
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India; (T.M.V.); (D.S.); (K.C.); (K.S.); (S.K.)
| | - Kaniraja Chitradevi
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India; (T.M.V.); (D.S.); (K.C.); (K.S.); (S.K.)
| | | | - Velu Rajesh Kannan
- Department of Microbiology, Bharathidasan University, Tiruchirappalli 620024, India; (V.K.); (V.R.K.)
| | | | - Krishnan Sundar
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India; (T.M.V.); (D.S.); (K.C.); (K.S.); (S.K.)
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India; (T.M.V.); (D.S.); (K.C.); (K.S.); (S.K.)
| | - Ewa Babkiewicz
- Department of Hydrobiology, Faculty of Biology, University of Warsaw, 02-089 Warsaw, Poland; (E.B.); (P.M.)
| | - Piotr Maszczyk
- Department of Hydrobiology, Faculty of Biology, University of Warsaw, 02-089 Warsaw, Poland; (E.B.); (P.M.)
| | - Thandavarayan Kathiresan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India; (T.M.V.); (D.S.); (K.C.); (K.S.); (S.K.)
- Correspondence: ; Tel.: +91-4563-289042; Fax: +91-4563-289322
| |
Collapse
|
5
|
The Landscape of PDK1 in Breast Cancer. Cancers (Basel) 2022; 14:cancers14030811. [PMID: 35159078 PMCID: PMC8834120 DOI: 10.3390/cancers14030811] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Given that 3-phosphoinositide-dependent kinase 1 (PDK1) plays a crucial role in the malignant biological behaviors of a wide range of cancers, we review the influence of PDK1 in breast cancer (BC). First, we describe the power of PDK1 in cellular behaviors and characterize the interaction networks of PDK1. Then, we establish the roles of PDK1 in carcinogenesis, growth and survival, metastasis, and chemoresistance in BC cells. More importantly, we sort the current preclinical or clinical trials of PDK1-targeted therapy in BC and find that, even though no selective PDK1 inhibitor is currently available for BC therapy, the combination trials of PDK1-targeted therapy and other agents have provided some benefit. Thus, there is increasing anticipation that PDK1-targeted therapy will have its space in future therapeutic approaches related to BC, and we hope the novel approaches of targeted therapy will be conducive to ameliorating the dismal prognosis of BC patients.
Collapse
|
6
|
Lai YJ, Ko MC, Chan SY, Chou YS, Wang CC, Ku PW, Chen LJ, Hsu LF, Chuang PH, Chen CC, Yen YF. Hospice Care Services Associated With a Lower Utilization of Life-Sustaining Treatments During End-Of-Life Care Among People Living With HIV/AIDS: A Population-Based Cohort Study. Am J Hosp Palliat Care 2022; 39:1165-1173. [PMID: 35044895 DOI: 10.1177/10499091211072240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Hospice care involves improving quality of end-of-life (EOL) care and respecting patients' preferences regarding EOL treatment. However, the impact of hospice care services on the utilization of life-sustaining treatments during EOL care in patients with life-limiting diseases has not been extensively studied. Objectives: This nationwide cohort study aimed to determine the impact of hospice care services on the utilization of life-sustaining treatments during the last 3 months of life among people living with HIV/AIDS (PLWHA) in Taiwan. Methods: From 2000 to 2018, we identified adult PLWHA from Taiwan centers for disease control HIV Surveillance System. HIV-infected individuals were defined as positive HIV-1 Western blot. Life-sustaining treatments included cardiopulmonary resuscitation, intubation, mechanical ventilation support, and defibrillation. The association of hospice care services with the utilization of life-sustaining treatments was determined using multiple logistic regression. Results: Of 5691 PLWHA, 2595 (45.9%) subjects utilized life-sustaining treatments during the last 3 months of life. After adjusting for other covariates, PLWHA with hospice care services were less likely to receive life-sustaining treatments during the last 3 months of life than those without the services (adjusted odds ratio [AOR] = .50, 95% confidence interval [CI]: .37-.66). Considering the type of life-sustaining treatments, hospice care services were associated with lower likelihood of receiving cardiopulmonary resuscitation (AOR = .22, 95% CI: .13-.39), endotracheal intubation (AOR = .48, 95% CI: .35-.65), and mechanical ventilation support (AOR = .56, 95% CI: .42-.75). Conclusion: Hospice care services were associated with a lower utilization of life-sustaining treatments during the last 3 months of life among PLWHA.
Collapse
Affiliation(s)
- Yun-Ju Lai
- Department of Health Care Management, 38028National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.,School of Medicine, 34914National Yang Ming Chiao Tung University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, 63501Puli Branch of Taichung Veterans General Hospital, Nantou, Taiwan.,Department of Exercise Health Science, 145475National Taiwan University of Sport, Taichung, Taiwan
| | - Ming-Chung Ko
- Department of Health Care Management, 38028National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.,Department of Urology, 38010Taipei City Hospital, Taipei, Taiwan
| | - Shang-Yih Chan
- Department of Health Care Management, 38028National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.,Department of Internal Medicine, 205692Taipei City Hospital Yangming Branch, Taipei, Taiwan.,63390University of Taipei, Taipei, Taiwan
| | - Yi-Sheng Chou
- School of Medicine, 34914National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Hematology and Oncology, 156947Taipei City Hospital Renai Branch, Taipei, Taiwan
| | - Chun-Chieh Wang
- Division of Chest Medicine, Department of Internal Medicine, 63501Puli Branch of Taichung Veterans General Hospital, Nantou, Taiwan.,Department of Eldercare, 63340Central Taiwan University of Science and Technology Taichung, Nantou, Taiwan
| | - Po-Wen Ku
- Graduate Institute of Sports and Health Management, 34916National Chung Hsing University, Taichung, Taiwan
| | - Li-Jung Chen
- Department of Exercise Health Science, 145475National Taiwan University of Sport, Taichung, Taiwan
| | - Li-Fei Hsu
- College of Public Health, 33561National Taiwan University, Taipei, Taiwan
| | - Pei-Hung Chuang
- Taipei Association of Health and Welfare Data Science, Taipei, Taiwan
| | - Chu-Chieh Chen
- Department of Health Care Management, 38028National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Yung-Feng Yen
- Department of Health Care Management, 38028National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.,63390University of Taipei, Taipei, Taiwan.,Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Section of Infectious Diseases, 205692Taipei City Hospital Yangming Branch, Taipei, Taiwan.,Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| |
Collapse
|
7
|
Therapeutic potential of the PI3K inhibitor LY294002 and PARP inhibitor Talazoparib combination in BRCA-deficient triple negative breast cancer cells. Cell Signal 2021; 91:110229. [PMID: 34958867 DOI: 10.1016/j.cellsig.2021.110229] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors provide a promising therapeutic strategy for triple-negative breast cancers (TNBCs) with BRCA1/2 mutation. However, acquire resistance mechanisms and genetic alterations limit the clinical efficacy of PARP inhibitors. The aberrant activation of phosphatidylinositol 3-kinase (PI3K) is a significant problem for cancer development and thus the inhibition of PI3K by PI3K inhibitors is a novel targeted therapy in advanced breast cancer. Here, we, for the first time, investigated that the combined inhibition of PARP by Talazoparib (TAL) and PI3K by LY294002 synergistically inhibited proliferation of BRCA1 mutant HCC1937 TNBC cells through apoptosis, G0/G1 arrest, oxidative stress and increased DNA damage compared to drug alone. Additionally, TAL and LY294002 combination could be a promising strategy for overcoming TAL resistance. Co-treatment of TAL with LY294002 considerably suppressed the activation of PI3K, Akt1 and mTOR expression and phosphorylated protein levels in TNBC cells and caused changes in the multiple kinase phosphorylation. Our findings revealed that the dual inhibition of PARP and PI3K might represent an effective therapeutic strategy for TNBC and potentially overcome TAL resistance.
Collapse
|
8
|
Li J, Wen S, Li B, Li N, Zhan X. Phosphorylation-Mediated Molecular Pathway Changes in Human Pituitary Neuroendocrine Tumors Identified by Quantitative Phosphoproteomics. Cells 2021; 10:cells10092225. [PMID: 34571875 PMCID: PMC8471408 DOI: 10.3390/cells10092225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
To investigate the biological role of protein phosphorylation in human nonfunctional pituitary neuroendocrine tumors (NF-PitNETs), proteins extracted from NF-PitNET and control tissues were analyzed with tandem mass tag (TMT)-based quantitative proteomics coupled with TiO2 enrichment of phosphopeptides. A total of 595 differentially phosphorylated proteins (DPPs) with 1412 phosphosites were identified in NF-PitNETs compared to controls (p < 0.05). KEGG pathway network analysis of 595 DPPs identified nine statistically significant signaling pathways, including the spliceosome pathway, the RNA transport pathway, proteoglycans in cancer, SNARE interactions in vesicular transport, platelet activation, bacterial invasion of epithelial cells, tight junctions, vascular smooth muscle contraction, and protein processing in the endoplasmic reticulum. GO analysis revealed that these DPPs were involved in multiple cellular components (CCs), biological processes (BPs), and molecule functions (MFs). The kinase analysis of 595 DPPs identified seven kinases, including GRP78, WSTF, PKN2, PRP4, LOK, NEK1, and AMPKA1, and the substrate of these kinases could provide new ideas for seeking drug targets for NF-PitNETs. The randomly selected DPP calnexin was further confirmed with immunoprecipitation (IP) and Western blot (WB). These findings provide the first DPP profiling, phosphorylation-mediated molecular network alterations, and the key kinase profiling in NF-PitNET pathogenesis, which are a precious resource for understanding the biological roles of protein phosphorylation in NF-PitNET pathogenesis and discovering effective phosphoprotein biomarkers and therapeutic targets and drugs for the management of NF-PitNETs.
Collapse
Affiliation(s)
- Jiajia Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Central South University, 87 Xiangya Road, Changsha 410008, China; (J.L.); (S.W.); (B.L.)
- Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China;
| | - Siqi Wen
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Central South University, 87 Xiangya Road, Changsha 410008, China; (J.L.); (S.W.); (B.L.)
- Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China;
| | - Biao Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Central South University, 87 Xiangya Road, Changsha 410008, China; (J.L.); (S.W.); (B.L.)
- Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China;
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China;
- Shandong Key Laboratory of Radiation Oncology, Shandong First Medical University, 440 Jiyan Road, Jinan 250117, China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan 250117, China;
- Shandong Key Laboratory of Radiation Oncology, Shandong First Medical University, 440 Jiyan Road, Jinan 250117, China
- Correspondence: or
| |
Collapse
|
9
|
Cao W, Zeng Z, Pan R, Wu H, Zhang X, Chen H, Nie Y, Yu Z, Lei S. Hypoxia-Related Gene FUT11 Promotes Pancreatic Cancer Progression by Maintaining the Stability of PDK1. Front Oncol 2021; 11:675991. [PMID: 34221996 PMCID: PMC8247946 DOI: 10.3389/fonc.2021.675991] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Background Hypoxia is associated with the development of pancreatic cancer (PC). However, genes associated with hypoxia response and their regulatory mechanism in PC cells were unclear. The current study aims to investigate the role of the hypoxia associated gene fucosyltransferase 11 (FUT11) in the progression of PC. Methods In the preliminary study, bioinformatics analysis predicted FUT11 as a key hypoxia associated gene in PC. The expression of FUT11 in PC was evaluated using quantitative real-time PCR (qRT-PCR), Western blot and immunohistochemistry. The effects of FUT11 on PC cells proliferation and migration under normoxia and hypoxia were evaluated using Cell Counting Kit 8, 5-ethynyl-2'-deoxyuridine (EDU) assay, colony formation assay and transwell assay. The effects of FUT11 in vivo was examined in mouse tumor models of liver metastasis and subcutaneous xenograft. Furthermore, Western blot, luciferase assay and immunoprecipitation were performed to explore the regulatory relationship among FUT11, hypoxia-inducible factor 1α (HIF1α) and pyruvate dehydrogenase kinase 1 (PDK1) in PC. Results FUT11 was markedly increased of PC cells with hypoxia, upregulated in the PC clinical tissues, and predicted a poor outcome of PC patients. Inhibition of FUT11 reduced PC cell growth and migratory ability of PC cells under normoxia and hypoxia conditions in vitro, and growth and tumor cell metastasis in vivo. FUT11 bound to PDK1 and regulated the expression PDK1 under normoxia and hypoxia. FUT11 interacted with PDK1 and decreased the ubiquitination of PDK1, lead to the activation of AKT/mTOR signaling pathway. FUT11 knockdown significantly increased the degradation of PDK1 under hypoxia, while treatment with MG132 can relieve the degradation of PDK1 induced by FUT11 knockdown. Overexpression of PDK1 in PC cells under hypoxia conditions reversed the suppressive impacts of FUT11 knockdown on PC cell growth and migration. In addition, HIF1α bound to the promoter of FUT11 and increased its expression, as well as co-expressed with FUT11 in PC tissues. Furthermore, overexpression of FUT11 partially rescued the suppressive effects of HIF1α knockdown on PC cell growth and migration in hypoxia condition. Conclusion Our data implicate that hypoxia-induced FUT11 contributes to proliferation and metastasis of PC by maintaining the stability of PDK1, thus mediating activation of AKT/mTOR signaling pathway, and suggest that FUT11 could be a novel and effective target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Wenpeng Cao
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Zhirui Zeng
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Runsang Pan
- Department of Orthopedics, Guiyang Maternal and Child Health Care Hospital, Guiyang, China
| | - Hao Wu
- Department of Pediatric Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiangyan Zhang
- NHC Key Laboratory of Pulmonary, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hui Chen
- NHC Key Laboratory of Pulmonary, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yingjie Nie
- The Clinical Lab Center, Guizhou Provincial People's Hospital, Guiyang, China
| | - Zijiang Yu
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Shan Lei
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| |
Collapse
|
10
|
Pyruvate dehydrogenase kinases (PDKs): an overview toward clinical applications. Biosci Rep 2021; 41:228121. [PMID: 33739396 PMCID: PMC8026821 DOI: 10.1042/bsr20204402] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/01/2023] Open
Abstract
Pyruvate dehydrogenase kinase (PDK) can regulate the catalytic activity of pyruvate decarboxylation oxidation via the mitochondrial pyruvate dehydrogenase complex, and it further links glycolysis with the tricarboxylic acid cycle and ATP generation. This review seeks to elucidate the regulation of PDK activity in different species, mainly mammals, and the role of PDK inhibitors in preventing increased blood glucose, reducing injury caused by myocardial ischemia, and inducing apoptosis of tumor cells. Regulations of PDKs expression or activity represent a very promising approach for treatment of metabolic diseases including diabetes, heart failure, and cancer. The future research and development could be more focused on the biochemical understanding of the diseases, which would help understand the cellular energy metabolism and its regulation by pharmacological effectors of PDKs.
Collapse
|
11
|
Kiss E, Forika G, Mohacsi R, Nemeth Z, Krenacs T, Dank M. Methyl-Donors Can Induce Apoptosis and Attenuate Both the Akt and the Erk1/2 Mediated Proliferation Pathways in Breast and Lung Cancer Cell Lines. Int J Mol Sci 2021; 22:ijms22073598. [PMID: 33808426 PMCID: PMC8036837 DOI: 10.3390/ijms22073598] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 01/04/2023] Open
Abstract
Dietary methyl-donors play important roles in physiological processes catalyzed by B vitamins as coenzymes, and are used for complementary support in oncotherapy. Our hypothesis was that methyl-donors can not only assist in tolerating cancer treatment but may also directly interfere with tumor growth and proliferation. Therefore, we investigated the proposed cancer inhibitory effects of methyl-donors (in a mixture of L-methionine, choline chloride, folic acid, and vitamin B12) on MCF7 and T47D breast cancer as well as A549 and H1650 lung cancer cell lines. Indeed, methyl-donor treatment significantly reduced the proliferation in all cell lines, possibly through the downregulation of MAPK/ERK and AKT signaling. These were accompanied by the upregulation of the pro-apoptotic Bak and Bax, both in MCF7 and H1650 cells, at reduced anti-apoptotic Mcl-1 and Bcl-2 levels in MCF7 and H1650 cells, respectively. The treatment-induced downregulation of p-p53(Thr55) was likely to contribute to protecting the nuclear localization and apoptosis inducing functions of p53. The presented features are known to improve the sensitivity of cancer therapy. Therefore, these data support the hypothesis, i.e., that methyl-donors may promote apoptotic signaling by protecting p53 functions through downregulating both the MAPK/ERK and the AKT pathways both in breast and lung adenocarcinoma cell lines. Our results can emphasize the importance and benefits of the appropriate dietary supports in cancer treatments. However, further studies are required to confirm these effects without any adverse outcome in clinical settings.
Collapse
Affiliation(s)
- Eva Kiss
- 1st Department of Internal Medicine and Oncology, Oncology Profile, Semmelweis University, 1085 Budapest, Hungary; (E.K.); (R.M.); (M.D.)
| | - Gertrud Forika
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (G.F.); (T.K.)
| | - Reka Mohacsi
- 1st Department of Internal Medicine and Oncology, Oncology Profile, Semmelweis University, 1085 Budapest, Hungary; (E.K.); (R.M.); (M.D.)
| | - Zsuzsanna Nemeth
- 1st Department of Internal Medicine and Oncology, Oncology Profile, Semmelweis University, 1085 Budapest, Hungary; (E.K.); (R.M.); (M.D.)
- Correspondence: ; Tel.: +36-20-670-1025
| | - Tibor Krenacs
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (G.F.); (T.K.)
| | - Magdolna Dank
- 1st Department of Internal Medicine and Oncology, Oncology Profile, Semmelweis University, 1085 Budapest, Hungary; (E.K.); (R.M.); (M.D.)
| |
Collapse
|
12
|
Gilreath C, Boerma M, Qin Z, Hudson MK, Wang S. The Hypoxic Microenvironment of Breast Cancer Cells Promotes Resistance in Radiation Therapy. Front Oncol 2021; 10:629422. [PMID: 33680952 PMCID: PMC7930560 DOI: 10.3389/fonc.2020.629422] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 12/29/2020] [Indexed: 11/13/2022] Open
Abstract
The American Cancer Society has estimated an expected 279,100 new breast cancer cases, and an expected 42,690 breast cancer deaths in the U.S. for the year 2020. This includes an estimated 276,480 women who are expected to be diagnosed. Radiation therapy, also called ionizing radiation therapy, is one of the most frequently used methods in the treatment of breast cancer. While radiation therapy is used in the treatment of more than 50% of all cancer cases, tumor resistance to ionizing radiation presents a major challenge for effective cancer treatment. Most tumor cells are in a hypoxic microenvironment that promotes resistance to radiation therapy. In addition to radiation resistance, the hypoxic microenvironment also promotes cancer proliferation and metastasis. In this review, we will discuss the hypoxic microenvironment of breast cancer tumors, related signaling pathways, breast cancer stem-like cells, and the resistance to radiation therapy. Recent developments in our understanding of tumor hypoxia and hypoxic pathways may assist us in developing new strategies to increase cancer control in radiation therapy.
Collapse
Affiliation(s)
- Cordell Gilreath
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - Marjan Boerma
- Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Zhiqiang Qin
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - M Keith Hudson
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, United States
| | - Shanzhi Wang
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, United States
| |
Collapse
|
13
|
Yen YF, Lai YJ, Ko MC, Chen YY, Chen YT, Chou YS, Huang CT, Chen CC. Association of Hospice Care Services With the Utilization of Life-Sustaining Treatments During End-of-Life Care Among Patients With Cancer: A Nationwide 11-Year Cohort Study. J Pain Symptom Manage 2021; 61:323-330. [PMID: 32739560 DOI: 10.1016/j.jpainsymman.2020.07.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/18/2020] [Accepted: 07/23/2020] [Indexed: 12/16/2022]
Abstract
CONTEXT The impact of hospice care services on the utilization of life-sustaining treatments during end-of-life care in terminally ill patients has not been extensively studied. OBJECTIVES To determine the impact of hospice care services on the utilization of life-sustaining treatments during the last three months of life among patients with cancer. METHODS This nationwide population-based cohort study identified adults with cancer diagnosis from the Taiwan Registry for Catastrophic Illness, 2006-2016. Life-sustaining treatments included cardiopulmonary resuscitation, intubation, mechanical ventilation support, nasogastric tube feeding, and total parenteral nutrition. Hospice care services consisted of hospice inpatient care, hospice-shared care, and hospice home care. The association of hospice care services with the utilization of life-sustaining treatments was determined using multiple logistic regression. RESULTS Of 516,409 patients with cancer, 310,722 (60.2%) patients used life-sustaining treatments during the last three months of life. After adjusting for covariates, patients with hospice care services were less likely to receive life-sustaining treatments during the last three months of life than those without the services (adjusted odds ratio [AOR]: 0.70; 95% CI: 0.69-0.71). While type of life-sustaining treatments were considered, hospice care services were associated with a lower likelihood of receiving cardiopulmonary resuscitation (AOR: 0.125; 95% CI: 0.118-0.131), endotracheal intubation (AOR: 0.204; 95% CI: 0.199-0.210), mechanical ventilation support (AOR: 0.265; 95% CI: 0.260-0.270), nasogastric tube feeding (AOR: 0.736; 95% CI: 0.727-0.744), and total parenteral nutrition (AOR: 0.86; 95% CI: 0.84-0.88). CONCLUSION Hospice care services were associated with a lower likelihood of receiving life-sustaining treatments during the last three months of life in patients with cancer.
Collapse
Affiliation(s)
- Yung-Feng Yen
- Section of Infectious Diseases, Taipei City Hospital, Taipei, Taiwan; Institute of Public Health, National Yang-Ming University, Taipei, Taiwan; Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan; Department of Education and Research, Taipei City Hospital, Taiwan.
| | - Yun-Ju Lai
- School of Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Endocrinology and Metabolism, Department of Internal Medicine, Puli Branch of Taichung Veterans General Hospital, Nantou, Taiwan; Department of Exercise Health Science, National Taiwan University of Sport, Taichung, Taiwan
| | - Ming-Chung Ko
- Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan; Department of Urology, Taipei City Hospital, Taipei, Taiwan
| | - Yu-Yen Chen
- Institute of Public Health, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Ophthalmology, Taichung Veterans General Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Tui Chen
- Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Yi-Sheng Chou
- Department of Hematology and Oncology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
| | - Chun-Teng Huang
- Department of Hematology and Oncology, Taipei City Hospital, Yang-Ming Branch, Taipei, Taiwan
| | - Chu-Chieh Chen
- Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| |
Collapse
|
14
|
Huang G, Song C, Wang N, Qin T, Sui S, Obr A, Zeng L, Wood TL, Leroith D, Li M, Wu Y. RNA-binding protein CUGBP1 controls the differential INSR splicing in molecular subtypes of breast cancer cells and affects cell aggressiveness. Carcinogenesis 2020; 41:1294-1305. [PMID: 31958132 PMCID: PMC7513956 DOI: 10.1093/carcin/bgz141] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/18/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
The insulin receptor gene (INSR) undergoes alternative splicing to give rise to two functionally related, but also distinct, isoforms IR-A and IR-B, which dictate proliferative and metabolic regulations, respectively. Previous studies identified the RNA-binding protein CUGBP1 as a key regulator of INSR splicing. In this study, we show that the differential splicing of INSR occurs more frequently in breast cancer than in non-tumor breast tissues. In breast cancer cell lines, the IR-A:IR-B ratio varies in different molecular subtypes, knockdown or overexpression of CUGBP1 gene in breast cancer cells altered IR-A:IR-B ratio through modulation of IR-A expression, thereby reversed or enhanced the insulin-induced oncogenic behavior of breast cancer cells, respectively. Our data revealed the predominant mitogenic role of IR-A isoform in breast cancer and depicted a novel interplay between INSR and CUGBP1, implicating CUGBP1 and IR-A isoform as the potential therapeutic targets and biomarkers for breast cancer.
Collapse
Affiliation(s)
- Gena Huang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Chen Song
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Qin
- Department of Pathology, Dalian Medical University, Dalian, Liaoning, China
| | - Silei Sui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Alison Obr
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Li Zeng
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Derek Leroith
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
| | - Man Li
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
- College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
15
|
Theivendran S, Tang J, Lei C, Yang Y, Song H, Gu Z, Wang Y, Yang Y, Jin L, Yu C. Post translational modification-assisted cancer immunotherapy for effective breast cancer treatment. Chem Sci 2020; 11:10421-10430. [PMID: 34123182 PMCID: PMC8162284 DOI: 10.1039/d0sc02803g] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 09/09/2020] [Indexed: 01/24/2023] Open
Abstract
Post translational modifications (PTM) such as phosphorylation are often correlated with tumorigenesis and malignancy in breast cancer. Herein, we report a PTM-assisted strategy as a simplified version of a personalized cancer vaccine for enhanced cancer immunotherapy. Titanium modified dendritic mesoporous silica nanoparticles (TiDMSN) are applied to assist the specific enrichment of phosphorylated tumor antigens released upon immunogenic cell death. This strategy significantly improved the tumor inhibition efficacy in a bilateral breast cancer model and the expansion of both CD8+ and CD4+ T cells in the distant tumor site. The nanotechnology based PTM-assisted strategy provides a simple and generalizable methodology for effective personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Shevanuja Theivendran
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
| | - Chang Lei
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
| | - Zhengying Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
- School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
| | - Yang Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
| | - Lei Jin
- School of Medicine and Public Health, University of Newcastle NSW 2308 Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland QLD 4072 Australia
- School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| |
Collapse
|
16
|
Wei Y, Han X, Zhao C. PDK1 regulates the survival of the developing cortical interneurons. Mol Brain 2020; 13:65. [PMID: 32366272 PMCID: PMC7197138 DOI: 10.1186/s13041-020-00604-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 04/22/2020] [Indexed: 01/08/2023] Open
Abstract
Inhibitory interneurons are critical for maintaining the excitatory/inhibitory balance. During the development cortical interneurons originate from the ganglionic eminence and arrive at the dorsal cortex through two tangential migration routes. However, the mechanisms underlying the development of cortical interneurons remain unclear. 3-Phosphoinositide-dependent protein kinase-1 (PDK1) has been shown to be involved in a variety of biological processes, including cell proliferation and migration, and plays an important role in the neurogenesis of cortical excitatory neurons. However, the function of PDK1 in interneurons is still unclear. Here, we reported that the disruption of Pdk1 in the subpallium achieved by crossing the Dlx5/6-Cre-IRES-EGFP line with Pdk1fl/fl mice led to the severely increased apoptosis of immature interneurons, subsequently resulting in a remarkable reduction in cortical interneurons. However, the tangential migration, progenitor pools and cell proliferation were not affected by the disruption of Pdk1. We further found the activity of AKT-GSK3β signaling pathway was decreased after Pdk1 deletion, suggesting it might be involved in the regulation of the survival of cortical interneurons. These results provide new insights into the function of PDK1 in the development of the telencephalon.
Collapse
Affiliation(s)
- Yongjie Wei
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiaoning Han
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
17
|
Zhao S, Li X, Yin L, Hou L, Lan J, Zhu X. TCRP1 induces tamoxifen resistance by promoting the activation of SGK1 in MCF‑7 cells. Oncol Rep 2020; 43:2017-2027. [PMID: 32323833 PMCID: PMC7160545 DOI: 10.3892/or.2020.7577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 03/04/2020] [Indexed: 02/07/2023] Open
Abstract
Tamoxifen is widely used as a highly effective drug for treating estrogen‑receptor (ER) alpha‑positive breast cancer. However, tamoxifen resistance developed during cancer treatment remains a significant challenge. Tongue cancer resistance‑related protein1 (TCRP1), which is recognized as a novel drug target, is related to chemo‑resistance in human cancers, moreover, it is often overexpressed in various cancer cells, such as in lung cancer, breast cancer, and tongue cancer. However, the effects of TCRP1 on tamoxifen‑resistant breast cancer cells and tissues are far from clear. The present study revealed that TCRP1 induced tamoxifen resistance in breast cancer cells. Western blotting, quantitative real‑time polymerase chain reaction (RT‑PCR) and immunohistochemical staining were performed to detect the expression level of TCRP1 in vivo and in vitro between primary breast cancer tissues and tamoxifen‑resistant breast cancer tissues. The data revealed that the expression of TCRP1 was upregulated in the tamoxifen‑resistant breast cancer tissues and human breast cancer cell line, MCF‑7. Further study revealed that knocking down TCRP1 inhibited the growth of MCF‑7 cells with tamoxifen‑resistance (MCF7‑R cells) and induced cell apoptosis. Moreover, TCRP1 promoted serum‑ and glucocorticoid‑inducible kinase 1 (SGK1) activation via phosphorylation of phosphoinositide‑dependent kinase 1 (PDK1) in MCF7‑R cells. In addition, it was also observed that knocking down TCRP1 inhibited tumorigenesis of MCF‑7 cells in nude mice. In conclusion, these data indicated that TCRP1 could induce tamoxifen resistance by regulating the PDK1/SGK1 signaling pathway. Thus, TCRP1 could be explored as a promising candidate for treating tamoxifen‑resistant breast cancer in the future.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Breast and Thyroid Surgery, Wuzhong People's Hospital of Suzhou City, Suzhou, Jiangsu 215128, P.R. China
| | - Xiaohua Li
- Department of Breast and Thyroid Surgery, Wuzhong People's Hospital of Suzhou City, Suzhou, Jiangsu 215128, P.R. China
| | - Lei Yin
- Department of Breast and Thyroid Surgery, Wuzhong People's Hospital of Suzhou City, Suzhou, Jiangsu 215128, P.R. China
| | - Lili Hou
- Department of Breast and Thyroid Surgery, Wuzhong People's Hospital of Suzhou City, Suzhou, Jiangsu 215128, P.R. China
| | - Jing Lan
- Department of General Surgery, The First Hospital Affiliated of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Xun Zhu
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
18
|
Zhao C, Thompson BJ, Chen K, Gao F, Blouw B, Marella M, Zimmerman S, Kimbler T, Garrovillo S, Bahn J, Huang L, Huang Z, Shepard HM, Rosengren S, Thanos CD, Maneval DC. The growth of a xenograft breast cancer tumor model with engineered hyaluronan-accumulating stroma is dependent on hyaluronan and independent of CD44. Oncotarget 2019; 10:6561-6576. [PMID: 31762938 PMCID: PMC6859925 DOI: 10.18632/oncotarget.27302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/19/2019] [Indexed: 12/18/2022] Open
Abstract
Hyaluronan accumulation in the tumor microenvironment is associated with poor prognosis in several solid human cancers. To understand the role of stromal hyaluronan in tumor progression, we engineered 3T3HAS3, a hyaluronan-producing fibroblast cell line, by lentiviral transduction of Balb/c 3T3 cells with the human hyaluronan synthase 3(HAS3) gene. 3T3HAS3 cells significantly enhanced tumor growth when co-grafted with MDA-MB-468 cells in nude mice. Immunohistochemical analysis of the xenograft tumors showed that MDA-MB-468 cells were surrounded by hyaluronan-accumulating stroma, closely resembling the morphology observed in human breast cancer specimens. Tumor growth of MDA-MB-468 + 3T3HAS3 co-grafts was greatly reduced upon hyaluronan degradation by lentiviral transduction of a human hyaluronidase gene in 3T3HAS3 cells, or by systemic administration of pegvorhyaluronidase alfa (PEGPH20). In contrast, the growth of the co-graft tumors was not inhibited when CD44 expression was reduced or ablated by small hairpin RNA-mediated CD44 knockdown in MDA-MB-468 cells, CD44 CRISPR knockout in 3T3HAS3 cells, or by grafting these cells in CD44 knockout nude mice. Collectively, these data demonstrate that tumor growth of an engineered xenograft breast cancer model with hyaluronan-accumulating stroma can be dependent on hyaluronan and independent of CD44.
Collapse
Affiliation(s)
- Chunmei Zhao
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Benjamin J Thompson
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Kelly Chen
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Feng Gao
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Barbara Blouw
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Mathieu Marella
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | | | - Trevor Kimbler
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Sheryl Garrovillo
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Jesse Bahn
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Lei Huang
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Zhongdong Huang
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - H Michael Shepard
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Sanna Rosengren
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | - Christopher D Thanos
- Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA.,Formerly of Halozyme Therapeutics, Inc., San Diego, CA, 92121, USA
| | | |
Collapse
|
19
|
Niu Y, Xu J, Sun T. Cyclin-Dependent Kinases 4/6 Inhibitors in Breast Cancer: Current Status, Resistance, and Combination Strategies. J Cancer 2019; 10:5504-5517. [PMID: 31632494 PMCID: PMC6775706 DOI: 10.7150/jca.32628] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 08/06/2019] [Indexed: 12/26/2022] Open
Abstract
Dysregulated activation of the cyclin-dependent kinases (CDKs) 4/6, leading to uncontrolled cell division, is hallmark of cancers. Further study of the cell cycle will advance the cancer treatment. As powerful and effective drugs, inhibitors of CDK 4/6 have been widely used in clinical practice for several malignancies, particularly against breast cancers driven by the estrogen receptor (ER). Three CDK4/6 inhibitors, including palbociclib (PD0332991), ribociclib (LEE011) and abemaciclib (LY2835219), have been approved by the US Food and Drug Administration (FDA) for the treatment of hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced or metastatic breast cancer. However, CDK4/6 inhibitors act downstream of many mitogenic signaling pathways, and this has implications for resistance. It is worth to note that the mechanisms of resistance are not very clear. Up to now, a small number of preclinical and clinical studies have explored potential mechanisms of CDK4/6 inhibitors resistance in breast cancer. On this basis, rational and effective combination therapy is under development. Here we review the current knowledge about the mechanisms and efficacy of CDK4/6 inhibitors, and summarize data on resistance mechanisms to make future combination therapies more accurate and reasonable.
Collapse
Affiliation(s)
- Ying Niu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China
| | - Junnan Xu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China.,Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning 110042, P.R. China
| | - Tao Sun
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
20
|
Siriwardhana C, Khadka VS, Chen JJ, Deng Y. Development of a miRNA-seq based prognostic signature in lung adenocarcinoma. BMC Cancer 2019; 19:34. [PMID: 30621620 PMCID: PMC6325795 DOI: 10.1186/s12885-018-5206-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/10/2018] [Indexed: 12/29/2022] Open
Abstract
Background We utilized miRNAs expression and clinical data to develop a prognostic signature for patients with lung adenocarcinoma, with respect to their overall survival, to identify high-risk subjects based on their miRNA genomic profile. Methods MiRNA expressions based on miRNA sequencing and clinical data of lung adenocarcinoma patients (n = 479) from the Cancer Genome Atlas were randomly partitioned into non-overlapping Model (n = 320) and Test (n = 159) sets, respectively, for model estimation and validation. Results Among the ten miRNAs identified using the univariate Cox analysis, six from miR-8, miR-181, miR-326, miR-375, miR-99a, and miR-10, families showed improvement of the overall survival chance, while two miRNAs from miR-582 and miR-584 families showed a worsening of survival chances. The final prognostic signature was developed with five miRNAs—miR-375, miR-582-3p, miR-326, miR-181c-5p, and miR-99a-5p—utilizing a stepwise variable selection procedure. Using the KEGG pathway analysis, we found potential evidence supporting their significance in multiple cancer pathways, including non-small cell lung cancer. We defined two risk groups with a score calculated using the Cox regression coefficients. The five-year survival rates for the low-risk group was approximately 48.76% (95% CI = (36.15, 63.93)); however, it was as low as 7.50% (95% CI = (2.34, 24.01)) for the high-risk group. Furthermore, we demonstrated the effect of the genomic profile using the miRNA signature, quantifying survival rates for hypothetical subjects in different pathological stages of cancer. Conclusions The proposed prognostic signature can be used as a reliable tool for identifying high-risk subjects regarding survival based on their miRNA genomic profile. Electronic supplementary material The online version of this article (10.1186/s12885-018-5206-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chathura Siriwardhana
- Bioinformatics and Biostatistics Cores, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA.
| | - Vedbar S Khadka
- Bioinformatics and Biostatistics Cores, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA
| | - John J Chen
- Bioinformatics and Biostatistics Cores, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA
| | - Youping Deng
- Bioinformatics and Biostatistics Cores, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA.
| |
Collapse
|
21
|
Gopalakrishnan K, Venkatesan S, Low ESH, Hande MP. Effects of rapamycin on the mechanistic target of rapamycin (mTOR) pathway and telomerase in breast cancer cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 836:103-113. [DOI: 10.1016/j.mrgentox.2018.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 03/09/2018] [Accepted: 03/27/2018] [Indexed: 01/24/2023]
|
22
|
Luo D, Xu X, Li J, Chen C, Chen W, Wang F, Xie Y, Li F. The PDK1/c‑Jun pathway activated by TGF‑β induces EMT and promotes proliferation and invasion in human glioblastoma. Int J Oncol 2018; 53:2067-2080. [PMID: 30106127 DOI: 10.3892/ijo.2018.4525] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 07/23/2018] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignant tumor affecting the human brain. Despite improvements in therapeutic technologies, patients with GBM have a poor clinical result and the molecular mechanisms responsible for the development of GBM have not yet been fully elucidated. 3-phosphoinositide dependent protein kinase 1 (PDK1) is upregulated in various tumors and promotes tumor invasion. In glioma, transforming growth factor-β (TGF‑β) promotes cell invasion; however, whether TGF‑β directly regulates PDK1 protein and promotes proliferation and invasion is not yet clear. In this study, PDK1 levels were measured in glioma tissues using tissue microarray (TMA) by immunohistochemistry (IHC) and RT‑qPCR. Kaplan-Meier analyses were used to calculate the survival rate of patients with glioma. In vitro, U251 and U87 glioma cell lines were used for functional analyses. Cell proliferation and invasion were analyzed using siRNA transfection, MTT assay, RT‑qPCR, western blot analysis, flow cytometry and invasion assay. In vivo, U251 glioma cell xenografts were established. The results revealed that PDK1 protein was significantly upregulated in glioma tissues compared with non-tumorous tissues. Furthermore, the higher PDK1 levels were associated with a large tumor size (>5.0 cm), a higher WHO grade and a shorter survival of patients with GBM. Univariate and multivariate analyses indicated that PDK1 was an independent prognostic factor. In vivo, PDK1 promoted glioma tumor xenograft growth. In vitro, functional analyses confirmed that TGF‑β upregulated PDK1 protein expression and PDK1 promoted cell migration and invasion, and functioned as an oncogene in GBM, by upregulating c‑Jun protein and inducing epithelial-mesenchymal transition (EMT). c‑Jun protein were overexpressed in glioma tissues and positively correlated with PDK1 levels. Moreover, our findings were further validated by the online Oncomine database. On the whole, the findings of this study indicate that in GBM, PDK1 functions as an oncogene, promoting proliferation and invasion.
Collapse
Affiliation(s)
- Dingyuan Luo
- Department of Vascular and Thyroid Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xinke Xu
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510120, P.R. China
| | - Junliang Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510120, P.R. China
| | - Cheng Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510120, P.R. China
| | - Wei Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510120, P.R. China
| | - Fangyu Wang
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510120, P.R. China
| | - Yanping Xie
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510120, P.R. China
| | - Fangcheng Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
23
|
Liu X, Montissol S, Uber A, Ganley S, Grossestreuer AV, Berg K, Heydrick S, Donnino MW. The Effects of Thiamine on Breast Cancer Cells. Molecules 2018; 23:molecules23061464. [PMID: 29914147 PMCID: PMC6100081 DOI: 10.3390/molecules23061464] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/06/2018] [Accepted: 06/09/2018] [Indexed: 12/31/2022] Open
Abstract
(1) Background: Thiamine is an important cofactor for multiple metabolic processes. Its role in cancer has been debated for years. Our aim is to determine if thiamine can convert the cellular metabolic state of breast cancer cells from anaerobic to aerobic, thus reducing their growth. (2) Methods: Breast cancer (MCF7) and non-tumorigenic (MCF10A) cell lines were treated with various doses of thiamine and assessed for changes in cell growth. The mechanism of this relationship was identified through the measurement of enzymatic activity and metabolic changes. (3) Results: A high dose of thiamine reduced cell proliferation in MCF7 (63% decrease, p < 0.0001), but didn’t affect apoptosis and the cell-cycle profile. Thiamine had a number of effects in MCF7; it (1) reduced extracellular lactate levels in growth media, (2) increased cellular pyruvate dehydrogenase (PDH) activities and the baseline and maximum cellular oxygen consumption rates, and (3) decreased non-glycolytic acidification, glycolysis, and glycolytic capacity. MCF10A cells preferred mitochondrial respiration instead of glycolysis. In contrast, MCF7 cells were more resistant to mitochondrial respiration, which may explain the inhibitory effect of thiamine on their proliferation. (4) Conclusions: The treatment of MCF7 breast cancer cells with 1 μg/mL and 2 μg/mL of thiamine for 24 h significantly reduced their proliferation. This reduction is associated with a reduction in glycolysis and activation of the PDH complex in breast cancer cells.
Collapse
Affiliation(s)
- Xiaowen Liu
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Sophia Montissol
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Amy Uber
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Sarah Ganley
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Anne V Grossestreuer
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Katherine Berg
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
- Department of Medicine, Division of Pulmonary and Critical Care, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Stanley Heydrick
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Michael W Donnino
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
- Department of Medicine, Division of Pulmonary and Critical Care, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Dunlop EA, Johnson CE, Wiltshire M, Errington RJ, Tee AR. Targeting protein homeostasis with nelfinavir/salinomycin dual therapy effectively induces death of mTORC1 hyperactive cells. Oncotarget 2018; 8:48711-48724. [PMID: 28415776 PMCID: PMC5564719 DOI: 10.18632/oncotarget.16232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/03/2017] [Indexed: 02/05/2023] Open
Abstract
Uncontrolled cell growth in Tuberous Sclerosis Complex occurs due to inappropriate activation of mechanistic (mammalian) target of rapamycin complex 1 (mTORC1). The current therapy, rapamycin, produced promising clinical trial results, but patient tumours regrow if treatment is discontinued, revealing rapamycin has cytostatic properties rather than a cytotoxic effect. Taking advantage of the enhanced levels of endoplasmic reticulum (ER) stress present in TSC2-null cells, we investigated drug combinations producing a cytotoxic response. We found a nelfinavir and salinomycin combination specifically killed TSC2-deficient, mTORC1 hyperactive cells. Cytotoxicity was rescued by reducing protein synthesis, either through mTORC1 inhibition or cycloheximide treatment. This indicates that the drug combination targets the cells by tipping the protein homeostasis balance of the already metabolically stressed TSC2-deficient cells in favour of cell death. Furthermore, this drug combination also inhibited tumour formation in TSC2-deficient cell models and caused tumour spheroid death in 3D culture. Importantly, the 3D assay could differentiate the cytostatic agent, rapamycin, from the cytotoxic nelfinavir/salinomycin combination. Sporadic cancer cell lines with hyperactive mTORC1 signalling were also susceptible to this nelfinavir/salinomycin drug combination. This work indicates that the protein homeostasis pathway is an attractive therapeutic target in both Tuberous Sclerosis Complex and mTORC1-driven sporadic cancers.
Collapse
Affiliation(s)
- Elaine A Dunlop
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Charlotte E Johnson
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Marie Wiltshire
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Rachel J Errington
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Andrew R Tee
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| |
Collapse
|
25
|
Ding Y, Liu W, Wang X, Zhang L, Zhao M, Deng H, Liu Y. Bufalin induces apoptosis in human esophageal carcinoma ECA109 cells by inhibiting the activation of the mTOR/p70S6K pathway. Oncol Lett 2018; 15:9339-9346. [PMID: 29805658 DOI: 10.3892/ol.2018.8526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 03/07/2018] [Indexed: 01/30/2023] Open
Abstract
The present study examined whether bufalin could induce human esophageal carcinoma ECA109 cells apoptosis via inhibiting the activation of mechanistic target of rapamycin (mTOR)/p70 S6 kinase (p70S6K) pathway is discussed in this article. The present study used the esophageal squamous cell carcinoma ECA109 cell line to assess the apoptosis-inducing effects of bufalin via inhibition of the mTOR/p70S6K pathways. A plasmid containing the wild-type mTOR gene (wtmTOR) was transfected into ECA109 cells. The levels of p70S6K, phosphorylated (p)-p70S6K, cellular inhibitor of apoptosis-1 (cIAP-1) and Bcl-2-associated death promoter (BAD) in ECA109 cells were examined by western blot analysis, and apoptosis was detected by flow cytometry analysis and Giemsa staining. The results revealed that the expression of p-p70S6K was increased as the time progressed (at 0, 12 and 24 h), and then decreased at 30, 36, 42 and 48 h after transfection. The expression of cIAP-1 was significantly decreased as time progressed following the addition of bufalin, whereas that of BAD was increased. The levels of p-p70S6K and cIAP-1 were significantly higher in the wtmTOR-transfected group than in the control and empty vector-transfected groups, and then reduced following addition of bufalin; however, BAD expression was significantly lower in the wtmTOR-transfected group. The results of flow cytometry revealed the cell cycle of ECA109 was arrested at G2/M phase and the apoptotic rate was significantly lower in the wtmTOR-transfected group than in the control and empty vector-transfected groups, and then increased following addition of bufalin. In conclusion, the findings of the present study demonstrated that bufalin induced apoptosis in esophageal carcinoma cells via the inhibition of the mTOR/p70S6K pathway and indicated that treatment with bufalin could be combined with chemotherapy to overcome the resistance of esophageal carcinoma cells to chemotherapeutic-induced apoptosis.
Collapse
Affiliation(s)
- Yan Ding
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Wei Liu
- Department of Surgery, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaoling Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Lingling Zhang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Meng Zhao
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Huiyan Deng
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
26
|
Kaemmerer E, Turner D, Peters AA, Roberts-Thomson SJ, Monteith GR. An automated epifluorescence microscopy imaging assay for the identification of phospho-AKT level modulators in breast cancer cells. J Pharmacol Toxicol Methods 2018; 92:13-19. [PMID: 29438745 DOI: 10.1016/j.vascn.2018.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/28/2018] [Accepted: 02/08/2018] [Indexed: 11/19/2022]
Abstract
AKT is an enzyme of the PI3K/pAKT pathway, regulating proliferation and cell survival. High basal levels of active, phosphorylated AKT (pAKT) are associated with tumor progression and therapeutic resistance in some breast cancer subtypes, including HER2 positive breast cancers. Various stimuli can increase pAKT levels and elevated basal pAKT levels are a feature of PTEN-deficient breast cancer cell lines. The aim of this study was to develop an assay able to identify modulators of pAKT levels using an automated epifluorescence microscope and high content analysis. To develop this assay, we used HCC-1569, a PTEN-deficient, HER2-overexpressing breast cancer cell line with elevated basal pAKT levels. HCC-1569 cells were treated with a selective pharmacological inhibitor of AKT (MK-2206) to reduce basal pAKT levels or EGF to increase pAKT levels. Immunofluorescence images were acquired using an automated epifluorescence microscope and integrated intensity of cytoplasmic pAKT staining was calculated using high content analysis software. Mean and median integrated cytoplasmic intensity were normalized using fold change and standard score to assess assay quality and to identify most robust data analysis. The highest z' factor was achieved for median data normalization using the standard score method (z' = 0.45). Using our developed assay we identified the calcium homeostasis regulating proteins TPRV6, STIM1 and TRPC1 as modulators of pAKT levels in HCC-1569 cells. Calcium signaling controls a diverse array of cellular processes and some calcium homeostasis regulating proteins are involved in modulating pAKT levels in cancer cells. Thus, these identified hits present promising targets for further assessment.
Collapse
Affiliation(s)
- Elke Kaemmerer
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia.
| | - Dane Turner
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia.
| | - Amelia A Peters
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia.
| | | | - Gregory R Monteith
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
27
|
Li Y, Liu C, Tang K, Chen Y, Tian K, Feng Z, Chen J. Novel multi‑kinase inhibitor, T03 inhibits Taxol‑resistant breast cancer. Mol Med Rep 2017; 17:2373-2383. [PMID: 29207185 PMCID: PMC5783483 DOI: 10.3892/mmr.2017.8179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 07/26/2017] [Indexed: 11/06/2022] Open
Abstract
Activation of kinase-associated signaling pathways is one of the leading causes of various malignant phenotypes in breast tumors. Strategies of drug discovery and development have investigated approaches to target the inhibition of protein kinase signaling. In the current study, the anti‑tumor activities of a novel multi‑kinase inhibitor, T03 were evaluated in breast cancer. T03 inhibited Taxol‑resistant breast cancer cell proliferation and induced cell cycle arrest and apoptosis in vitro and in vivo. The current results demonstrated that T03 downregulated c‑Raf, platelet‑derived growth factor receptor‑β and other kinases, thus inhibited Raf/mitogen‑activated protein kinase kinase/extracellular signal‑regulated kinase and Akt/mechanistic target of rapamycin survival pathways in MCF‑7 and MCF‑7/Taxol xenograft tumors. At a dose of 100 mg/kg, T03 inhibited tumor growth by 62.90 and 59.98% in tumor weight in MX‑1 and MX‑1/T xenograft models, respectively and by 62.60 and 60.22% in MCF‑7 and MCF‑7/T tumors, respectively. These data indicate that the novel multi‑kinase inhibitor, T03, may present as a potential compound to develop novel treatments against breast cancer and Taxol‑resistant breast tumors.
Collapse
Affiliation(s)
- Yan Li
- Department of Pharmacology, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Chunxia Liu
- Department of Pharmacology, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Ke Tang
- Department of Pharmacology, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Yan Chen
- Department of Pharmacology, State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Kang Tian
- Department of Synthetic Medicinal Chemistry, Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Zhiqiang Feng
- Department of Synthetic Medicinal Chemistry, Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Jindong Chen
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
28
|
Targeting PDK1 for Chemosensitization of Cancer Cells. Cancers (Basel) 2017; 9:cancers9100140. [PMID: 29064423 PMCID: PMC5664079 DOI: 10.3390/cancers9100140] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 01/01/2023] Open
Abstract
Despite the rapid development in the field of oncology, cancer remains the second cause of mortality worldwide, with the number of new cases expected to more than double in the coming years. Chemotherapy is widely used to decelerate or stop tumour development in combination with surgery or radiation therapy when appropriate, and in many cases this improves the symptomatology of the disease. Unfortunately though, chemotherapy is not applicable to all patients and even when it is, there are many cases where a successful initial treatment period is followed by chemotherapeutic drug resistance. This is caused by a number of reasons, ranging from the genetic background of the patient (innate resistance) to the formation of tumour-initiating cells (acquired resistance). In this review, we discuss the potential role of PDK1 in the development of chemoresistance in different types of malignancy, and the design and application of potent inhibitors which can promote chemosensitization.
Collapse
|
29
|
Ishihara S, Inman DR, Li WJ, Ponik SM, Keely PJ. Mechano-Signal Transduction in Mesenchymal Stem Cells Induces Prosaposin Secretion to Drive the Proliferation of Breast Cancer Cells. Cancer Res 2017; 77:6179-6189. [PMID: 28972074 DOI: 10.1158/0008-5472.can-17-0569] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/27/2017] [Accepted: 09/25/2017] [Indexed: 01/15/2023]
Abstract
In response to chemical stimuli from cancer cells, mesenchymal stem cells (MSC) can differentiate into cancer-associated fibroblasts (CAF) and promote tumor progression. How mechanical stimuli such as stiffness of the extracellular matrix (ECM) contribute to MSC phenotype in cancer remains poorly understood. Here, we show that ECM stiffness leads to mechano-signal transduction in MSC, which promotes mammary tumor growth in part through secretion of the signaling protein prosaposin. On a stiff matrix, MSC cultured with conditioned media from mammary cancer cells expressed increased levels of α-smooth muscle actin, a marker of CAF, compared with MSC cultured on a soft matrix. By contrast, MSC cultured on a stiff matrix secreted prosaposin that promoted proliferation and survival of mammary carcinoma cells but inhibited metastasis. Our findings suggest that in addition to chemical stimuli, increased stiffness of the ECM in the tumor microenvironment induces differentiation of MSC to CAF, triggering enhanced proliferation and survival of mammary cancer cells. Cancer Res; 77(22); 6179-89. ©2017 AACR.
Collapse
Affiliation(s)
- Seiichiro Ishihara
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin.
| | - David R Inman
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Wan-Ju Li
- Departments of Orthopedics and Rehabilitation, and Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
30
|
Zavialova MG, Zgoda VG, Nikolaev EN. [Analysis of contribution of protein phosphorylation in the development of the diseases]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2017; 63:101-114. [PMID: 28414281 DOI: 10.18097/pbmc20176302101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In recent decades, studies in the molecular origins of socially significant diseases have made a big step forward with the development and using of high-performance methods in genomics and proteomics. Numerous studies in the framework of the global program "Human Proteome" were aimed at the identification of all possible proteins in various cell cultures and tissues, including cancer. One of the objectives was to identify biomarkers - proteins with high specificity to certain pathologies. However, in many cases, it is shown that the development of the disease is not associated with the appearance of new proteins, but depends on the level of gene expression or forming of proteoforms - splice variants, single amino acid substitutions (SAP variants), and post-translational modifications (PTM) of proteins. PTM may play a key role in the development of pathology because they activate a variety of regulatory or structural proteins in the majority of cell physiological processes. Phosphorylation is among the most significant of these protein modifications.This review will describe methods for analysis of protein phosphorylation used in the studies of such diseases as cancer and neurodegenerative diseases, as well as examples of cases when the modified proteins are involved directly to their development, and screening such significant PTM is used for the diagnosis and choice of treatment.
Collapse
Affiliation(s)
| | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| | - E N Nikolaev
- Institute of Biomedical Chemistry, Moscow, Russia; Skolkovo Institute of Science and Technology (Skoltech), Moscow, Russia
| |
Collapse
|
31
|
Jansen VM, Bhola NE, Bauer JA, Formisano L, Lee KM, Hutchinson KE, Witkiewicz AK, Moore PD, Estrada MV, Sánchez V, Ericsson PG, Sanders ME, Pohlmann PR, Pishvaian MJ, Riddle DA, Dugger TC, Wei W, Knudsen ES, Arteaga CL. Kinome-Wide RNA Interference Screen Reveals a Role for PDK1 in Acquired Resistance to CDK4/6 Inhibition in ER-Positive Breast Cancer. Cancer Res 2017; 77:2488-2499. [PMID: 28249908 PMCID: PMC5421398 DOI: 10.1158/0008-5472.can-16-2653] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/23/2016] [Accepted: 02/22/2017] [Indexed: 12/13/2022]
Abstract
Acquired resistance to cyclin-dependent kinases 4 and 6 (CDK4/6) small-molecule inhibitors in breast cancer arises through mechanisms that are yet uncharacterized. In this study, we used a kinome-wide siRNA screen to identify kinases that, when downregulated, yield sensitivity to the CDK4/6 inhibitor ribociclib. In this manner, we identified 3-phosphoinositide-dependent protein kinase 1 (PDK1) as a key modifier of ribociclib sensitivity in estrogen receptor-positive MCF-7 breast cancer cells. Pharmacologic inhibition of PDK1 with GSK2334470 in combination with ribociclib or palbociclib, another CDK4/6 inhibitor, synergistically inhibited proliferation and increased apoptosis in a panel of ER-positive breast cancer cell lines. Ribociclib-resistant breast cancer cells selected by chronic drug exposure displayed a relative increase in the levels of PDK1 and activation of the AKT pathway. Analysis of these cells revealed that CDK4/6 inhibition failed to induce cell-cycle arrest or senescence. Mechanistic investigations showed that resistant cells coordinately upregulated expression of cyclins A, E, and D1, activated phospho-CDK2, and phospho-S477/T479 AKT. Treatment with GSK2334470 or the CDK2 inhibitor dinaciclib was sufficient to reverse these events and to restore the sensitivity of ribociclib-resistant cells to CDK4/6 inhibitors. Ribociclib, in combination with GSK2334470 or the PI3Kα inhibitor alpelisib, decreased xenograft tumor growth more potently than each drug alone. Taken together, our results highlight a role for the PI3K-PDK1 signaling pathway in mediating acquired resistance to CDK4/6 inhibitors. Cancer Res; 77(9); 2488-99. ©2017 AACR.
Collapse
Affiliation(s)
- Valerie M Jansen
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Neil E Bhola
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joshua A Bauer
- Department of Biochemistry, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Luigi Formisano
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kyung-Min Lee
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Katherine E Hutchinson
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Preston D Moore
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mónica Valéria Estrada
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Violeta Sánchez
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paula G Ericsson
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Melinda E Sanders
- Department of Pathology, Microbiology and Immunology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paula R Pohlmann
- Department of Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Michael J Pishvaian
- Department of Medicine, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - David A Riddle
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Teresa C Dugger
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Erik S Knudsen
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Carlos L Arteaga
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
32
|
TCRP1 promotes NIH/3T3 cell transformation by over-activating PDK1 and AKT1. Oncogenesis 2017; 6:e323. [PMID: 28436990 PMCID: PMC5520495 DOI: 10.1038/oncsis.2017.18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/30/2016] [Accepted: 01/24/2017] [Indexed: 01/04/2023] Open
Abstract
Tongue cancer resistance-related protein 1 (TCRP1) gene was first cloned from the multidrug resistance tongue cancer cell (Tca8113/pingyangmycin) in our lab. Our precious studies demonstrated that TCRP1 was involving in chemotherapy and radiotherapy resistance of tongue cancer cells, lung cancer cells and ovarian cancer cells. In this study, we showed that TCRP1 overexpression promotes cell transformation and tumorigenesis through hyperphosphorylation of the oncogenic kinase 3-phosphoinositide-dependent protein kinase-1 (PDK1) and AKT1, whereas inhibition of PDK1 by OSU-03012 or PDK1 small interfering RNA reversed TCRP1-mediated cell transformation. Importantly, TCRP1 was able to directly interact with PDK1, and 93–107 amino-acid and 109–124 amino-acid sites of TCRP1 were the common binding domain of PDK1. Moreover, in line with its oncogenic activity, we found that TCRP1 is often overexpressed in human in lung cancer, glioma, ovarian cancer, thyroid cancer, nasopharyngeal carcinoma, pancreatic cancer, stomach cancer and tongue carcinoma tissues. Spearman correlation analysis showed that the expression of TCRP1 has a positive correlation with p-PDK1, as well as p-AKT1 in lung cancer and gliomas tissues. Thus, TCRP1 may be a candidate as human oncoprotein that promotes cancer development by activation of PDK1/AKT1 signaling.
Collapse
|
33
|
Di Blasio L, Gagliardi PA, Puliafito A, Primo L. Serine/Threonine Kinase 3-Phosphoinositide-Dependent Protein Kinase-1 (PDK1) as a Key Regulator of Cell Migration and Cancer Dissemination. Cancers (Basel) 2017; 9:cancers9030025. [PMID: 28287465 PMCID: PMC5366820 DOI: 10.3390/cancers9030025] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 02/03/2023] Open
Abstract
Dissecting the cellular signaling that governs the motility of eukaryotic cells is one of the fundamental tasks of modern cell biology, not only because of the large number of physiological processes in which cell migration is crucial, but even more so because of the pathological ones, in particular tumor invasion and metastasis. Cell migration requires the coordination of at least four major processes: polarization of intracellular signaling, regulation of the actin cytoskeleton and membrane extension, focal adhesion and integrin signaling and contractile forces generation and rear retraction. Among the molecular components involved in the regulation of locomotion, the phosphatidylinositol-3-kinase (PI3K) pathway has been shown to exert fundamental role. A pivotal node of such pathway is represented by the serine/threonine kinase 3-phosphoinositide-dependent protein kinase-1 (PDPK1 or PDK1). PDK1, and the majority of its substrates, belong to the AGC family of kinases (related to cAMP-dependent protein kinase 1, cyclic Guanosine monophosphate-dependent protein kinase and protein kinase C), and control a plethora of cellular processes, downstream either to PI3K or to other pathways, such as RAS GTPase-MAPK (mitogen-activated protein kinase). Interestingly, PDK1 has been demonstrated to be crucial for the regulation of each step of cell migration, by activating several proteins such as protein kinase B/Akt (PKB/Akt), myotonic dystrophy-related CDC42-binding kinases alpha (MRCKα), Rho associated coiled-coil containing protein kinase 1 (ROCK1), phospholipase C gamma 1 (PLCγ1) and β3 integrin. Moreover, PDK1 regulates cancer cell invasion as well, thus representing a possible target to prevent cancer metastasis in human patients. The aim of this review is to summarize the various mechanisms by which PDK1 controls the cell migration process, from cell polarization to actin cytoskeleton and focal adhesion regulation, and finally, to discuss the evidence supporting a role for PDK1 in cancer cell invasion and dissemination.
Collapse
Affiliation(s)
- Laura Di Blasio
- Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo, Torino, Italy.
| | | | | | - Luca Primo
- Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo, Torino, Italy.
- Department of Oncology, University of Torino, 10043 Orbassano, Torino, Italy.
| |
Collapse
|
34
|
Regulation of PI3K effector signalling in cancer by the phosphoinositide phosphatases. Biosci Rep 2017; 37:BSR20160432. [PMID: 28082369 PMCID: PMC5301276 DOI: 10.1042/bsr20160432] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/24/2022] Open
Abstract
Class I phosphoinositide 3-kinase (PI3K) generates phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) at the plasma membrane in response to growth factors, activating a signalling cascade that regulates many cellular functions including cell growth, proliferation, survival, migration and metabolism. The PI3K pathway is commonly dysregulated in human cancer, and drives tumorigenesis by promoting aberrant cell growth and transformation. PtdIns(3,4,5)P3 facilitates the activation of many pleckstrin homology (PH) domain-containing proteins including the serine/threonine kinase AKT. There are three AKT isoforms that are frequently hyperactivated in cancer through mutation, amplification or dysregulation of upstream regulatory proteins. AKT isoforms have converging and opposing functions in tumorigenesis. PtdIns(3,4,5)P3 signalling is degraded and terminated by phosphoinositide phosphatases such as phosphatase and tensin homologue (PTEN), proline-rich inositol polyphosphate 5-phosphatase (PIPP) (INPP5J) and inositol polyphosphate 4-phosphatase type II (INPP4B). PtdIns(3,4,5)P3 is rapidly hydrolysed by PIPP to generate phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2), which is further hydrolysed by INPP4B to form phosphatidylinositol 3-phosphate (PtdIns3P). PtdIns(3,4)P2 and PtdIns3P are also important signalling molecules; PtdIns(3,4)P2 together with PtdIns(3,4,5)P3 are required for maximal AKT activation and PtdIns3P activates PI3K-dependent serum and glucocorticoid-regulated kinase (SGK3) signalling. Loss of Pten, Pipp or Inpp4b expression or function promotes tumour growth in murine cancer models through enhanced AKT isoform-specific signalling. INPP4B inhibits PtdIns(3,4)P2-mediated AKT activation in breast and prostate cancer; however, INPP4B expression is increased in acute myeloid leukaemia (AML), melanoma and colon cancer where it paradoxically promotes cell proliferation, transformation and/or drug resistance. This review will discuss how PTEN, PIPP and INPP4B distinctly regulate PtdIns(3,4,5)P3 signalling downstream of PI3K and how dysregulation of these phosphatases affects cancer outcomes.
Collapse
|
35
|
Biological Activities of Essential Oils: From Plant Chemoecology to Traditional Healing Systems. Molecules 2017; 22:molecules22010070. [PMID: 28045446 PMCID: PMC6155610 DOI: 10.3390/molecules22010070] [Citation(s) in RCA: 361] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/25/2016] [Indexed: 02/06/2023] Open
Abstract
Essential oils are complex mixtures of hydrocarbons and their oxygenated derivatives arising from two different isoprenoid pathways. Essential oils are produced by glandular trichomes and other secretory structures, specialized secretory tissues mainly diffused onto the surface of plant organs, particularly flowers and leaves, thus exerting a pivotal ecological role in plant. In addition, essential oils have been used, since ancient times, in many different traditional healing systems all over the world, because of their biological activities. Many preclinical studies have documented antimicrobial, antioxidant, anti-inflammatory and anticancer activities of essential oils in a number of cell and animal models, also elucidating their mechanism of action and pharmacological targets, though the paucity of in human studies limits the potential of essential oils as effective and safe phytotherapeutic agents. More well-designed clinical trials are needed in order to ascertain the real efficacy and safety of these plant products.
Collapse
|
36
|
Lion M, Harlé A, Salleron J, Ramacci C, Campone M, Merlin JL. Trastuzumab as a preoperative monotherapy does not inhibit HER2 downstream signaling in HER2-positive breast cancer. Oncol Lett 2016; 12:2028-2032. [PMID: 27602133 DOI: 10.3892/ol.2016.4891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 05/23/2016] [Indexed: 01/07/2023] Open
Abstract
Human epidermal growth factor 2 (HER2) is overexpressed in 15-20% of breast carcinomas. The overexpression of HER2 was previously associated with a poor prognosis until the development of the first anti-HER2 therapy, trastuzumab, which drastically improves the prognosis of HER2-overexpressing breast cancers. However, its mechanism of action remains not fully understood. Several studies have proposed that the behavior and mechanism of action of trastuzumab may be drastically altered in vitro and in vivo. The present study assesses the ability of trastuzumab to inhibit the phosphorylation of the key-proteins of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin and Ras/Raf/mitogen-activated protein kinase (MAPK) signaling pathways in vitro, in breast cancer cell lines and in tumor biopsies obtained from patients treated with trastuzumab preoperative monotherapy as part of the Unicancer GEP04 RADHER phase II clinical trial. HER2-positive SKBR3 and HER2-negative MCF-7 cell lines were exposed to trastuzumab for 72 h. In total, 41 patients received trastuzumab alone for 6 weeks of preoperative treatment. Biopsies were collected at the baseline and at surgery. A total of 19 pairs of associated baseline and surgery tumor specimens were eligible for protein extraction and comparative phosphoprotein expression analysis, prior to and subsequent to treatment. The expression of phosphoproteins was quantitatively assessed using a multiplex immunoassay. In the SKBR3 cell line, a statistically significant decrease of the expression level of phosphorylated (p-)AKT, p-ribosomal protein S6 kinase B1, p-extracellular signal regulated kinase 1/2 and p-mitogen-activated protein kinase kinase 1 was observed after exposure to trastuzumab. In contrast, no statistically significant variations for levels expression of these phosphoproteins were observed in patients following treatment. The lack of downregulation of PI3K and MAPK pathways could probably be explained by the implementation of a predominant immunological mechanism of action for trastuzumab, a type of antibody-dependent cell-mediated toxicity, which has previously been reported in preoperative monotherapy settings. The present study confirms that trastuzumab involves various modes of action when assayed in vitro and used clinically.
Collapse
Affiliation(s)
- Maëva Lion
- Cellular Oncology Laboratory, Faculty of Pharmacy, Université de Lorraine, Nancy 54000, France; Centre National pour la Recherche Scientifique, Unité Mixte de Recherche 7039 CRAN, Université de Lorraine, Vandœuvre-lès-Nancy 54500, France; Department of Biopathology, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy 54519, France
| | - Alexandre Harlé
- Cellular Oncology Laboratory, Faculty of Pharmacy, Université de Lorraine, Nancy 54000, France; Centre National pour la Recherche Scientifique, Unité Mixte de Recherche 7039 CRAN, Université de Lorraine, Vandœuvre-lès-Nancy 54500, France; Department of Biopathology, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy 54519, France
| | - Julia Salleron
- Biostatistics Unit, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy 54519, France
| | - Carole Ramacci
- Department of Biopathology, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy 54519, France
| | - Mario Campone
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche 892, Nantes 44007, France; Department of Medical Oncology, Institut de Cancérologie de l'Ouest, St Herblain, Nantes 44800, France
| | - Jean-Louis Merlin
- Cellular Oncology Laboratory, Faculty of Pharmacy, Université de Lorraine, Nancy 54000, France; Centre National pour la Recherche Scientifique, Unité Mixte de Recherche 7039 CRAN, Université de Lorraine, Vandœuvre-lès-Nancy 54500, France; Department of Biopathology, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy 54519, France
| |
Collapse
|
37
|
Akkiprik M, Nicorici D, Cogdell D, Jia YJ, Hategan A, Tabus I, Yli-Harja O, Y D, Sahin A, Zhang W. Dissection of Signaling Pathways in Fourteen Breast Cancer Cell Lines Using Reverse-Phase Protein Lysate Microarray. Technol Cancer Res Treat 2016; 5:543-51. [PMID: 17121430 DOI: 10.1177/153303460600500601] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Signal transduction pathways play a crucial role in breast cancer development, progression, and response to different therapies. A major problem in breast cancer therapy is the heterogeneity among different tumor types and cell lines commonly used in preclinical studies. To characterize the signaling pathways of some of the commonly used breast cancer cell lines and dissect the relationship among a number of pathways and some key genetic and molecular events in breast cancer development, such as p53 mutation, ErbB2 expression, and estrogen receptor (ER)/progesterone receptor (PR) status, we performed pathway profiling of 14 breast cancer cell lines by measuring the expression and phosphorylation status of 40 different cell signaling proteins with 53 specific antibodies using a protein lysate array. Cluster analysis of the expression data showed that there was close clustering of phosphatidylinositol 3-kinase, Akt, mammalian target of rapamycin (mTOR), Src, and platelet-derived growth factor receptor β (PDGFRβ) in all of the cell lines. The most differentially expressed proteins between ER- and PR-positive and ER- and PR-negative breast cells were mTOR, Akt (pThr308), PDGFRβ, PDGFRβ (pTyr751), panSrc, Akt (pSer473), insulin-like growth factor-binding protein 5 (IGFBP5), Src (pTyr418), mTOR (pSer2448), and IGFBP2. Many apoptotic proteins, such as apoptosis-inducing factor, IGFBP3, bad, bax, and cleaved caspase 9, were overexpressed in mutant p53-carrying breast cancer cells. Hexokinase isoenzyme 1, ND2, and c-kit were the most differentially expressed proteins in high and low ErbB2-expressing breast cancer cells. This study demonstrated that ER/PR status, ErbB2 expression, and p53 status are major molecules that impact downstream signaling pathways.
Collapse
Affiliation(s)
- M Akkiprik
- Department of Pathology, Unit 85, The University of Texas, M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ren HY, Wang J, Yang F, Zhang XL, Wang AL, Sun LL, Diao KX, Wang EH, Mi XY. Cytoplasmic TRAF4 contributes to the activation of p70s6k signaling pathway in breast cancer. Oncotarget 2016; 6:4080-96. [PMID: 25738361 PMCID: PMC4414174 DOI: 10.18632/oncotarget.2977] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 12/18/2014] [Indexed: 12/18/2022] Open
Abstract
Tumor necrosis factor receptor associated factor 4 (TRAF4) is an important adaptor protein that plays a significant role in several signaling pathways. By studying the relationship between TRAF4 and 70 kDa ribosomal protein S6 kinase (p70s6k) in vivo, we demonstrated that cytoplasmic TRAF4 was correlated with the activation of p70s6k in breast cancer. Moreover, we found that cytoplasmic TRAF4 expression in breast cancer patients was significantly associated with a poor prognosis. To determine the exact mechanism, we analyzed the interaction between TRAF4 and p70s6k and identified the Zinc fingers domain of TRAF4 was responsible for their interaction in MCF7 cells. Furthermore, we found that activation of p70s6k/S6 signaling pathway by TRAF4 requires the mammalian target of rapamycin (mTOR) activity; TRAF4 acted as a sensitizer. Tumor necrosis factor receptor associated factor 2 (TRAF2), as a binding partner of TRAF4, could also promoted activation of p70s6k signaling via upregulating cytoplasm expression of TRAF4 and played a critical role in TNFa-induced activation of p70s6k/S6 pathway. Finally, we demonstrated p70s6k/S6 signaling pathway played an important role in the promoting function of TRAF4 on cell proliferation. In summary, our work suggests a new direction for understanding the oncogenic function of TRAF4 in breast cancer.
Collapse
Affiliation(s)
- Hua-Yan Ren
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - Jian Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - Fan Yang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - Xiao-Li Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - Ai-Lian Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - Li-Li Sun
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - Ke-Xin Diao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - En-Hua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| | - Xiao-Yi Mi
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, P.R. China
| |
Collapse
|
39
|
Yu L, Andruska N, Zheng X, Shapiro DJ. Anticipatory activation of the unfolded protein response by epidermal growth factor is required for immediate early gene expression and cell proliferation. Mol Cell Endocrinol 2016; 422:31-41. [PMID: 26551735 PMCID: PMC4919024 DOI: 10.1016/j.mce.2015.11.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 12/13/2022]
Abstract
The onco-protein epidermal growth factor (EGF) initiates a cascade that includes activation of the ERK and AKT signaling pathways and alters gene expression. We describe a new action of EGF-EGF receptor (EGFR), rapid anticipatory activation of the endoplasmic reticulum stress sensor, the unfolded protein response (UPR). Within 2 min, EGF elicits EGFR dependent activation of phospholipase C γ (PLCγ), producing inositol triphosphate (IP3), which binds to IP3 receptor (IP3R), opening the endoplasmic reticulum IP3R Ca(2+) channels, resulting in increased intracellular Ca(2+). This calcium release leads to transient and moderate activation of the IRE1α and ATF6α arms of the UPR, resulting in induction of BiP chaperone. Knockdown or inhibition of EGFR, PLCγ or IP3R blocks the increase in intracellular Ca(2+). While blocking the increase in intracellular Ca(2+) by locking the IP3R calcium channel with 2-APB had no effect on EGF activation of the ERK or AKT signaling pathways, it abolished the rapid EGF-mediated induction and repression of gene expression. Knockdown of ATF6α or XBP1, which regulate UPR-induced chaperone production, inhibited EGF stimulated cell proliferation. Supporting biological relevance, increased levels of EGF receptor during tumor progression were correlated with increased expression of the UPR gene signature. Anticipatory activation of the UPR is a new role for EGF. Since UPR activation occurs in <2 min, it is an initial cell response when EGF binds EGFR.
Collapse
Affiliation(s)
- Liqun Yu
- Department of Biochemistry, University of Illinois, Urbana, IL, 61801, USA
| | - Neal Andruska
- Department of Biochemistry, University of Illinois, Urbana, IL, 61801, USA; College of Medicine, University of Illinois, USA
| | - Xiaobin Zheng
- Department of Biochemistry, University of Illinois, Urbana, IL, 61801, USA
| | - David J Shapiro
- Department of Biochemistry, University of Illinois, Urbana, IL, 61801, USA; University of Illinois Cancer Center, USA; College of Medicine, University of Illinois, USA.
| |
Collapse
|
40
|
PDK1 promotes tumor growth and metastasis in a spontaneous breast cancer model. Oncogene 2015; 35:3314-23. [DOI: 10.1038/onc.2015.393] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 09/06/2015] [Accepted: 09/08/2015] [Indexed: 12/25/2022]
|
41
|
Ruiz de Garibay G, Herranz C, Llorente A, Boni J, Serra-Musach J, Mateo F, Aguilar H, Gómez-Baldó L, Petit A, Vidal A, Climent F, Hernández-Losa J, Cordero Á, González-Suárez E, Sánchez-Mut JV, Esteller M, Llatjós R, Varela M, López JI, García N, Extremera AI, Gumà A, Ortega R, Plà MJ, Fernández A, Pernas S, Falo C, Morilla I, Campos M, Gil M, Román A, Molina-Molina M, Ussetti P, Laporta R, Valenzuela C, Ancochea J, Xaubet A, Casanova Á, Pujana MA. Lymphangioleiomyomatosis Biomarkers Linked to Lung Metastatic Potential and Cell Stemness. PLoS One 2015; 10:e0132546. [PMID: 26167915 PMCID: PMC4500593 DOI: 10.1371/journal.pone.0132546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 06/17/2015] [Indexed: 12/23/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare lung-metastasizing neoplasm caused by the proliferation of smooth muscle-like cells that commonly carry loss-of-function mutations in either the tuberous sclerosis complex 1 or 2 (TSC1 or TSC2) genes. While allosteric inhibition of the mechanistic target of rapamycin (mTOR) has shown substantial clinical benefit, complementary therapies are required to improve response and/or to treat specific patients. However, there is a lack of LAM biomarkers that could potentially be used to monitor the disease and to develop other targeted therapies. We hypothesized that the mediators of cancer metastasis to lung, particularly in breast cancer, also play a relevant role in LAM. Analyses across independent breast cancer datasets revealed associations between low TSC1/2 expression, altered mTOR complex 1 (mTORC1) pathway signaling, and metastasis to lung. Subsequently, immunohistochemical analyses of 23 LAM lesions revealed positivity in all cases for the lung metastasis mediators fascin 1 (FSCN1) and inhibitor of DNA binding 1 (ID1). Moreover, assessment of breast cancer stem or luminal progenitor cell biomarkers showed positivity in most LAM tissue for the aldehyde dehydrogenase 1 (ALDH1), integrin-ß3 (ITGB3/CD61), and/or the sex-determining region Y-box 9 (SOX9) proteins. The immunohistochemical analyses also provided evidence of heterogeneity between and within LAM cases. The analysis of Tsc2-deficient cells revealed relative over-expression of FSCN1 and ID1; however, Tsc2-deficient cells did not show higher sensitivity to ID1-based cancer inhibitors. Collectively, the results of this study reveal novel LAM biomarkers linked to breast cancer metastasis to lung and to cell stemness, which in turn might guide the assessment of additional or complementary therapeutic opportunities for LAM.
Collapse
Affiliation(s)
- Gorka Ruiz de Garibay
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Carmen Herranz
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Alicia Llorente
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Jacopo Boni
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Jordi Serra-Musach
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Francesca Mateo
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Helena Aguilar
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Laia Gómez-Baldó
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Anna Petit
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - August Vidal
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Fina Climent
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | | | - Álex Cordero
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Eva González-Suárez
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - José Vicente Sánchez-Mut
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
- Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Roger Llatjós
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Mar Varela
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - José Ignacio López
- Cruces University Hospital, BioCruces Research Institute, University of the Basque Country, Barakaldo, Spain
| | - Nadia García
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Ana I. Extremera
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Anna Gumà
- Department of Radiology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Raúl Ortega
- Department of Radiology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - María Jesús Plà
- Department of Gynecology, University Hospital of Bellvitge, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Adela Fernández
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Sònia Pernas
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Catalina Falo
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Idoia Morilla
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Miriam Campos
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Miguel Gil
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Antonio Román
- Department of Pulmonology, Lung Transplant Unit, Lymphangioleiomyomatosis (LAM) Clinic, Vall d'Hebron University Hospital, Barcelona, Catalonia, Spain
| | - María Molina-Molina
- Department of Pneumology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
- Biomedical Research Centre Network for Respiratory Diseases (CIBERES), Madrid, Spain
| | - Piedad Ussetti
- Department of Pneumology, University Hospital Clínica Puerta del Hierro, Madrid, Spain
| | - Rosalía Laporta
- Department of Pneumology, University Hospital Clínica Puerta del Hierro, Madrid, Spain
| | - Claudia Valenzuela
- Department of Pneumology, Instituto de Investigación Sanitaria La Princesa, Hospital La Princesa, Madrid, Spain
| | - Julio Ancochea
- Department of Pneumology, Instituto de Investigación Sanitaria La Princesa, Hospital La Princesa, Madrid, Spain
| | - Antoni Xaubet
- Biomedical Research Centre Network for Respiratory Diseases (CIBERES), Madrid, Spain
- Department of Pneumology, Hospital Clinic of Barcelona, Agusti Pi Suñer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain
| | | | - Miguel Angel Pujana
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
- * E-mail:
| |
Collapse
|
42
|
Abstract
Mechanistic target of rapamycin (mTOR) is a central component of the essential signaling pathway that regulates cell growth and proliferation by controlling anabolic processes in cells. mTOR exists in two distinct mTOR complexes known as mTORC1 and mTORC2 that reside mostly in cytoplasm. In our study, the biochemical characterization of mTOR led to discovery of its novel localization on nuclear envelope where it associates with a critical regulator of nuclear import Ran Binding Protein 2 (RanBP2). We show that association of mTOR with RanBP2 is dependent on the mTOR kinase activity that regulates the nuclear import of ribosomal proteins. The mTOR kinase inhibitors within thirty minutes caused a substantial decrease of ribosomal proteins in the nuclear but not cytoplasmic fraction. Detection of a nuclear accumulation of the GFP-tagged ribosomal protein rpL7a also indicated its dependence on the mTOR kinase activity. The nuclear abundance of ribosomal proteins was not affected by inhibition of mTOR Complex 1 (mTORC1) by rapamycin or deficiency of mTORC2, suggesting a distinctive role of the nuclear envelope mTOR complex in the nuclear import. Thus, we identified that mTOR in association with RanBP2 mediates the active nuclear import of ribosomal proteins.
Collapse
|
43
|
Xu Z, Liao B, Zhang R, Yao J, Shi R, Wang L. Expression of 3-phosphoinositide-dependent protein kinase 1 in colorectal cancer as a potential therapeutic target. Med Oncol 2015; 32:198. [PMID: 26055151 DOI: 10.1007/s12032-015-0645-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 05/21/2015] [Indexed: 01/05/2023]
Abstract
3-Phosphoinositide-dependent protein kinase 1 (PDK1) is centrally involved in cancer progression, including proliferation, apoptosis and invasion. However, its expression pattern and possible cellular functions in human colorectal cancer remain unclear. In the present study, we show that PDK1 expression is up-regulated at both mRNA and protein levels in colorectal cancer clinical specimens and cell lines. Transient knockdown of PDK1 suppresses cellular growth, induces cellular apoptosis and causes abnormal cell cycle distribution. Meanwhile, decreased PDK1 level is closely associated with reduced Akt/cyclin D1 activity. Activating AKT activity and reintroducing cyclin D1 expression significantly compromised the oncogenic activity induced by PDK1. Together, our findings elucidate a key role for PDK1 in colorectal cellular functions trigged by the Akt/cyclin D1 pathway, thus providing a novel insight of PDK1 in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Zhenglei Xu
- Department of Gastroenterology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, 1017#, North Dongmen Road, Shenzhen, 518000, Guangdong Province, People's Republic of China
| | | | | | | | | | | |
Collapse
|
44
|
Lapin V, Shirdel EA, Wei X, Mason JM, Jurisica I, Mak TW. Kinome-wide screening of HER2+ breast cancer cells for molecules that mediate cell proliferation or sensitize cells to trastuzumab therapy. Oncogenesis 2014; 3:e133. [PMID: 25500906 PMCID: PMC4275559 DOI: 10.1038/oncsis.2014.45] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 08/19/2014] [Indexed: 01/18/2023] Open
Abstract
Understanding the signaling differences that distinguish human HER2-amplified (HER2-positive (HER2+)) breast cancers from other breast cancer subtypes may help to identify protein drug targets for the specific treatment of HER2+ breast cancers. We performed two kinome-wide small interfering RNA (siRNA) screens on five HER2+ breast cancer cell lines, seven breast cancer cell lines in which HER2 was not amplified and two normal breast cell lines. To pinpoint the main kinases driving HER2 signaling, we performed a comprehensive siRNA screen that identified loss of the HER2/HER3 heterodimer as having the most prominent inhibitory effect on the growth of HER2+ breast cancer cells. In a second siRNA screen focused on identifying genes that could sensitize HER2+ cells to trastuzumab treatment, we found that loss of signaling members downstream of phosphatidylinositol 3 kinase (PI3K) potentiated the growth inhibitory effects of trastuzumab. Loss of HER2 and HER3, as well as proteins involved in mitogenic and environmental stress pathways inhibited the proliferation of HER2+ cells only in the absence of trastuzumab, suggesting that these pathways are inhibited by trastuzumab treatment. Loss of essential G2/M cell cycle mediators or proteins involved in vesicle organization exerted inhibitory effects on HER2+ cell growth that were unaffected by trastuzumab. Furthermore, the use of a sensitization index (SI) identified targeting the PI3K pathway to sensitize to trastuzumab treatment. Antagonism using the SI identified MYO3A, MYO3B and MPZL1 as antagonizers to trastuzumab treatment among HER2+ cell lines. Our results suggest that the dimerization partners of HER2 are important for determining the activation of downstream proliferation pathways. Understanding the complex layers of signaling triggered downstream of HER2 homodimers and heterodimers will facilitate the selection of better targets for combination therapies intended to treat HER2+ breast cancers.
Collapse
Affiliation(s)
- V Lapin
- 1] Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada [2] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - E A Shirdel
- 1] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada [2] Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - X Wei
- Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada
| | - J M Mason
- Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada
| | - I Jurisica
- 1] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada [2] Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - T W Mak
- 1] Campbell Family Institute for Breast Cancer Research, Toronto, Ontario, Canada [2] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Duan H, Qu L, Shou C. Activation of EGFR-PI3K-AKT signaling is required for Mycoplasma hyorhinis-promoted gastric cancer cell migration. Cancer Cell Int 2014; 14:135. [PMID: 25505372 PMCID: PMC4262230 DOI: 10.1186/s12935-014-0135-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 11/20/2014] [Indexed: 01/26/2023] Open
Abstract
Persistent infection of Mycoplasma hyorhinis (M. hyorhinis) was associated with gastric cancer cell migration and invasion, but the mechanisms were not well understood. Herein, we found that M. hyorhinis activated phosphoinositide 3-kinase (PI3K)-AKT signaling axis in gastric cancer cell lines. Epidermal growth factor receptor (EGFR) was upstream of PI3K-AKT signaling in the context of M. hyorhinis infection, because phosphorylation of AKT Serine 473 was almost completely attenuated by the EGFR inhibitor AG1478 or by EGFR knockdown. Phosphorylation of AKT S473 induced by M. hyorhinis infection was also abolished by PI3K inhibitor wortmannin. Furthermore, we found that p37, a membrane protein of M. hyorhinis, could also promote M. hyorhinis-induced PI3K-AKT signaling activation and cell migration. In addition, pre-treatment with AG1478 or wortmannin significantly inhibited cell migration induced by M. hyorhinis infection or p37 treatment. In conclusion, EGFR-PI3K-AKT signaling plays an important role in M. hyorhinis-promoted cell migration in gastric cancer cells, thus providing a clue to the pathogenesis of M. hyorhinis in gastric cancer.
Collapse
Affiliation(s)
- Hongying Duan
- Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 52 Fucheng Road, Beijing, 100142 China
| | - Like Qu
- Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 52 Fucheng Road, Beijing, 100142 China
| | - Chengchao Shou
- Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 52 Fucheng Road, Beijing, 100142 China
| |
Collapse
|
46
|
Hu Y, Zhu Q, Tang L. MiR-99a antitumor activity in human breast cancer cells through targeting of mTOR expression. PLoS One 2014; 9:e92099. [PMID: 24637915 PMCID: PMC3956864 DOI: 10.1371/journal.pone.0092099] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 02/19/2014] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) play an important role in human tumorigenesis as oncogenes or tumor suppressors. miR-99a has been reported as a tumor suppressor gene in various cancers in humans. However, only limited information about the function of miR-99a in human breast cancers is available. Here we investigated the expression of miR-99a in breast cancer tissue specimens and its antitumor activity in breast cancer cells. We initially identified that the expression of miR-99a was significantly reduced in four breast cancer cell lines. More importantly, we found downregulation of miR-99a in breast cancer specimens from ten different patients. We then analyzed the mechanism of miR-99a in inhibiting tumorigenesis. Cell-based assays that showed overexpression of miR-99a not only reduced breast cancer cell viability by inducing accumulation of cells at sub-G1 phase and cell apoptosis, but also inhibited tumorigenicity in vivo. As a critical miR-99a target, we have shown that the function of mammalian target of rapamycin (mTOR) was greatly inhibited by miR-99a-based Luciferase report assay; overexpression of miR-99a reduced the expression of mTOR and its downstream phosphorylated proteins (p-4E-BP1 and p-S6K1). Similar to restoring miR-99a expression, mTOR downregulation suppressed cell viability and increased cell apoptosis, whereas restoration of mTOR expression significantly reversed the inhibitory effects of miR-99a on the mTOR/p-4E-BP1/p-S6K1 signal pathway and the miR-99a antitumor activity. In clinical specimens and cell lines, mTOR was commonly overexpressed and its protein levels were statistically inversely correlated with miR-99a expression. Taken together, these results have demonstrated that miR-99a antitumor activity is achieved by targeting the mTOR/p-4E-BP1/p-S6K1 pathway in human breast cancer cells. This study suggests a potential therapeutic strategy to effectively control breast cancer development.
Collapse
Affiliation(s)
- Yu Hu
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Zhu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lili Tang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, China
- * E-mail:
| |
Collapse
|
47
|
Hanberry BS, Berger R, Zastre JA. High-dose vitamin B1 reduces proliferation in cancer cell lines analogous to dichloroacetate. Cancer Chemother Pharmacol 2014; 73:585-94. [PMID: 24452394 DOI: 10.1007/s00280-014-2386-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 01/02/2014] [Indexed: 12/26/2022]
Abstract
PURPOSE The dichotomous effect of thiamine supplementation on cancer cell growth is characterized by growth stimulation at low doses and growth suppression at high doses. Unfortunately, how thiamine reduces cancer cell proliferation is currently unknown. Recent focuses on metabolic targets for cancer therapy have exploited the altered regulation of the thiamine-dependent enzyme pyruvate dehydrogenase (PDH). Cancer cells inactivate PDH through phosphorylation by overexpression of pyruvate dehydrogenase kinases (PDKs). Inhibition of PDKs by dichloracetate (DCA) exhibits a growth suppressive effect in many cancers. Recently, it has been shown that the thiamine coenzyme, thiamine pyrophosphate reduces PDK-mediated phosphorylation of PDH. Therefore, the objective of this study was to determine whether high-dose thiamine supplementation reduces cell proliferation through a DCA-like mechanism. METHODS Cytotoxicity of thiamine and DCA was assessed in SK-N-BE and Panc-1 cancer cell lines. Comparative effects of high-dose thiamine and DCA on PDH phosphorylation were measured by Western blot. The metabolic impact of PDH reactivation was determined by glucose and lactate assays. Changes in the mitochondrial membrane potential, reactive oxygen species (ROS) production, and caspase-3 activation were assessed to characterize the mechanism of action. RESULTS Thiamine exhibited a lower IC50 value in both cell lines compared with DCA. Both thiamine and DCA reduced the extent of PDH phosphorylation, reduced glucose consumption, lactate production, and mitochondrial membrane potential. High-dose thiamine and DCA did not increase ROS, but increased caspase-3 activity. CONCLUSION Our findings suggest that high-dose thiamine reduces cancer cell proliferation by a mechanism similar to that described for dichloroacetate.
Collapse
Affiliation(s)
- Bradley S Hanberry
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, R.C. Wilson Pharmacy Building, Athens, GA, 30602, USA
| | | | | |
Collapse
|
48
|
Godovac-Zimmermann J, Mulvey C, Konstantoulaki M, Sainsbury R, Brown LR. Promise of multiphoton detection in discovery and diagnostic proteomics. Expert Rev Proteomics 2014; 4:161-73. [PMID: 17425453 DOI: 10.1586/14789450.4.2.161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Proteomics has lacked adequate methods for handling the complexity (hundreds of thousands of different proteins) and range of protein concentrations (> or =10(6)) of eukaryotic proteomes. New multiphoton-detection methods for ultrasensitive detection of proteins produce 10,000-fold gains in sensitivity and allow highly quantitative, linear detection of 50 zmol (30,000 molecules) to 500 fmol of proteins in complex samples. The potential of multiphoton detection in top-down proteomics analyses is illustrated with applications in monitoring proteomes in very small numbers of cells, in identifying and monitoring complex functional isoforms of cancer-related proteins, and in super-sensitive immunoassays of serum proteins for high-performance detection of cancer.
Collapse
|
49
|
Takahashi Y, Kohashi K, Yamada Y, Endo M, Setsu N, Ishii T, Yamamoto H, Iwamoto Y, Oda Y. Activation of the Akt/mammalian target of rapamycin pathway in myxofibrosarcomas. Hum Pathol 2014; 45:984-93. [PMID: 24746202 DOI: 10.1016/j.humpath.2013.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 10/25/2022]
Abstract
The Akt/mammalian target of rapamycin (mTOR) pathway plays important roles in modulating cellular function in response to extracellular signals such as growth factors and cytokines. The Akt/mTOR signaling pathway is activated in certain kinds of sarcomas. Myxofibrosarcoma is a soft tissue sarcoma, characterized by abundant myxoid stroma and frequent local recurrence. Here, we conducted a large-scale examination of the clinicopathological and activation statuses of the Akt/mTOR pathways in myxofibrosarcoma. The phosphorylation status of Akt, mTOR, S6 ribosomal protein, and the eukaryotic translation initiation factor 4E-binding protein, and mitogen-activated protein kinase were assessed by immunohistochemistry in 101 formalin-fixed, paraffin-embedded samples, including 68 primary tumors in myxofibrosarcoma. Immunohistochemical expressions were confirmed by Western blotting with 20 frozen samples, which were paired with normal tissue samples. PIK3CA and AKT1 gene mutations were also analyzed using 12 primary tumor frozen samples. Immunohistochemically, phosphorylations of Akt, mTOR, S6 ribosomal protein, 4E-binding protein, and mitogen-activated protein kinase 1/2 were observed in 64.7%, 45.6%, 42.6%, 63.2%, and 64.7% of samples. Phosphorylated Akt/mTOR pathway proteins were correlated with one another and were also correlated with the phosphorylation of these proteins in the concordant recurrent tumors. Immunoblotting showed a high degree of phosphorylation in tumor samples, compared with that in normal tissue samples. Activation of the Akt/mTOR pathway was correlated with histologic grade and tumor progression. Mutational analysis failed to reveal any PIK3CA or AKT1 mutations around the hot spots. Activation of the Akt/mTOR pathway was associated with histologic malignancy and tumor progression in primary and recurrent myxofibrosarcoma.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Makoto Endo
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Nokitaka Setsu
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takeaki Ishii
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yukihide Iwamoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
50
|
Yamada Y, Kohashi K, Fushimi F, Takahashi Y, Setsu N, Endo M, Yamamoto H, Tokunaga S, Iwamoto Y, Oda Y. Activation of the Akt-mTOR pathway and receptor tyrosine kinase in patients with solitary fibrous tumors. Cancer 2013; 120:864-76. [DOI: 10.1002/cncr.28506] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 10/30/2013] [Accepted: 11/04/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Yuichi Yamada
- Department of Anatomic Pathology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | | | - Yusuke Takahashi
- Department of Anatomic Pathology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Nokitaka Setsu
- Department of Anatomic Pathology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Makoto Endo
- Department of Anatomic Pathology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Shoji Tokunaga
- Medical Information Center; Kyushu University Hospital; Fukuoka Japan
| | - Yukihide Iwamoto
- Department of Orthopaedic Surgery; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| |
Collapse
|