1
|
Xu S, Zhang Z, Zhou X, Liao Y, Peng Z, Meng Z, Nüssler AK, Ma L, Xia H, Liu L, Yang W. Gouqi-derived Nanovesicles (GqDNVs) promoted MC3T3-E1 cells proliferation and improve fracture healing. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156755. [PMID: 40252435 DOI: 10.1016/j.phymed.2025.156755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Lycium barbarum L., also known as Gouqi, a traditional Chinese herbal medicine, is widely utilized in health care products and clinical therapies. Its muscle and bone strengthening efficacy has been recorded in medical classics for a long time. In addition, plant exosome-like nanovesicles (PELNVs) have attracted more and more attention owing to their biological traits. Therefore, we intended to explore the functions, regulatory role, and underlying mechanism of Gouqi-derived Nanovesicles (GqDNVs) on fracture healing. METHODS In this study, we employed the sucrose density gradient differential ultracentrifugation to isolate GqDNVs. The effects of GqDNVs on the proliferation and differentiation of MC3T3-E1 cells were evaluated using the CCK-8 assay, ALP activity measurement, and cell scratch assay. Additionally, leveraging a fracture mouse model, we utilized Micro-CT, immunological staining, and histologic analyses to comprehensively assess the impact of GqDNVs on fracture healing in mice. RESULTS GqDNVs stimulated cell viability, increased ALP activity, and promoted cellular osteogenic protein expression (OPN, ALP, and RUNX2). Subsequently, in the mouse fracture model, trabecular thickness, and bone marrow density were increased in the GqDNVs treatment group after 28 days of injection. Meanwhile, the expressions of OPN and BGP were significantly elevated after both 14 and 28 days. Additionally, the expressions of p-PI3K/PI3K, p-Akt/Akt, p-mTOR/mTOR, p-4EBP1/4EBP1 and p-p70S6K/ p70S6K were also increased after14 days of treatment. CONCLUSIONS GqDNVs effectively promoted the proliferation and differentiation of MC3T3-E1 cells. Furthermore, GqDNVs could improve fracture healing, which is associated with PI3K/Akt/mTOR/p70S6K/4EBP1 signaling pathway.
Collapse
Affiliation(s)
- Shiyin Xu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zixuan Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Xiaolei Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zitong Meng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, Tübingen 72076, Germany
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China.
| |
Collapse
|
2
|
Liu J, Wang H, Wan H, Yang J, Gao L, Wang Z, Zhang X, Han W, Peng J, Yang L, Hong L. NEK6 dampens FOXO3 nuclear translocation to stabilize C-MYC and promotes subsequent de novo purine synthesis to support ovarian cancer chemoresistance. Cell Death Dis 2024; 15:661. [PMID: 39256367 PMCID: PMC11387829 DOI: 10.1038/s41419-024-07045-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
De novo purine synthesis metabolism plays a crucial role in tumor cell survival and malignant progression. However, the specific impact of this metabolic pathway on chemoresistance in ovarian cancer remains unclear. This study aims to elucidate the influence of de novo purine synthesis on chemoresistance in ovarian cancer and its underlying regulatory mechanisms. We analyzed metabolic differences between chemosensitive and chemoresistant ovarian cancer tissues using mass spectrometry-based metabolomics. Cell growth, metabolism, chemoresistance, and DNA damage repair characteristics were assessed in vitro using cell line models. Tumor growth and chemoresistance were assessed in vivo using ovarian cancer xenograft tumors. Intervention of purines and NEK6-mediated purine metabolism on chemoresistance was investigated at multiple levels. Chemoresistant ovarian cancers exhibited higher purine abundance and NEK6 expression. Inhibiting NEK6 led to decreased de novo purine synthesis, resulting in diminished chemoresistance in ovarian cancer cells. Mechanistically, NEK6 directly interacted with FOXO3, contributing to the phosphorylation of FOXO3 at S7 through its kinase activity, thereby inhibiting its nuclear translocation. Nuclear FOXO3 promoted FBXW7 transcription, leading to c-MYC ubiquitination and suppression of de novo purine synthesis. Paeonol, by inhibiting NEK6, suppressed de novo purine synthesis and enhanced chemosensitivity. The NEK6-mediated reprogramming of de novo purine synthesis emerges as a critical pathway influencing chemoresistance in ovarian cancer. Paeonol exhibits the potential to interfere with NEK6, thereby inhibiting chemoresistance.
Collapse
Affiliation(s)
- Jingchun Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Haoyu Wang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Huanzhi Wan
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Jiang Yang
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, Xiangyang, Hubei, China
| | - Likun Gao
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Zhi Wang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Xiaoyi Zhang
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Wuyue Han
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Jiaxin Peng
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Lian Yang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- The First Clinical School of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Brugnoli F, Dell’Aira M, Tedeschi P, Grassilli S, Pierantoni M, Foschi R, Bertagnolo V. Effects of Garlic on Breast Tumor Cells with a Triple Negative Phenotype: Peculiar Subtype-Dependent Down-Modulation of Akt Signaling. Cells 2024; 13:822. [PMID: 38786044 PMCID: PMC11119207 DOI: 10.3390/cells13100822] [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: 03/29/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Breast cancer includes tumor subgroups with morphological, molecular, and clinical differences. Intrinsic heterogeneity especially characterizes breast tumors with a triple negative phenotype, often leading to the failure of even the most advanced therapeutic strategies. To improve breast cancer treatment, the use of natural agents to integrate conventional therapies is the subject of ever-increasing attention. In this context, garlic (Allium sativum) shows anti-cancerous potential, interfering with the proliferation, motility, and malignant progression of both non-invasive and invasive breast tumor cells. As heterogeneity could be at the basis of variable effects, the main objective of our study was to evaluate the anti-tumoral activity of a garlic extract in breast cancer cells with a triple negative phenotype. Established triple negative breast cancer (TNBC) cell lines from patient-derived xenografts (PDXs) were used, revealing subtype-dependent effects on morphology, cell cycle, and invasive potential, correlated with the peculiar down-modulation of Akt signaling, a crucial regulator in solid tumors. Our results first demonstrate that the effects of garlic on TNBC breast cancer are not unique and suggest that only more precise knowledge of the mechanisms activated by this natural compound in each tumor will allow for the inclusion of garlic in personalized therapeutic approaches to breast cancer.
Collapse
Affiliation(s)
- Federica Brugnoli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Marcello Dell’Aira
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Paola Tedeschi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Silvia Grassilli
- Department of Environmental Sciences and Prevention and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Marina Pierantoni
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Rebecca Foschi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| | - Valeria Bertagnolo
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.B.); (M.D.); (M.P.); (R.F.)
| |
Collapse
|
4
|
Önel T, Arıcıoğlu F, Yıldırım E, Zortul H, Yaba A. The effect of maternal separation stress-induced depression on ovarian reserve in Sprague Dawley Rats: The possible role of imipramine and agmatine through a mTOR signal pathway. Physiol Behav 2023:114270. [PMID: 37308044 DOI: 10.1016/j.physbeh.2023.114270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
PURPOSE To examine the possible role of impramine and agmatine through a mTOR signal pathway on rat ovary after maternal separation stress-induced depression. METHODS Sprague Dawley neonatal female rats were divided into control, maternal separation (MS), MS+imipramine, and MS+agmatine groups. Rats were subjected to MS for 4 hours daily from postnatal day (PND) 2 to PND 21 and pups were exposed to social isolation (SI) on PND23 for 37 days for model establishment treated with imipramine (30 mg/kg; ip) or agmatine (40 mg/kg; ip) for 15 days. In order to examine behavioral changes rats were all subjected to locomotor activity and forced swimming tests (FST). Ovaries were isolated for morphological evaluation, follicle counting and mTOR signal pathway protein expression levels were detected. RESULTS Increased number of primordial follicles and diminished ovarian reserve in the MS groups were detected. Imipramine treatment caused diminished ovarian reserve and atretic follicle; however, agmatine treatment provided the maintenance of ovarian follicular reserve after MS. mTOR signal pathway may have an important role during rat ovarian follicular development in model of MS. CONCLUSIONS Our findings suggest that agmatine may help to protect ovarian reserve during follicular development by controlling cell growth.
Collapse
Affiliation(s)
- Tuğçe Önel
- Yeditepe University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Türkiye
| | - Feyza Arıcıoğlu
- Marmara University, Institute of Health Sciences, İstanbul, Türkiye
| | - Ecem Yıldırım
- Yeditepe University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Türkiye
| | - Hacer Zortul
- Marmara University, Institute of Health Sciences, İstanbul, Türkiye
| | - Aylin Yaba
- Yeditepe University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Türkiye..
| |
Collapse
|
5
|
Rajendran AK, Sankar D, Amirthalingam S, Kim HD, Rangasamy J, Hwang NS. Trends in mechanobiology guided tissue engineering and tools to study cell-substrate interactions: a brief review. Biomater Res 2023; 27:55. [PMID: 37264479 DOI: 10.1186/s40824-023-00393-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Sensing the mechanical properties of the substrates or the matrix by the cells and the tissues, the subsequent downstream responses at the cellular, nuclear and epigenetic levels and the outcomes are beginning to get unraveled more recently. There have been various instances where researchers have established the underlying connection between the cellular mechanosignalling pathways and cellular physiology, cellular differentiation, and also tissue pathology. It has been now accepted that mechanosignalling, alone or in combination with classical pathways, could play a significant role in fate determination, development, and organization of cells and tissues. Furthermore, as mechanobiology is gaining traction, so do the various techniques to ponder and gain insights into the still unraveled pathways. This review would briefly discuss some of the interesting works wherein it has been shown that specific alteration of the mechanical properties of the substrates would lead to fate determination of stem cells into various differentiated cells such as osteoblasts, adipocytes, tenocytes, cardiomyocytes, and neurons, and how these properties are being utilized for the development of organoids. This review would also cover various techniques that have been developed and employed to explore the effects of mechanosignalling, including imaging of mechanosensing proteins, atomic force microscopy (AFM), quartz crystal microbalance with dissipation measurements (QCMD), traction force microscopy (TFM), microdevice arrays, Spatio-temporal image analysis, optical tweezer force measurements, mechanoscanning ion conductance microscopy (mSICM), acoustofluidic interferometric device (AID) and so forth. This review would provide insights to the researchers who work on exploiting various mechanical properties of substrates to control the cellular and tissue functions for tissue engineering and regenerative applications, and also will shed light on the advancements of various techniques that could be utilized to unravel the unknown in the field of cellular mechanobiology.
Collapse
Affiliation(s)
- Arun Kumar Rajendran
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Deepthi Sankar
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwan D Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Jayakumar Rangasamy
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
6
|
Chen XR, Igumenova TI. Regulation of eukaryotic protein kinases by Pin1, a peptidyl-prolyl isomerase. Adv Biol Regul 2023; 87:100938. [PMID: 36496344 PMCID: PMC9992314 DOI: 10.1016/j.jbior.2022.100938] [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: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
The peptidyl-prolyl isomerase Pin1 cooperates with proline-directed kinases and phosphatases to regulate multiple oncogenic pathways. Pin1 specifically recognizes phosphorylated Ser/Thr-Pro motifs in proteins and catalyzes their cis-trans isomerization. The Pin1-catalyzed conformational changes determine the stability, activity, and subcellular localization of numerous protein substrates. We conducted a survey of eukaryotic protein kinases that are regulated by Pin1 and whose Pin1 binding sites have been identified. Our analyses reveal that Pin1 target sites in kinases do not fall exclusively within the intrinsically disordered regions of these enzymes. Rather, they fall into three groups based on their location: (i) within the catalytic kinase domain, (ii) in the C-terminal kinase region, and (iii) in regulatory domains. Some of the kinases downregulated by Pin1 activity are tumor-suppressing, and all kinases upregulated by Pin1 activity are functionally pro-oncogenic. These findings further reinforce the rationale for developing Pin1-specific inhibitors as attractive pharmaceuticals for cancer therapy.
Collapse
Affiliation(s)
- Xiao-Ru Chen
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Tatyana I Igumenova
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
7
|
Ju Z, Thomas TN, Chiu YJ, Yamanouchi S, Yoshida Y, Abe JI, Takahashi A, Wang J, Fujiwara K, Hada M. Adaptation and Changes in Actin Dynamics and Cell Motility as Early Responses of Cultured Mammalian Cells to Altered Gravitational Vector. Int J Mol Sci 2022; 23:6127. [PMID: 35682810 PMCID: PMC9181735 DOI: 10.3390/ijms23116127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
Cultured mammalian cells have been shown to respond to microgravity (μG), but the molecular mechanism is still unknown. The study we report here is focused on molecular and cellular events that occur within a short period of time, which may be related to gravity sensing by cells. Our assumption is that the gravity-sensing mechanism is activated as soon as cells are exposed to any new gravitational environment. To study the molecular events, we exposed cells to simulated μG (SμG) for 15 min, 30 min, 1 h, 2 h, 4 h, and 8 h using a three-dimensional clinostat and made cell lysates, which were then analyzed by reverse phase protein arrays (RPPAs) using a panel of 453 different antibodies. By comparing the RPPA data from cells cultured at 1G with those of cells under SμG, we identified a total of 35 proteomic changes in the SμG samples and found that 20 of these changes took place, mostly transiently, within 30 min. In the 4 h and 8 h samples, there were only two RPPA changes, suggesting that the physiology of these cells is practically indistinguishable from that of cells cultured at 1 G. Among the proteins involved in the early proteomic changes were those that regulate cell motility and cytoskeletal organization. To see whether changes in gravitational environment indeed activate cell motility, we flipped the culture dish upside down (directional change in gravity vector) and studied cell migration and actin cytoskeletal organization. We found that compared with cells grown right-side up, upside-down cells transiently lost stress fibers and rapidly developed lamellipodia, which was supported by increased activity of Ras-related C3 botulinum toxin substrate 1 (Rac1). The upside-down cells also increased their migratory activity. It is possible that these early molecular and cellular events play roles in gravity sensing by mammalian cells. Our study also indicated that these early responses are transient, suggesting that cells appear to adapt physiologically to a new gravitational environment.
Collapse
Affiliation(s)
- Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Tamlyn N. Thomas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.N.T.); (J.-i.A.)
- Aab Cardiovascular Research Institute, University of Rochester Medical School, Rochester, NY 14642, USA;
| | - Yi-Jen Chiu
- Aab Cardiovascular Research Institute, University of Rochester Medical School, Rochester, NY 14642, USA;
| | - Sakuya Yamanouchi
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (S.Y.); (Y.Y.); (A.T.)
| | - Yukari Yoshida
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (S.Y.); (Y.Y.); (A.T.)
| | - Jun-ichi Abe
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.N.T.); (J.-i.A.)
| | - Akihisa Takahashi
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (S.Y.); (Y.Y.); (A.T.)
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Keigi Fujiwara
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.N.T.); (J.-i.A.)
| | - Megumi Hada
- Radiation Institute for Science & Engineering, Prairie View A&M University, Prairie View, TX 77446, USA;
| |
Collapse
|
8
|
Lin Q, Liu M, Yue GGL, Cheung MK, Lai Z, Kwok FHF, Lee JKM, Wang Z, Lau CBS, Tan N. Anti-inflammatory activities of natural cyclopeptide RA-XII in colitis-associated colon cancer mouse model and its effect on gut microbiome. Phytother Res 2022; 36:2641-2659. [PMID: 35537703 DOI: 10.1002/ptr.7482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC), the third most common cancer globally, is associated with intestinal inflammation that leads to poor prognosis. RA-XII, a natural cyclopeptide, has previously been reported to possess anti-tumor activities. Here, the anti-inflammatory activities of RA-XII were investigated in colitis-associated colon cancer mice and a co-culture in vitro model, in which colon cancer cells HCT116 and macrophages RAW264.7 were grown together to mimic the inflammatory microenvironment of CRC. Changes of inflammatory-related molecules and protein expressions in cells were evaluated after RA-XII incubation. Besides, azoxymethane and dextran sulfate sodium-induced colitis-associated colon cancer mice were treated with RA-XII for 24 days, inflammatory parameters and gut microbiome alterations were studied. Our results showed that RA-XII reversed the inflammatory responses of RAW264.7 cells induced by LPS and modulated the protein expressions of AKT, STAT3/p-STAT3, P70S6K, NF-κB and GSK3β and suppressed the expression of LC3A/B in HCT116 cells in co-culture system. RA-XII treatment restored the colitis damage in colon, reduced colon tumors numbers and decreased inflammatory factors (IL-6, IL-10 and TNF-α). The role of RA-XII on regulating gut microbiome was also demonstrated for the first time. In conclusion, our findings provided new scientific evidence for developing RA-XII as a potent anti-inflammatory agent for CRC.
Collapse
Affiliation(s)
- Qianwen Lin
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mingyu Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Grace Gar-Lee Yue
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Man Kit Cheung
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhixing Lai
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Frankie Hin-Fai Kwok
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Julia Kin-Ming Lee
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhe Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Clara Bik-San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ninghua Tan
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
9
|
Loite U, Raam L, Reimann E, Reemann P, Prans E, Traks T, Vasar E, Silm H, Kingo K, Kõks S. The Expression Pattern of Genes Related to Melanogenesis and Endogenous Opioids in Psoriasis. Int J Mol Sci 2021; 22:ijms222313056. [PMID: 34884858 PMCID: PMC8657874 DOI: 10.3390/ijms222313056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
The melanocortin system is a major regulator of stress responses in the skin and is responsible for the induction of melanin synthesis through activation of melanogenesis enzymes. The expression of both melanocortin system genes and melanogenesis enzyme genes is altered in psoriasis, and the focus here was on twelve genes related to the signal transduction between them. Additionally, five endogenous opioid system genes that are involved in cutaneous inflammation were examined. Quantitative real-time-PCR was utilized to measure mRNA expression in punch biopsies from lesional and non-lesional skin of psoriasis patients and from the skin of healthy control subjects. Most of the genes related to melanogenesis were down-regulated in patients (CREB1, MITF, LEF1, USF1, MAPK14, ICAM1, PIK3CB, RPS6KB1, KIT, and ATRN). Conversely, an up-regulation occurred in the case of opioids (PENK, PDYN, and PNOC). The suppression of genes related to melanogenesis is in agreement with the reported reduction in pigmentation signaling in psoriatic skin and potentially results from the pro-inflammatory environment. The increase in endogenous opioids can be associated with their involvement in inflammatory dysregulation in psoriasis.
Collapse
Affiliation(s)
- Ulvi Loite
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
| | - Liisi Raam
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Dermatology Clinic, Tartu University Hospital, 31 Raja, 50417 Tartu, Estonia
| | - Ene Reimann
- Institute of Genomics, University of Tartu, 23b/2 Riia, 51010 Tartu, Estonia;
| | - Paula Reemann
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
| | - Ele Prans
- Department of Anaesthesiology and Intensive Care, Tartu University Hospital, 8 L. Puusepa, 51014 Tartu, Estonia;
| | - Tanel Traks
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Correspondence:
| | - Eero Vasar
- Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia;
| | - Helgi Silm
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Dermatology Clinic, Tartu University Hospital, 31 Raja, 50417 Tartu, Estonia
| | - Külli Kingo
- Department of Dermatology and Venerology, University of Tartu, 31 Raja, 50417 Tartu, Estonia; (U.L.); (L.R.); (P.R.); (H.S.); (K.K.)
- Dermatology Clinic, Tartu University Hospital, 31 Raja, 50417 Tartu, Estonia
| | - Sulev Kõks
- The Perron Institute for Neurological and Translational Science, 8 Verdun St., Nedlands, WA 6009, Australia;
- Centre for Comparative Genomics, Murdoch University, 90 South St., Murdoch, WA 6150, Australia
| |
Collapse
|
10
|
Yaguchi M, Ikeya S, Kozaki A. The activation mechanism of plant S6 kinase (S6K), a substrate of TOR kinase, is different from that of mammalian S6K. FEBS Lett 2019; 594:776-787. [PMID: 31705659 DOI: 10.1002/1873-3468.13661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 11/06/2022]
Abstract
The S6 kinases (S6Ks) are known to be activated by the target of rapamycin through phosphorylation of their hydrophobic motif (HM). However, our previous research showed that the HM site of plant S6Ks is not phosphorylated and is not essential for their activity in yeast cells lacking Ypk3, an ortholog of mammalian S6K. Here, we demonstrate that the HM site of mammalian S6Ks is phosphorylated and is indispensable for their activity in yeast ypk3∆ cells. Furthermore, pseudo-phosphorylation at the HM site of plant S6Ks results in regaining of activity that is lost due to mutation in the conserved phosphorylation sites, namely the T-loop and Turn motif. These results indicate the activation mechanism of plant S6Ks is different from that of mammals.
Collapse
Affiliation(s)
| | - Shun Ikeya
- Department of Biology, Shizuoka University, Japan
| | - Akiko Kozaki
- Department of Biology, Shizuoka University, Japan
| |
Collapse
|
11
|
Murugan AK, Liu R, Xing M. Identification and characterization of two novel oncogenic mTOR mutations. Oncogene 2019; 38:5211-5226. [PMID: 30918329 PMCID: PMC6597304 DOI: 10.1038/s41388-019-0787-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/12/2019] [Accepted: 03/07/2019] [Indexed: 12/29/2022]
Abstract
Mammalian target of rapamycin (mTOR) signaling is often aberrantly activated, particularly when genetically altered, in human cancers. mTOR inhibitors targeting the activated mTOR signaling are highly promising anti-cancer drugs. Knowing the activating genetic change in mTOR can help guide the use of mTOR inhibitors for cancer treatment. This study was conducted to identify and characterize novel oncogenic mTOR mutations that can potentially be therapeutic targets in human cancer. We sequenced 30 exons of the mTOR gene in 12 thyroid cancer cell lines, 3 melanoma cell lines, 20 anaplastic thyroid cancer (ATC) tumors, and 23 melanoma tumors and functionally characterized the identified novel mTOR mutations in vitro and in vivo. We identified a novel point mutation A1256G in ATC cell line and G7076A in melanoma tumor in exon 9 and exon 51 of the mTOR gene, respectively. Over-expression of the corresponding mTOR mutants H419R and G2359E created through induced mutagenesis showed markedly elevated protein kinase activities associated with the activation of mTOR/p70S6K signaling in HEK293T cells. Stable expression of the two mTOR mutants in NIH3T3 cells strongly activated the mTOR/p70S6K signaling pathway and induced morphologic transformation, cell focus formation, anchorage-independent cell growth, and invasion. Inoculation of these mutant-expressing cells in athymic nude mice induced rapid tumor development, showing their driving oncogenicity. We also demonstrated that transfection with the novel mutants conferred cells high sensitivities to the mTOR inhibitor temsirolimus. We speculate that human cancers harboring these mTOR mutations, such as ATC and melanoma, may be effectively treated with inhibitors targeting mTOR.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Rengyun Liu
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Mingzhao Xing
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
12
|
Regulatory mechanisms of miR-145 expression and the importance of its function in cancer metastasis. Biomed Pharmacother 2018; 109:195-207. [PMID: 30396077 DOI: 10.1016/j.biopha.2018.10.037] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs are post-transcriptional mediators of gene expression and regulation, which play influential roles in tumorigenesis and cancer metastasis. The expression of tumor suppressor miR-145 is reduced in various cancer cell lines, containing both solid tumors and blood malignancies. However, the responsible mechanisms of its down-regulation are a complicated network. miR-145 is potentially able to inhbit tumor cell metastasis by targeting of multiple oncogenes, including MUC1, FSCN1, Vimentin, Cadherin, Fibronectin, Metadherin, GOLM1, ARF6, SMAD3, MMP11, Snail1, ZEB1/2, HIF-1α and Rock-1. This distinctive role of miR-145 in the regulation of metastasis-related gene expression may introduce miR-145 as an ideal candidate for controlling of cancer metastasis by miRNA replacement therapy. The present review aims to discuss the current understanding of the different aspects of molecular mechanisms of miR-145 regulation as well as its role in r metastasis regulation.
Collapse
|
13
|
Pinkham C, Ahmed A, Bracci N, Narayanan A, Kehn-Hall K. Host-based processes as therapeutic targets for Rift Valley fever virus. Antiviral Res 2018; 160:64-78. [PMID: 30316916 DOI: 10.1016/j.antiviral.2018.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/27/2018] [Accepted: 10/05/2018] [Indexed: 12/28/2022]
Abstract
Rift Valley fever virus (RVFV) is an enveloped, segmented, negative sense RNA virus that replicates within the host's cytoplasm. To facilitate its replication, RVFV must utilize host cell processes and as such, these processes may serve as potential therapeutic targets. This review summarizes key host cell processes impacted by RVFV infection. Specifically the influence of RVFV on host transcriptional regulation, post-transcriptional regulation, protein half-life and availability, host signal transduction, trafficking and secretory pathways, cytoskeletal modulation, and mitochondrial processes and oxidative stress are discussed. Therapeutics targeted towards host processes that are essential for RVFV to thrive as well as their efficacy and importance to viral pathogenesis are highlighted.
Collapse
Affiliation(s)
- Chelsea Pinkham
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Aslaa Ahmed
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Nicole Bracci
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA.
| |
Collapse
|
14
|
Wojarski J, Żegleń S, Ochman M, Karolak W. Early Sirolimus-Based Immunosuppression is Safe for Lung Transplantation Patients: Retrospective, Single Arm, Exploratory Study. Ann Transplant 2018; 23:598-607. [PMID: 30135417 PMCID: PMC6248054 DOI: 10.12659/aot.908471] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Sirolimus, a mechanistic target of sirolimus inhibitor, is an immunosuppression medication for patients undergoing heart and abdominal transplantation. Sirolimus-based immunosuppression administered de novo post-lung transplantation is associated with bronchial anastomosis healing-related complications. We hypothesized that sirolimus administration within the first postoperative month in selected lung transplant recipients is safe and may be associated with favorable short-term and long-term outcomes due to its anti-proliferative properties and minimal adverse side effects. Material/Methods Thirteen patients (13.3%; mean age, 46.8±11.9 years) received early sirolimus-based immunosuppression along with cyclosporine and prednisone; 10 patients received single-lung transplantation, 3 received double-lung transplantation, and all received induction immunosuppressants. Patients received early sirolimus-based immunosuppression after an uncomplicated postoperative course and detailed bronchoscopic assessment. Results Sirolimus was begun on a mean of 20.6±4.7 days postoperatively (range, 14–32 days). The in-hospital and 30-day mortality rate was 0%. At long-term follow-up, 5 patients died (due to bacterial infection in 4 patients and pneumocystis jiroveci pneumonia in 1 patient). The mean overall survival was 4.4±2.53 (range, 0.8–10.0) years, 1-year survival was 92%, and 5-year survival was 62%. In 4 patients (30.8%), sirolimus was stopped due to infection in 3 patients and re-transplantation in 1 patient. Only one of the 13 patients developed bronchiolitis obliterans syndrome. In patients still taking sirolimus, renal function, systolic blood pressure, and lipid profile were within normal ranges; however, these patients required statin therapy. Conclusions In selected lung transplant recipients, early sirolimus-based immunosuppression is safe and associated with beneficial short-term and long-term outcomes.
Collapse
Affiliation(s)
- Jacek Wojarski
- Department of Cardiac, Vascular, and Endovascular Surgery and Transplantology, Medical University of Silesia in Katowice, Silesian Centre for Heart Diseases, Zabrze, Poland
| | - Sławomir Żegleń
- Department of Cardiac, Vascular, and Endovascular Surgery and Transplantology, Medical University of Silesia in Katowice, Silesian Centre for Heart Diseases, Zabrze, Poland
| | - Marek Ochman
- Department of Cardiac, Vascular, and Endovascular Surgery and Transplantology, Medical University of Silesia in Katowice, Silesian Centre for Heart Diseases, Zabrze, Poland.,Department of Pharmacology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Wojtek Karolak
- Department of Cardiac, Vascular, and Endovascular Surgery and Transplantology, Medical University of Silesia in Katowice, Silesian Centre for Heart Diseases, Zabrze, Poland
| |
Collapse
|
15
|
Zhang J, Guo J, Qin X, Wang B, Zhang L, Wang Y, Gan W, Pandolfi PP, Chen W, Wei W. The p85 isoform of the kinase S6K1 functions as a secreted oncoprotein to facilitate cell migration and tumor growth. Sci Signal 2018; 11:11/523/eaao1052. [PMID: 29588411 DOI: 10.1126/scisignal.aao1052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Cancer cells can remodel surrounding microenvironments to facilitate cell growth, invasion, and migration by secreting proteins that educate surrounding stromal cells. We report that p85S6K1, the longest isoform of S6K (ribosomal protein S6 kinase), but not the shorter isoform p70S6K1 or p56S6K2, was secreted from cancer cells through its HIV TAT-like, N-terminal six-arginine motif. The exogenously produced p85S6K1 protein entered cultured transformed and nontransformed cells to promote or confer malignant behaviors, leading to increased cell growth and migration. When injected into mice, the p85S6K1 protein enhanced the growth of xenografted breast cancer cells and lung metastasis. Hence, our findings reveal a role for p85S6K1 as a secreted oncogenic kinase and provide a mechanism by which cancer cells remodel their microenvironment by transforming the surrounding cells to drive tumorigenesis.
Collapse
Affiliation(s)
- Jianjun Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Xing Qin
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Bin Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Linli Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yingnan Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Wenjian Gan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pier Paolo Pandolfi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wantao Chen
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Research Institute of Stomatology and Shanghai Key Laboratory of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
16
|
In silico binding affinity studies of N-9 substituted 6-(4-(4-propoxyphenyl)piperazin-1-yl)-9H-purine derivatives-Target for P70-S6K1 & PI3K-δ kinases. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2018. [DOI: 10.1016/j.bjbas.2017.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
17
|
Chai X, Sun D, Han Q, Yi L, Wu Y, Liu X. Hypoxia induces pulmonary arterial fibroblast proliferation, migration, differentiation and vascular remodeling via the PI3K/Akt/p70S6K signaling pathway. Int J Mol Med 2018; 41:2461-2472. [PMID: 29436587 PMCID: PMC5846667 DOI: 10.3892/ijmm.2018.3462] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 11/29/2017] [Indexed: 12/20/2022] Open
Abstract
The present study was designed to examine whether hypoxia induces the proliferation, migration and differentiation of pulmonary arterial fibroblasts (PAFs) via the PI3K/Akt/p70S6K signaling pathway. PAFs were subjected to normoxia (21% O2) or hypoxia (1% O2). The proliferation, migration, differentiation and cellular p110α, p-Akt, and p-p70S6K expression levels of the PAFs were examined in vitro. In addition, rats were maintained under hypoxic conditions, and the right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI) and right ventricular weight/body weight ratio (RV/BW) were examined. The expression levels of p110α, p-Akt, p70S6K, fibronectin and α-SMA in the rat pulmonary vessels were also examined. Hypoxia significantly elevated the proliferation, migration and differentiation of rat PAFs. It also strongly elevated the expression of p110α, p-Akt and p-p70S6K in PAFs in vitro. NVP-BEZ235 was revealed to significantly reduce the hypoxia-induced proliferation, migration and differentiation. In vivo experiments demonstrated that hypoxia significantly induced the elevation of RVSP, RVHI, RV/BW, medial thickening, adventitious thickening, and fibronectin and collagen deposition around pulmonary artery walls. The expression of p110α, p-Akt and p70S6K was evident in the pulmonary arteries of the hypoxic rats. NVP-BEZ235 significantly reduced the hypoxia-induced hypoxic pulmonary vascular remodeling, as well as fibronectin and collagen deposition in the pulmonary arteries. Therefore, hypoxia was demonstrated to induce the proliferation, migration and differentiation of PAFs and the hypoxic pulmonary vascular remodeling of rats via the PI3K/Akt/p70S6K signaling pathway.
Collapse
Affiliation(s)
- Xiaoyu Chai
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Dan Sun
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Qian Han
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Liang Yi
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Yanping Wu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| | - Xinmin Liu
- Department of Geriatrics, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
18
|
Kwon NH, Lee MR, Kong J, Park SK, Hwang BJ, Kim BG, Lee ES, Moon HG, Kim S. Transfer-RNA-mediated enhancement of ribosomal proteins S6 kinases signaling for cell proliferation. RNA Biol 2018; 15:635-648. [PMID: 28816616 PMCID: PMC6103689 DOI: 10.1080/15476286.2017.1356563] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While transfer-RNAs (tRNAs) are known to transport amino acids to ribosome, new functions are being unveiled from tRNAs and their fragments beyond protein synthesis. Here we show that phosphorylation of 90-kDa RPS6K (ribosomal proteins S6 kinase) was enhanced by tRNALeu overexpression under amino acids starvation condition. The phosphorylation of 90-kDa RPS6K was decreased by siRNA specific to tRNALeu and was independent to mTOR (mammalian target of rapamycin) signaling. Among the 90-kDa RPS6K family, RSK1 (ribosomal S6 kinase 1) and MSK2 (mitogen-and stress-activated protein kinase 2) were the major kinases phosphorylated by tRNALeu overexpression. Through SILAC (stable isotope labeling by/with amino acids in cell culture) and combined mass spectrometry analysis, we identified EBP1 (ErbB3-binding protein 1) as the tRNALeu-binding protein. We suspected that the overexpression of free tRNALeu would reinforce ErbB2/ErbB3 signaling pathway by disturbing the interaction between ErbB3 and EBP1, resulting in RSK1/MSK2 phosphorylation, improving cell proliferation and resistance to death. Analysis of samples from patients with breast cancer also indicated an association between tRNALeu overexpression and the ErbB2-positive population. Our results suggested a possible link between tRNALeu overexpression and RSK1/MSK2 activation and ErbB2/ErbB3 signaling.
Collapse
Affiliation(s)
- Nam Hoon Kwon
- a Medicinal Bioconvergence Research Center , Seoul National University , Suwon , Gyeonggi , Korea
| | - Mi Ran Lee
- a Medicinal Bioconvergence Research Center , Seoul National University , Suwon , Gyeonggi , Korea
| | - Jiwon Kong
- a Medicinal Bioconvergence Research Center , Seoul National University , Suwon , Gyeonggi , Korea.,b Department of Pharmacy , Seoul National University , Seoul , Korea
| | - Seung Kyun Park
- c Department of Molecular Bioscience , College of Biomedical Science, Kangwon National University , Chuncheon , Kangwon , Korea
| | - Byung Joon Hwang
- c Department of Molecular Bioscience , College of Biomedical Science, Kangwon National University , Chuncheon , Kangwon , Korea
| | - Byung Gyu Kim
- a Medicinal Bioconvergence Research Center , Seoul National University , Suwon , Gyeonggi , Korea
| | - Eun-Shin Lee
- d Department of Surgery , Seoul National University College of Medicine , Seoul , Korea
| | - Hyeong-Gon Moon
- d Department of Surgery , Seoul National University College of Medicine , Seoul , Korea
| | - Sunghoon Kim
- a Medicinal Bioconvergence Research Center , Seoul National University , Suwon , Gyeonggi , Korea.,e Department of Molecular Medicine and Biopharmaceutical Sciences , Graduate School of Convergence Science and Technology, Seoul National University , Suwon , Gyeonggi , Korea
| |
Collapse
|
19
|
Wu D, Cheng J, Sun G, Wu S, Li M, Gao Z, Zhai S, Li P, Su D, Wang X. p70S6K promotes IL-6-induced epithelial-mesenchymal transition and metastasis of head and neck squamous cell carcinoma. Oncotarget 2017; 7:36539-36550. [PMID: 27174914 PMCID: PMC5095019 DOI: 10.18632/oncotarget.9282] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 04/24/2016] [Indexed: 12/14/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the fifth most common cancer worldwide and a common cause of cancer-related death, with a 5-year survival rate of less than 60%. IL-6 has been suggested to play an important role in cancer metastasis, but its mechanism in HNSCC has not been fully clarified. p70S6K has been reported to induce epithelial-mesenchymal transition (EMT) of ovarian cancer, but its role in HNSCC remains unknown. In this study, we found that p70S6K and IL-6 were upregulated in high-metastatic HNSCC cell lines that underwent EMT when compared to paired low-metastatic cell lines. Overexpression of p70S6K promoted EMT and migration of HNSCC cells, while downregulation of p70S6K attenuated IL-6-induced EMT and cell migration. Furthermore, IL-6-induced p70S6K activation was attenuated by inhibitors of the PI3K/Akt/mTOR, MAPK/ERK, and JAK/STAT3 signaling pathways, suggesting that it located downstream of these pathways. These findings suggest that p70S6K promotes IL-6-induced EMT and metastasis of HNSCC. Targeting p70S6K for HNSCC therapy may benefit patients through the inhibition of tumor growth, as well as metastasis.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China.,Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, Jiangsu Province 222000, China
| | - Jie Cheng
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Geng Sun
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Shengjie Wu
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Min Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Zhongyuan Gao
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Sulan Zhai
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Ping Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Dongming Su
- Center for Clinical Pathology and Laboratory, Affiliated Hospital of Yifu, Nanjing Medical University, Nanjing, Jiangsu Province 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Xuerong Wang
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China.,Center for Clinical Pathology and Laboratory, Affiliated Hospital of Yifu, Nanjing Medical University, Nanjing, Jiangsu Province 211166, China.,Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| |
Collapse
|
20
|
Van Ostade X, Dom M, Tjalma W, Van Raemdonck G. Candidate biomarkers in the cervical vaginal fluid for the (self-)diagnosis of cervical precancer. Arch Gynecol Obstet 2017; 297:295-311. [PMID: 29143101 PMCID: PMC5778162 DOI: 10.1007/s00404-017-4587-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/06/2017] [Indexed: 11/29/2022]
Abstract
Purpose Despite improvement in vaccines against human papilloma virus (HPV), the causative agent of cervical cancer, screening women for cervical precancer will remain indispensable in the coming 30–40 years. A simple test that could be performed at home or at a doctor’s practice and that informs the woman whether she is at risk would significantly help make a broader group of patients who aware that they need medical treatment. Cervical vaginal fluid (CVF) is a body fluid that is very well suited for such a test. Methods Narrative review of cervical (pre)cancer candidate biomarkers from cervicovaginal fluid, is based on a detailed review of the literature. We will also discuss the possibilities that these biomarkers create for the development of a self-test or point-of-care test for cervical (pre)cancer. Results Several DNA, DNA methylation, miRNA, and protein biomarkers were identified in the cervical vaginal fluid; however, not all of these biomarkers are suited for development of a simple diagnostic assay. Conclusions Proteins, especially alpha-actinin-4, are most suited for development of a simple assay for cervical (pre)cancer. Accuracy of the test could further be improved by combination of several proteins or by combination with a new type of biomarker, e.g., originating from the cervicovaginal microbiome or metabolome.
Collapse
Affiliation(s)
- Xaveer Van Ostade
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES), University of Antwerp, Wilrijk, Belgium. .,Centre for Proteomics (CfP), University of Antwerp, Wilrijk, Belgium.
| | - Martin Dom
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES), University of Antwerp, Wilrijk, Belgium.,Centre for Proteomics (CfP), University of Antwerp, Wilrijk, Belgium
| | - Wiebren Tjalma
- Gynecological Oncology Unit, Department of Obstetrics and Gynecology, Multidisciplinary Breast Clinic, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Geert Van Raemdonck
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES), University of Antwerp, Wilrijk, Belgium.,Centre for Proteomics (CfP), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
21
|
LOC285629 regulates cell proliferation and motility in colorectal cancer cells. Clin Transl Oncol 2017; 20:775-784. [DOI: 10.1007/s12094-017-1788-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/23/2017] [Indexed: 10/18/2022]
|
22
|
Mohl BP, Emmott E, Roy P. Phosphoproteomic Analysis Reveals the Importance of Kinase Regulation During Orbivirus Infection. Mol Cell Proteomics 2017; 16:1990-2005. [PMID: 28851738 PMCID: PMC5672004 DOI: 10.1074/mcp.m117.067355] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 08/08/2017] [Indexed: 01/03/2023] Open
Abstract
Bluetongue virus (BTV) causes infections in wild and domesticated ruminants with high morbidity and mortality and is responsible for significant economic losses in both developing and developed countries. BTV serves as a model for the study of other members of the Orbivirus genus. Previously, the importance of casein kinase 2 for BTV replication was demonstrated. To identify intracellular signaling pathways and novel host-cell kinases involved during BTV infection, the phosphoproteome of BTV infected cells was analyzed. Over 1000 phosphosites were identified using mass spectrometry, which were then used to determine the corresponding kinases involved during BTV infection. This analysis yielded protein kinase A (PKA) as a novel kinase activated during BTV infection. Subsequently, the importance of PKA for BTV infection was validated using a PKA inhibitor and activator. Our data confirmed that PKA was essential for efficient viral growth. Further, we showed that PKA is also required for infection of equid cells by African horse sickness virus, another member of the Orbivirus genus. Thus, despite their preference in specific host species, orbiviruses may utilize the same host signaling pathways during their replication.
Collapse
Affiliation(s)
- Bjorn-Patrick Mohl
- From the ‡Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Edward Emmott
- §University of Cambridge, Division of Virology, Department of Pathology, Lab block level 5, Box 237, Addenbrookes Hospital, Cambridge, UK
| | - Polly Roy
- From the ‡Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK;
| |
Collapse
|
23
|
Kim H, Kim DH, Jeong B, Kim JH, Lee SR, Sonn JK. Blebbistatin induces chondrogenesis of single mesenchymal cells via PI3K/PDK1/mTOR/p70S6K pathway. Biologia (Bratisl) 2017. [DOI: 10.1515/biolog-2017-0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
24
|
Britain CM, Dorsett KA, Bellis SL. The Glycosyltransferase ST6Gal-I Protects Tumor Cells against Serum Growth Factor Withdrawal by Enhancing Survival Signaling and Proliferative Potential. J Biol Chem 2017; 292:4663-4673. [PMID: 28154177 DOI: 10.1074/jbc.m116.763862] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/24/2017] [Indexed: 12/27/2022] Open
Abstract
A hallmark of cancer cells is the ability to survive and proliferate when challenged with stressors such as growth factor insufficiency. In this study, we report a novel glycosylation-dependent mechanism that protects tumor cells from serum growth factor withdrawal. Our results suggest that the β-galactoside α-2,6-sialyltransferase 1 (ST6Gal-I) sialyltransferase, which is up-regulated in numerous cancers, promotes the survival of serum-starved cells. Using ovarian and pancreatic cancer cell models with ST6Gal-I overexpression or knockdown, we find that serum-starved cells with high ST6Gal-I levels exhibit increased activation of prosurvival signaling molecules, including pAkt, p-p70S6K, and pNFκB. Correspondingly, ST6Gal-I activity augments the expression of tumor-promoting pNFκB transcriptional targets such as IL-6, IL-8, and the apoptosis inhibitor cIAP2. ST6Gal-I also potentiates expression of the cell cycle regulator cyclin D2, leading to increased phosphorylation and inactivation of the cell cycle inhibitor pRb. Consistent with these results, serum-starved cells with high ST6Gal-I expression maintain a greater number of S phase cells compared with low ST6Gal-I expressors, reflecting enhanced proliferation. Finally, selective enrichment in clonal variants with high ST6Gal-I expression is observed upon prolonged serum deprivation, supporting the concept that ST6Gal-I confers a survival advantage. Collectively, these results implicate a functional role for ST6Gal-I in fostering tumor cell survival within the serum-depleted tumor microenvironment.
Collapse
Affiliation(s)
- Colleen M Britain
- From the Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Kaitlyn A Dorsett
- From the Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Susan L Bellis
- From the Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
25
|
Lou K, Yang M, Duan E, Zhao J, Yu C, Zhang R, Zhang L, Zhang M, Xiao Z, Hu W, He Z. Rosmarinic acid stimulates liver regeneration through the mTOR pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1574-1582. [PMID: 27823621 DOI: 10.1016/j.phymed.2016.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 09/14/2016] [Accepted: 09/28/2016] [Indexed: 05/27/2023]
Abstract
BACKGROUND Rosemary (Rosmarinus offcinsalis L) has a liver protection function under various conditions of liver damage. Rosmarinic acid, one of the pharmacological constituents of rosemary, exhibited protective effects against organ injury, including acute liver injury. HYPOTHESIS We hypothesize that RA stimulates liver regeneration. STUDY DESIGN In the present study, we investigated the effects and mechanism of RA administration on liver regeneration using partial hepatectomy (PH), a well-validated liver regeneration model in mice. METHODS We use a 2/3 partial hepatectomy (PH) model to induce liver regeneration. RA was administered prior to and simultaneously with PH. The regeneration process was estimated by the index of the liver to body weight (ILBW) and the expression of proliferating cell nuclear antigen (PCNA) and liver transaminases. RESULTS The administration of rosmarinic acid stimulated hepatocyte proliferation based on activation of the mTOR/S6K pathway. Rosmarinic acid treatment also rescued impaired liver function due to PH. CONCLUSION These data demonstrate that RA is potentially useful to promote liver regeneration.
Collapse
Affiliation(s)
- Kaihan Lou
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Min Yang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Erdan Duan
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Jiahui Zhao
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Cong Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Rongping Zhang
- Biomedical Engineering Research Centre, Kunming Medical University, Kunming 650500, China
| | - Lanchun Zhang
- Biomedical Engineering Research Centre, Kunming Medical University, Kunming 650500, China
| | - Ming Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Zhicheng Xiao
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China; Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne 3800, Australia
| | - Weiyan Hu
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China.
| | - Zhiyong He
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China; Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne 3800, Australia.
| |
Collapse
|
26
|
Amaral CL, Freitas LB, Tamura RE, Tavares MR, Pavan ICB, Bajgelman MC, Simabuco FM. S6Ks isoforms contribute to viability, migration, docetaxel resistance and tumor formation of prostate cancer cells. BMC Cancer 2016; 16:602. [PMID: 27491285 PMCID: PMC4974797 DOI: 10.1186/s12885-016-2629-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background The S6 Kinase (S6K) proteins are some of the main downstream effectors of the mammalian Target Of Rapamycin (mTOR) and act as key regulators of protein synthesis and cell growth. S6K is overexpressed in a variety of human tumors and is correlated to poor prognosis in prostate cancer. Due to the current urgency to identify factors involved in prostate cancer progression, we aimed to reveal the cellular functions of three S6K isoforms–p70-S6K1, p85-S6K1 and p54-S6K2–in prostate cancer, as well as their potential as therapeutic targets. Methods In this study we performed S6K knockdown and overexpression and investigated its role in prostate cancer cell proliferation, colony formation, viability, migration and resistance to docetaxel treatment. In addition, we measured tumor growth in Nude mice injected with PC3 cells overexpressing S6K isoforms and tested the efficacy of a new available S6K1 inhibitor in vitro. Results S6Ks overexpression enhanced PC3-luc cell line viability, migration, resistance to docetaxel and tumor formation in Nude mice. Only S6K2 knockdown rendered prostate cancer cells more sensitive to docetaxel. S6K1 inhibitor PF-4708671 was particularly effective for reducing migration and proliferation of PC3 cell line. Conclusions These findings demonstrate that S6Ks play an important role in prostate cancer progression, enhancing cell viability, migration and chemotherapy resistance, and place both S6K1 and S6K2 as a potential targets in advanced prostate cancer. We also provide evidence that S6K1 inhibitor PF-4708671 may be considered as a potential drug for prostate cancer treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2629-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camila L Amaral
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Lidia B Freitas
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Rodrigo E Tamura
- Viral Vector Laboratory, Center for Translational Investigation in Oncology/LIM24, Cancer Institute of São Paulo, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mariana R Tavares
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Isadora C B Pavan
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Marcio C Bajgelman
- Brazilian Biosciences National Laboratory, Brazilian National Center for Research in Energy and Materials, Campinas, São Paulo, Brazil
| | - Fernando M Simabuco
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil.
| |
Collapse
|
27
|
Oehninger L, Spreckelmeyer S, Holenya P, Meier SM, Can S, Alborzinia H, Schur J, Keppler BK, Wölfl S, Ott I. Rhodium(I) N-Heterocyclic Carbene Bioorganometallics as in Vitro Antiproliferative Agents with Distinct Effects on Cellular Signaling. J Med Chem 2015; 58:9591-600. [PMID: 26595649 DOI: 10.1021/acs.jmedchem.5b01159] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Organometallics with N-heterocyclic carbene (NHC) ligands have triggered major interest in inorganic medicinal chemistry. Complexes of the type Rh(I)(NHC)(COD)X (where X is Cl or I, COD is cyclooctadiene, and NHC is a dimethylbenzimidazolylidene) represent a promising type of new metallodrugs that have been explored by advanced biomedical methods only recently. In this work, we have synthesized and characterized several complexes of this type. As observed by mass spectrometry, these complexes remained stable over at least 3 h in aqueous solution, after which hydrolysis of the halido ligands occurred and release of the NHC ligand was evident. Effects against mitochondria and general cell tumor metabolism were noted at higher concentrations, whereas phosphorylation of HSP27, p38, ERK1/2, FAK, and p70S6K was induced substantially already at lower exposure levels. Regarding the antiproliferative activity in tumor cells, a clear preference for iodido over chlorido secondary ligands was noted, as well as effects of the substituents of the NHC ligand.
Collapse
Affiliation(s)
- Luciano Oehninger
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig , Beethovenstraße 55, D-38106 Braunschweig, Germany
| | - Sarah Spreckelmeyer
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig , Beethovenstraße 55, D-38106 Braunschweig, Germany
| | - Pavlo Holenya
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg , Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Samuel M Meier
- Department of Analytical Chemistry, University of Vienna , Waehringer Straße 38, 1090 Vienna, Austria
| | - Suzan Can
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg , Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Hamed Alborzinia
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg , Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Julia Schur
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig , Beethovenstraße 55, D-38106 Braunschweig, Germany
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna , Waehringer Straße 42, 1090 Vienna, Austria
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-Universität Heidelberg , Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig , Beethovenstraße 55, D-38106 Braunschweig, Germany
| |
Collapse
|
28
|
Bahrami-B F, Ataie-Kachoie P, Pourgholami MH, Morris DL. p70 Ribosomal protein S6 kinase (Rps6kb1): an update. J Clin Pathol 2014; 67:1019-25. [PMID: 25100792 DOI: 10.1136/jclinpath-2014-202560] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Rps6kb1 gene encodes the 70 kDa ribosomal protein S6 kinase (p70S6K), which is a serine/threonine kinase regulated by phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway. p70S6K plays a crucial role in controlling cell cycle, growth and survival. The PI3K/mTOR signalling pathway is one of the major mechanisms for controlling cell survival, proliferation and metabolism and is the central regulator of translation of some components of protein synthesis system. Upon activation, this kinase phosphorylates S6 protein of ribosomal subunit 40S resulting in selective translation of unique family of mRNAs that contain oligopyrimidine tract on 5' transcriptional site (5'TOP). 5'TOP mRNAs are coding the components of translational apparatus including ribosomal proteins and elongation factors. Due to the role of p70S6K in protein synthesis and also its involvement in a variety of human diseases ranging from diabetes and obesity to cancer, p70S6K is now being considered as a new therapeutic target for drug development. Furthermore, p70S6K acts as a biomarker for response to immunosuppressant as well as anticancer effects of inhibitors of the mTOR. Because of the narrow therapeutic index of mTOR inhibitors, drug monitoring is essential, and this is usually done by measuring blood drug levels, therapeutic response and drug-induced adverse effects. Recent studies have suggested that plasma p70S6K is a reliable index for the monitoring of patient response to mTOR inhibitors. Therefore, a better understanding of p70S6K and its role in various pathological conditions could enable the development of strategies to aid diagnosis, prognosis and treatment schedules.
Collapse
Affiliation(s)
- Farnaz Bahrami-B
- Cancer research laboratories, Department of Surgery, St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | | | | | - David L Morris
- Cancer research laboratories, Department of Surgery, St George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| |
Collapse
|
29
|
JO JEONGRANG, PARK YUKYOUNG, JANG BYEONGCHURL. Short-term treatment with glucosamine hydrochloride specifically downregulates hypoxia-inducible factor-1α at the protein level in YD-8 human tongue cancer cells. Int J Oncol 2014; 44:1699-706. [DOI: 10.3892/ijo.2014.2336] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/04/2014] [Indexed: 11/06/2022] Open
|
30
|
Liu W, Liu K, Tao H, Chen C, Zhang JH, Sun X. Hyperoxia preconditioning: the next frontier in neurology? Neurol Res 2013; 34:415-21. [DOI: 10.1179/1743132812y.0000000034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Wenwu Liu
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Kan Liu
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Hengyi Tao
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| | - Chunhua Chen
- Department of Anatomy and EmbryologyPeking University Health Science Center, Beijing, China
| | - John H Zhang
- Department of AnesthesiologyLoma Linda Medical Center, Loma Linda, CA, USA
| | - Xuejun Sun
- Department of Diving MedicineThe Second Military Medical University, Shanghai, China
| |
Collapse
|
31
|
Koh PO. Ferulic acid attenuates focal cerebral ischemia-induced decreases in p70S6 kinase and S6 phosphorylation. Neurosci Lett 2013; 555:7-11. [DOI: 10.1016/j.neulet.2013.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 08/29/2013] [Accepted: 09/01/2013] [Indexed: 10/26/2022]
|
32
|
Murugan AK, Alzahrani A, Xing M. Mutations in critical domains confer the human mTOR gene strong tumorigenicity. J Biol Chem 2013; 288:6511-21. [PMID: 23322780 DOI: 10.1074/jbc.m112.399485] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that regulates cell growth, proliferation, and survival. mTOR is frequently activated in human cancers and is a commonly sought anticancer therapeutic target. However, whether the human mTOR gene itself is a proto-oncogene possessing tumorigenicity has not been firmly established. To answer this question, we mutated evolutionarily conserved amino acids, generated eight mutants in the HEAT repeats (M938T) and the FAT (W1456R and G1479N) and kinase (P2273S, V2284M, V2291I, T2294I, and E2288K) domains of mTOR, and studied their oncogenicity. On transient expression in HEK293T cells, these mTOR mutants displayed elevated protein kinase activities accompanied by activated mTOR/p70S6K signaling at varying levels, demonstrating the gain of function of the mTOR gene with these mutations. We selected P2273S and E2288K, the two most catalytically active mutants, to further examine their oncogenicity and tumorigenicity. Stable expression of the two mTOR mutants in NIH3T3 cells strongly activated mTOR/p70S6K signaling, induced cell transformation and invasion, and remarkably, caused rapid tumor formation and growth in athymic nude mice after subcutaneous inoculation of the transfected cells. This study confirms the oncogenic potential of mTOR suggested previously and demonstrates for the first time its tumorigenicity. Thus, beyond the pivotal position of mTOR to relay the oncogenic signals from the upstream phosphatidylinositol 3-kinase/Akt pathway in human cancer, mTOR is capable potentially of playing a direct role in human tumorigenesis if mutated. These results also further support the conclusion that mTOR is a major therapeutic target in human cancers.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology and Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | |
Collapse
|
33
|
Lo T, Tsai CF, Shih YRV, Wang YT, Lu SC, Sung TY, Hsu WL, Chen YJ, Lee OK. Phosphoproteomic Analysis of Human Mesenchymal Stromal Cells during Osteogenic Differentiation. J Proteome Res 2011; 11:586-98. [DOI: 10.1021/pr200868p] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ting Lo
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Chia-Feng Tsai
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yu-Ru V. Shih
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yi-Ting Wang
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Sheng-Chieh Lu
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Ting-Yi Sung
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Wen-Lian Hsu
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Yu-Ju Chen
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Oscar K. Lee
- Department of Medical Research and Education and ‡Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and ∥Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Chemistry and Genomics Research Center, ¶Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Institute of Chemistry, and #Institute of Information Science, Academia Sinica, Taipei, Taiwan
- Department of Chemistry and ○Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Xu M, Cao F, Liu L, Zhang B, Wang Y, Dong H, Cui Y, Dong M, Xu D, Liu Y, Zhao P, Niu W, Li Z. Tanshinone IIA-induced attenuation of lung injury in endotoxemic mice is associated with reduction of hypoxia-inducible factor 1α expression. Am J Respir Cell Mol Biol 2011; 45:1028-1035. [PMID: 21622293 DOI: 10.1165/rcmb.2011-0113oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Inhibiting hypoxia-inducible factor (HIF)-1α activity has been proposed as a novel therapeutic target in LPS-induced sepsis syndrome. We have reported that tanshinone IIA (TIIA) can reduce LPS-induced lethality and lung injury in mice, but the precise mechanisms have not been fully described. Therefore, the present study investigated whether the protective effect of TIIA was related to the inhibition of LPS-induced HIF-1α expression and what mechanisms accounted for it. This study showed that TIIA pretreatment improved LPS-induced biochemical and cellular changes and reduced the production of inflammatory cytokines. Pretreatment with TIIA decreased LPS-induced HIF-1α expression in vivo and in vitro. TIIA did not affect the LPS-induced HIF-1α mRNA level but inhibited HIF-1α protein translation by the inhibition of the PI3K/AKT and MAPK pathways and related protein translational regulators, such as p70S6K1, S6 ribosomal protein, 4E-BP1, and eIF4E, and promoted HIF-1α protein degradation via the proteasomal pathway in LPS-stimulated macrophages. These observations partially explain the antiinflammatory effects of TIIA, which provides scientific basis for its application for the treatment of acute lung injury/acute respiratory distress syndrome or sepsis.
Collapse
Affiliation(s)
- Min Xu
- Department of Pathology, Xijing Hospital, Xi’an, PR China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Aslan JE, Tormoen GW, Loren CP, Pang J, McCarty OJT. S6K1 and mTOR regulate Rac1-driven platelet activation and aggregation. Blood 2011; 118:3129-36. [PMID: 21757621 PMCID: PMC3175787 DOI: 10.1182/blood-2011-02-331579] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 06/24/2011] [Indexed: 01/20/2023] Open
Abstract
Platelet activation and thrombus formation are under the control of signaling systems that integrate cellular homeostasis with cytoskeletal dynamics. Here, we identify a role for the ribosome protein S6 kinase (S6K1) and its upstream regulator mTOR in the control of platelet activation and aggregate formation under shear flow. Platelet engagement of fibrinogen initiated a signaling cascade that triggered the activation of S6K1 and Rac1. Fibrinogen-induced S6K1 activation was abolished by inhibitors of Src kinases, but not Rac1 inhibitors, demonstrating that S6K1 acts upstream of Rac1. S6K1 and Rac1 interacted in a protein complex with the Rac1 GEF TIAM1 and colocalized with actin at the platelet lamellipodial edge, suggesting that S6K1 and Rac1 work together to drive platelet spreading. Pharmacologic inhibitors of mTOR and S6K1 blocked Rac1 activation and prevented platelet spreading on fibrinogen, but had no effect on Src or FAK kinase activation. mTOR inhibitors dramatically reduced collagen-induced platelet aggregation and promoted the destabilization of platelet aggregates formed under shear flow conditions. Together, these results reveal novel roles for S6K1 and mTOR in the regulation of Rac1 activity and provide insights into the relationship between the pharmacology of the mTOR system and the molecular mechanisms of platelet activation.
Collapse
Affiliation(s)
- Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| | | | | | | | | |
Collapse
|
36
|
Setshedi M, Longato L, Petersen DR, Ronis M, Chen WC, Wands JR, de la Monte SM. Limited therapeutic effect of N-acetylcysteine on hepatic insulin resistance in an experimental model of alcohol-induced steatohepatitis. Alcohol Clin Exp Res 2011; 35:2139-51. [PMID: 21790669 DOI: 10.1111/j.1530-0277.2011.01569.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Alcohol-related steatohepatitis is associated with increased oxidative stress, DNA damage, lipotoxicity, and insulin resistance in liver. As inflammation and oxidative stress can promote insulin resistance, effective treatment with antioxidants, for example, N-acetylcysteine (NAC), may restore ethanol-impaired insulin signaling in the liver. METHODS Adult male Sprague-Dawley rats were fed for 130 days with liquid diets containing 0 or 37% ethanol by caloric content, and simultaneously treated with vehicle or NAC. Chow-fed controls were studied in parallel. Liver tissues were used for histopathology, cytokine activation, and insulin/IGF-1 signaling assays. RESULTS We observed significant positive trends of increasing severity of steatohepatitis (p = 0.016) with accumulation of neutral lipid (p = 0.0002) and triglycerides (p = 0.0004) from chow to control, to the ethanol diet, irrespective of NAC treatment. In ethanol-fed rats, NAC reduced inflammation, converted the steatosis from a predominantly microvesicular to a mainly macrovesicular histological pattern, reduced pro-inflammatory cytokine gene expression, ceramide load, and acid sphingomyelinase activity, and increased expression of IGF-1 receptor and IGF-2 in liver. However, NAC did not abrogate ethanol-mediated impairments in signaling through insulin/IGF-1 receptors, IRS-1, Akt, GSK-3β, or p70S6K, nor did it significantly reduce pro-ceramide or GM3 ganglioside gene expression in liver. CONCLUSIONS Antioxidant treatments reduce the severity of chronic alcohol-related steatohepatitis, possibly because of the decreased expression of inflammatory mediators and ceramide accumulation, but they do not restore insulin/IGF-1 signaling in liver, most likely due to persistent elevation of GM3 synthase expression. Effective treatment of alcohol-related steatohepatitis most likely requires dual targeting of oxidative stress and insulin/IGF resistance.
Collapse
Affiliation(s)
- Mashiko Setshedi
- Department of Medicine, Brown University, Providence, Rhode Island, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Ohara T, Takaoka M, Toyooka S, Tomono Y, Nishikawa T, Shirakawa Y, Yamatsuji T, Tanaka N, Fujiwara T, Naomoto Y. Inhibition of mTOR by temsirolimus contributes to prolonged survival of mice with pleural dissemination of non-small-cell lung cancer cells. Cancer Sci 2011; 102:1344-9. [PMID: 21521416 DOI: 10.1111/j.1349-7006.2011.01967.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Temsirolimus (CCI-779), a recently synthesized analogue of rapamycin, specifically inhibits mTOR and has been approved for clinical use in renal cell carcinoma. Recent reports have indicated the growth inhibitory effect of temsirolimus in some cancers including non-small-cell lung carcinoma (NSCLC). In this study, we aimed to explore the potential therapeutic use of temsirolimus as a treatment for NSCLC. Using cultured NSCLC cells (A549, H1299, and H358), we determined the effect of temsirolimus on cell proliferation and its antitumor effects on subcutaneous tumors, as well as its contribution to the survival of mice having pleural dissemination of cancer cells, mimicking advanced NSCLC. Temsirolimus suppressed proliferation of NSCLC cells in a dose-dependent manner, with an IC(50) of <1 nM. Western blot analysis revealed that temsirolimus treatment specifically inhibited the phosphorylation of mTOR and its downstream effectors in 1 h, accompanied by an increased cell population in the G(0) /G(1) phase, but according to flow cytometry, the cell population did not increase in the sub-G(0) phase. When NSCLC subcutaneous tumor-bearing mice were treated with temsirolimus, tumor volume was significantly reduced (tumor volume on day 35: vehicle vs temsirolimus = 1239 vs 698 cm(3) ; P < 0.05). Furthermore, prolonged survival was observed in pleural disseminated tumor-bearing mice with temsirolimus treatment (median survival: vehicle vs temsirolimus = 53.5 vs 72.5 days; P < 0.05). These results suggest that temsirolimus could be useful for NSCLC treatment, due to its antiproliferative effect, and could be a potential treatment for advanced NSCLC, giving prolonged survival.
Collapse
Affiliation(s)
- Toshiaki Ohara
- Department of Gastroenterological Surgery, Transplant, and Surgical Oncology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Thrombin Preconditioning Attenuates Iron-Induced Neuronal Death. INTRACEREBRAL HEMORRHAGE RESEARCH 2011; 111:259-63. [DOI: 10.1007/978-3-7091-0693-8_43] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
39
|
Song S, Hu H, Hua Y, Wang J, Xi G. Thrombin preconditioning reduces iron-induced brain swelling and brain atrophy. ACTA NEUROCHIRURGICA. SUPPLEMENT 2011; 111:219-23. [PMID: 21725759 DOI: 10.1007/978-3-7091-0693-8_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Cerebral preconditioning with a low dose of thrombin attenuates brain edema induced by intracerebral hemorrhage (ICH), a large dose of thrombin or iron. This study examined whether or not thrombin preconditioning (TPC) reduces neuronal death and brain atrophy caused by iron. The right hippocampus of rats was pretreated with or without thrombin, and iron was then injected into the same location 3 days later. Rats were killed at 1 day or 7 days after iron injection, and the brains were used for histology. We found that TPC reduced neuronal death and brain swelling in the hippocampus 1 day after iron injection, and hippocampal atrophy 7 days later. Western blots showed that thrombin activates p44/42 mitogen-activated protein kinase (p44/42 MAPK) and 70-kDa ribosomal protein S6 kinase (p70 S6K). Our results indicate that TPC reduction of iron-induced neuronal death may be through the p44/42 MAPK /p70 S6K signal transduction pathway.
Collapse
Affiliation(s)
- Shuijiang Song
- The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | | | | | | | | |
Collapse
|
40
|
Karyala P, Namsa ND, Chilakalapudi DR. Translational up-regulation and high-level protein expression from plasmid vectors by mTOR activation via different pathways in PC3 and 293T cells. PLoS One 2010; 5:e14408. [PMID: 21203441 PMCID: PMC3010991 DOI: 10.1371/journal.pone.0014408] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 11/16/2010] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Though 293T cells are widely used for expression of proteins from transfected plasmid vectors, the molecular basis for the high-level expression is yet to be understood. We recently identified the prostate carcinoma cell line PC3 to be as efficient as 293T in protein expression. This study was undertaken to decipher the molecular basis of high-level expression in these two cell lines. METHODOLOGY/PRINCIPAL FINDINGS In a survey of different cell lines for efficient expression of platelet-derived growth factor-B (PDGF-B), β-galactosidase (β-gal) and green fluorescent protein (GFP) from plasmid vectors, PC3 was found to express at 5-50-fold higher levels compared to the bone metastatic prostate carcinoma cell line PC3BM and many other cell lines. Further, the efficiency of transfection and level of expression of the reporters in PC3 were comparable to that in 293T. Comparative analyses revealed that the high level expression of the reporters in the two cell lines was due to increased translational efficiency. While phosphatidic acid (PA)-mediated activation of mTOR, as revealed by drastic reduction in reporter expression by n-butanol, primarily contributed to the high level expression in PC3, multiple pathways involving PA, PI3K/Akt and ERK1/2 appear to contribute to the abundant reporter expression in 293T. Thus the extent of translational up-regulation attained through the concerted activation of mTOR by multiple pathways in 293T could be achieved through its activation primarily by the PA pathway in PC3. CONCLUSIONS/SIGNIFICANCE Our studies reveal that the high-level expression of proteins from plasmid vectors is effected by translational up-regulation through mTOR activation via different signaling pathways in the two cell lines and that PC3 is as efficient as 293T for recombinant protein expression. Further, PC3 offers an advantage in that the level of expression of the protein can be regulated by simple addition of n-butanol to the culture medium.
Collapse
Affiliation(s)
- Prashanthi Karyala
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Nima D. Namsa
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Durga Rao Chilakalapudi
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
- * E-mail:
| |
Collapse
|
41
|
Hu H, Yamashita S, Hua Y, Keep RF, Liu W, Xi G. Thrombin-induced neuronal protection: role of the mitogen activated protein kinase/ribosomal protein S6 kinase pathway. Brain Res 2010; 1361:93-101. [PMID: 20846511 PMCID: PMC2963703 DOI: 10.1016/j.brainres.2010.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 09/02/2010] [Accepted: 09/03/2010] [Indexed: 10/19/2022]
Abstract
Our previous studies have found that intracerebral pretreatment with a low dose of thrombin (thrombin preconditioning, TPC) reduces infarct volume and attenuates brain edema after focal cerebral ischemia. In this study, we examined whether TPC protects against the neuronal death induced by oxygen glucose deprivation (OGD), and whether the protection is through thrombin receptors and the p44/42 mitogen activated protein kinases (MAPK)/ribosomal protein S6 kinases (p70 S6K) pathway. Expression of protease-activated receptors (PARs) mRNA was detected in cultured primary rat neurons and thrombin upregulated PAR-1 and PAR-4 mRNA expression. TPC reduced OGD-induced neuronal death (e.g. dead cells: 52.5 ± 5.4% vs. 72.3 ± 7.2% in the control group, n=6, p<0.01). Agonists of PAR-1 and PAR-4 mimicked the effects of thrombin and reduced OGD-induced neuronal death. Pretreatment with thrombin or PAR agonists induced the upregulation of activated p44/42 MAPK and p70S6K (Thr 421/Ser 424). PD98059, an inhibitor of p44/42 MAPK kinase, blocked thrombin-induced upregulation of activated p44/42 MAPK and p70S6K. It also reduced TPC-induced neuronal protection (e.g. dead cells: 68.2 ± 5.2% vs. 56.9 ± 4.6% in vehicle+TPC group, n=6, p<0.05). These results suggest that TPC-induced ischemic tolerance is through activation of thrombin receptors and the p44/42 MAPK/p70S6K pathway.
Collapse
Affiliation(s)
- Haitao Hu
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | |
Collapse
|
42
|
Frondorf K, Henkels KM, Frohman MA, Gomez-Cambronero J. Phosphatidic acid is a leukocyte chemoattractant that acts through S6 kinase signaling. J Biol Chem 2010; 285:15837-47. [PMID: 20304930 DOI: 10.1074/jbc.m109.070524] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Phosphatidic acid (PA) is a pleiotropic lipid second messenger in mammalian cells. We report here that extracellular PA acts as a leukocyte chemoattractant, as membrane-soluble dioleoyl-PA (DOPA) elicits actin polymerization and chemotaxis of human neutrophils and differentiated proleukemic HL-60 cells. We show that the mechanism for this involves the S6 kinase (S6K) signaling enzyme. Chemotaxis was inhibited >90% by the S6K inhibitors rapamycin and bisindolylmaleimide and by S6K1 silencing using double-stranded RNA. However, it was only moderately ( approximately 30%) inhibited by mTOR siRNA, indicating the presence of an mTOR-independent mechanism for S6K. Exogenous PA led to robust time- and dose-dependent increases in S6K enzymatic activity and Thr(421)/Ser(424) phosphorylation, further supporting a PA/S6K connection. We also investigated whether intracellular PA production affects cell migration. Overexpression of phospholipase D2 (PLD2) and, to a lesser extent, PLD1, resulted in elevation of both S6K activity and chemokinesis, whereas PLD silencing was inhibitory. Because the lipase-inactive PLD2 mutants K444R and K758R neither activated S6K nor induced chemotaxis, intracellular PA is needed for this form of cell migration. Lastly, we demonstrated a connection between extracellular and intracellular PA. Using an enhanced green fluorescent protein-derived PA sensor (pEGFP-Spo20PABD), we showed that exogenous PA or PA generated in situ by bacterial (Streptomyces chromofuscus) PLD enters the cell and accumulates in vesicle-like cytoplasmic structures. In summary, we report the discovery of PA as a leukocyte chemoattractant via cell entry and activation of S6K to mediate the cytoskeletal actin polymerization and leukocyte chemotaxis required for the immune function of these cells.
Collapse
Affiliation(s)
- Kathleen Frondorf
- Department of Biochemistry and Molecular Biology, Wright State University School of Medicine, Dayton, Ohio 45435, USA
| | | | | | | |
Collapse
|
43
|
Hou P, Bojdani E, Xing M. Induction of thyroid gene expression and radioiodine uptake in thyroid cancer cells by targeting major signaling pathways. J Clin Endocrinol Metab 2010; 95:820-8. [PMID: 20008023 PMCID: PMC2840852 DOI: 10.1210/jc.2009-1888] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Radioiodine ablation is commonly used to treat thyroid cancer, but a major challenge is often the loss of radioiodine avidity of the cancer caused by aberrant silencing of iodide-handling genes. OBJECTIVES This study was conducted to test the therapeutic potential of targeting the aberrantly activated MAPK and PI3K/Akt/mTOR pathways and histone deacetylase to restore radioiodine avidity in thyroid cancer cells. EXPERIMENTAL DESIGN We tested the effects of specific inhibitors targeting these pathways/molecules that had established clinical applicability, including the MAPK kinase inhibitor RDEA119, mTOR inhibitor temsirolimus, Akt inhibitor perifosine, and histone deacetylase inhibitor SAHA, individually or in combinations, on the expression of iodide-handling genes and radioiodide uptake in a large panel of thyroid cancer cell lines. RESULTS The expression of a large number of iodide-handling genes could be restored, particularly the sodium/iodide symporter, TSH receptor, and thyroperoxidase, by treating cells with these inhibitors. The effect was particularly robust and synergistic when combinations of inhibitors containing SAHA were used. Robust expression of sodium/iodide symporter in the cell membrane, which plays the most important role in iodide uptake in thyroid cells, was confirmed by immunofluorescent microscopy. Radioiodide uptake by cells was correspondingly induced under these conditions. Thyroid gene expression and radioiodide uptake could both be further enhanced by TSH. CONCLUSIONS Targeting major signaling pathways could restore thyroid gene expression and radioiodide uptake in thyroid cancer cells. Further studies are warranted to test this therapeutic potential in restoring radioiodine avidity of thyroid cancer cells for effective ablation treatment.
Collapse
Affiliation(s)
- Peng Hou
- Division of Endocrinology and Metabolism, The Johns Hopkins University School of Medicine, 1830 East Monument Street, Suite 333, Baltimore, Maryland 21287, USA
| | | | | |
Collapse
|
44
|
Tang X, Zhang Q, Shi S, Yun Y, Li X, Zhang Y, Zhou K, Le AD. Bisphosphonates suppress insulin-like growth factor 1-induced angiogenesis via the HIF-1alpha/VEGF signaling pathways in human breast cancer cells. Int J Cancer 2010; 126:90-103. [PMID: 19569175 PMCID: PMC2784023 DOI: 10.1002/ijc.24710] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adjunctive chemotherapy with bisphosphonates has been reported to delay bone metastasis and improve overall survival in breast cancer. Aside from its antiresorptive effect, bisphosphonates exhibit antitumor activities, in vitro and in vivo, via several mechanisms, including antiangiogenesis. In this study, we investigated the potential molecular mechanisms underlying the antiangiogenic effect of non-nitrogen-containing and nitrogen-containing bisphosphonates, clodronate and pamidronate, respectively, in insulin-like growth factor (IGF)-1 responsive human breast cancer cells. We tested whether bisphosphonates had any effects on hypoxia-inducible factor (HIF)-1alpha/vascular endothelial growth factor (VEGF) axis that plays a pivotal role in tumor angiogenesis, and our results showed that both pamidronate and clodronate significantly suppressed IGF-1-induced HIF-1alpha protein accumulation and VEGF expression in MCF-7 cells. Mechanistically, we found that either pamidronate or clodronate did not affect mRNA expression of HIF-1alpha, but they apparently promoted the degradation of IGF-1-induced HIF-1alpha protein. Meanwhile, we found that the presence of pamidronate and clodronate led to a dose-dependent decease in the newly-synthesized HIF-1alpha protein induced by IGF-1 in breast cancer cells after proteasomal inhibition, thus, indirectly reflecting the inhibition of protein synthesis. In addition, our results indicated that the inhibitory effects of bisphosphonates on the HIF-1alpha/VEGF axis are associated with the inhibition of the phosphoinositide 3-kinase/AKT/mammalian target of rapamycin signaling pathways. Consistently, we demonstrated that pamidronate and clodronate functionally abrogated both in vitro and in vivo tumor angiogenesis induced by IGF-1-stimulated MCF-7 cells. These findings have highlighted an important mechanism of the pharmacological action of bisphosphonates in the inhibition of tumor angiogenesis in breast cancer cells.
Collapse
Affiliation(s)
- Xudong Tang
- Center for Craniofacial Molecular Biology, University of Southern California, School of Dentistry, Los Angeles, California, 90033
- Institute of Biochemistry and Molecular Biology, Guangdong Medical College, Zhanjiang, Guangdong 524023, P. R. China
| | - Qunzhou Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, School of Dentistry, Los Angeles, California, 90033
| | - Shihong Shi
- Center for Craniofacial Molecular Biology, University of Southern California, School of Dentistry, Los Angeles, California, 90033
| | - Yen Yun
- Clinical & Molecular Pharmacology, City of Hope, Duarte, CA 91010
| | - Xiangyong Li
- Institute of Biochemistry and Molecular Biology, Guangdong Medical College, Zhanjiang, Guangdong 524023, P. R. China
| | - Yuefei Zhang
- Department of Otorhinolaryngology, the First Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524023, P R China
| | - Keyuan Zhou
- Institute of Biochemistry and Molecular Biology, Guangdong Medical College, Zhanjiang, Guangdong 524023, P. R. China
| | - Anh D. Le
- Center for Craniofacial Molecular Biology, University of Southern California, School of Dentistry, Los Angeles, California, 90033
| |
Collapse
|
45
|
Liu D, Hou P, Liu Z, Wu G, Xing M. Genetic alterations in the phosphoinositide 3-kinase/Akt signaling pathway confer sensitivity of thyroid cancer cells to therapeutic targeting of Akt and mammalian target of rapamycin. Cancer Res 2009; 69:7311-9. [PMID: 19706758 DOI: 10.1158/0008-5472.can-09-1077] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We investigated the genotype-dependent therapeutic potential of targeting the phosphoinositide 3-kinase (PI3K)/Akt pathway for thyroid cancer. Proliferation of TPC1, Hth7, FTC133, OCUT1, K1, and BCPAP cells that harbored PI3K/Akt-activating genetic alterations was potently inhibited by the Akt inhibitor perifosine, whereas SW1736, Hth74, WRO, KAT18, and TAD2 cells that harbored no genetic alterations had no or only modest responses. Inhibition of Akt phosphorylation by perifosine was seen in these cells. Genetic-dependent apoptosis was induced by perifosine in cells selectively tested. Similarly, potent inhibition of cell proliferation by the mammalian target of rapamycin (mTOR) inhibitor temsirolimus occurred in virtually all the cells harboring genetic alterations, whereas modest inhibition was seen in some of the cells not harboring genetic alterations. Temsirolimus inhibited the phosphorylation of p70S6K, a substrate of mTOR. Knockdown of Akt1/2 or mTOR by shRNA approach inhibited the proliferation and colony formation of FTC133 and OCUT1 cells that harbored genetic alterations in the PI3K/Akt pathway but had no effect on SW1736 and KAT18 cells that did not. Transfection with PIK3CA mutants greatly sensitized SW1736 cells to perifosine and temsirolimus. Growth of xenograft tumors derived from FTC133 cells but not SW1736 cells in nude mice was dramatically inhibited by perifosine. Thus, this work for the first time shows that genetic alterations in the PI3K/Akt pathway confer thyroid cancer cells addiction to this pathway and their sensitivity to inhibition by targeting Akt and mTOR. This genotype-based targeting of the PI3K/Akt pathway using Akt and mTOR inhibitors may offer an effective therapeutic strategy for thyroid cancer and warrants further studies.
Collapse
Affiliation(s)
- Dingxie Liu
- Division of Endocrinology and Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | | | |
Collapse
|
46
|
Takashima M, Parsons CJ, Ikejima K, Watanabe S, White ES, Rippe RA. The tumor suppressor protein PTEN inhibits rat hepatic stellate cell activation. J Gastroenterol 2009; 44:847-55. [PMID: 19436944 PMCID: PMC2790377 DOI: 10.1007/s00535-009-0073-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Accepted: 04/05/2009] [Indexed: 02/04/2023]
Abstract
BACKGROUND Following a fibrogenic stimulus, the hepatic stellate cell (HSC) transforms from a quiescent to an activated cell type associated with increased proliferation, collagen and smooth muscle alpha-actin (alphaSMA) expression. Phosphatase and Tensin Homolog Deleted on Chromosome Ten (PTEN), a tumor suppressor phosphatase, has been shown to play a role in several nonmalignant diseases. Here, we investigated the role of PTEN during HSC activation. METHODS Rat HSCs 2 days after isolation were transduced with adenoviruses expressing either the wild-type (WT) or a dominant negative form of PTEN, and culture-associated activation of HSCs, including morphological changes, expression of alphaSMA and alpha1(I) collagen, and cell proliferation, were evaluated. Apoptosis of HSCs was detected by measuring activity of caspase 3/7. Phosphorylation status of Akt, p70(S6K), and Erk was detected by Western blotting. RESULTS Overexpression of WT-PTEN inhibited phenotypic changes were associated with HSC activation, including morphological changes, expression of alphaSMA and alpha1(I) collagen, and HSC proliferation, including cyclin D1 expression. WT-PTEN expression also induced apoptosis in HSCs with increased caspase 3/7 activity. Expression of WT-PTEN also caused decreased activation of Akt, p70(S6K), and Erk signaling pathways. CONCLUSIONS Taken together, these findings show that PTEN represents an important negative regulator for transactivation of HSCs. This may have important implications for the design of therapeutic strategies to prevent the progression of liver fibrosis.
Collapse
Affiliation(s)
- Motoki Takashima
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, CB #7032, Medical Biomolecular Research Building, Room 7340B, Chapel Hill, NC 27599-7032, USA
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Christopher J. Parsons
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, CB #7032, Medical Biomolecular Research Building, Room 7340B, Chapel Hill, NC 27599-7032, USA
| | - Kenichi Ikejima
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Sumio Watanabe
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Eric S. White
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Richard A. Rippe
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, CB #7032, Medical Biomolecular Research Building, Room 7340B, Chapel Hill, NC 27599-7032, USA
| |
Collapse
|
47
|
Lu J, Zhang K, Chen S, Wen W. Grape seed extract inhibits VEGF expression via reducing HIF-1alpha protein expression. Carcinogenesis 2009; 30:636-44. [PMID: 19131542 DOI: 10.1093/carcin/bgp009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Grape seed extract (GSE) is a widely consumed dietary supplement that has antitumor activity. Here, we have investigated the inhibitory effect of GSE on the expression of vascular endothelial growth factor (VEGF) and the mechanism underlying this action. We found that GSE inhibited VEGF messenger RNA (mRNA) and protein expression in U251 human glioma cells and MDA-MB-231 human breast cancer cells. GSE inhibited transcriptional activation of the VEGF gene through reducing protein but not mRNA expression of hypoxia-inducible factor (HIF) 1alpha. The inhibitory effect of GSE on HIF-1alpha expression was mainly through inhibiting HIF-1alpha protein synthesis rather than promoting protein degradation. Consistent with this result, GSE-suppressed phosphorylation of several important components involved in HIF-1alpha protein synthesis, such as Akt, S6 kinase and S6 protein. Furthermore, in the MDA-MB-231 tumor, we found that GSE treatment inhibited the expression of VEGF and HIF-1alpha and the phosphorylation of S6 kinase without altering the subcellular localization of HIF-1alpha, correlating with reduced vessel density and tumor size. Depletion of polyphenol with polyvinylpyrrolidone abolished the inhibitory activity of GSE, suggesting a water-soluble fraction of polyphenol in GSE is responsible for the inhibitory activity. Taken together, our results indicate that GSE inhibits VEGF expression by reducing HIF-1alpha protein synthesis through blocking Akt activation. This finding provides new insight into the mechanisms of anticancer activity of GSE and reveals a novel molecular mechanism underlying the antiangiogenic action of GSE.
Collapse
Affiliation(s)
- Jianming Lu
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | | | | | | |
Collapse
|
48
|
Sehon CA, Wang GZ, Viet AQ, Goodman KB, Dowdell SE, Elkins PA, Semus SF, Evans C, Jolivette LJ, Kirkpatrick RB, Dul E, Khandekar SS, Yi T, Wright LL, Smith GK, Behm DJ, Bentley R, Doe CP, Hu E, Lee D. Potent, selective and orally bioavailable dihydropyrimidine inhibitors of Rho kinase (ROCK1) as potential therapeutic agents for cardiovascular diseases. J Med Chem 2008; 51:6631-4. [PMID: 18842034 DOI: 10.1021/jm8005096] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent studies using known Rho-associated kinase isoform 1 (ROCK1) inhibitors along with cellular and molecular biology data have revealed a pivotal role of this enzyme in many aspects of cardiovascular function. Here we report a series of ROCK1 inhibitors which were originally derived from a dihydropyrimidinone core 1. Our efforts focused on the optimization of dihydropyrimidine 2, which resulted in the identification of a series of dihydropyrimidines with improved pharmacokinetics and P450 properties.
Collapse
Affiliation(s)
- Clark A Sehon
- Departments of Medicinal Chemistry, Investigative Biology, Vascular Biology, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, Pennsylvania 19406, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Li J, Zhang D, Stoner GD, Huang C. Differential effects of black raspberry and strawberry extracts on BaPDE-induced activation of transcription factors and their target genes. Mol Carcinog 2008; 47:286-94. [PMID: 18085529 DOI: 10.1002/mc.20377] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The chemopreventive properties of edible berries have been demonstrated both in vitro and in vivo, however, the specific molecular mechanisms underlying their anti-cancer effects are largely unknown. Our previous studies have shown that a methanol extract fraction of freeze-dried black raspberries inhibits benzoapyrene (BaP)-induced transformation of Syrian hamster embryo cells. This fraction also blocks activation of activator protein-1 (AP-1) and nuclear factor kappaB (NF-kappaB) induced by benzoapyrene diol-epoxide (BaPDE) in mouse epidermal JB6 Cl 41 cells. To determine if different berry types exhibit specific mechanisms for their anti-cancer effects, we compared the effects of extract fractions from both black raspberries and strawberries on BaPDE-induced activation of various signaling pathways in Cl 41 cells. Black raspberry fractions inhibited the activation of AP-1, NF-kappaB, and nuclear factor of activated T cells (NFAT) by BaPDE as well as their upstream PI-3K/Akt-p70(S6K) and mitogen-activated protein kinase pathways. In contrast, strawberry fractions inhibited NFAT activation, but did not inhibit the activation of AP-1, NF-kappaB or the PI-3K/Akt-p70(S6K) and mitogen-activated protein kinase pathways. Consistent with the effects on NFAT activation, tumor necrosis factor-alpha (TNF-alpha) induction by BaPDE was blocked by extract fractions of both black raspberries and strawberries, whereas vascular endothelial growth factor (VEGF) expression, which depends on AP-1 activation, was suppressed by black raspberry fractions but not strawberry fractions. These results suggest that black raspberry and strawberry components may target different signaling pathways in exerting their anti-carcinogenic effects.
Collapse
Affiliation(s)
- Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, New York, New York 10987, USA
| | | | | | | |
Collapse
|
50
|
Yoon SC, Seo MS, Kim SH, Jeon WJ, Ahn YM, Kang UG, Kim YS. The effect of MK-801 on mTOR/p70S6K and translation-related proteins in rat frontal cortex. Neurosci Lett 2008; 434:23-8. [PMID: 18262357 DOI: 10.1016/j.neulet.2008.01.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Revised: 12/26/2007] [Accepted: 01/08/2008] [Indexed: 10/22/2022]
Abstract
In experimental animals, including rats, MK-801 produces characteristic behavioural changes that model schizophrenia. It has been hypothesized that these changes accompany long-term synaptic changes, which require protein neosynthesis. We observed the effect of MK-801 on the "mammalian target of rapamycin" (mTOR)/70-kDa ribosomal protein S6 kinase (p70S6K) pathway that regulates protein synthesis in the rat frontal cortex. A single injection of MK-801 (0.5, 1, or 2mg/kg) induced an acute increase in the phosphorylation of Akt (Ser-473) eIF4E-binding protein (4E-BP1) (Thr-37/46) and p70S6K (Thr-389). In contrast, after repeated treatment with MK-801 (1mg/kg for 5 or 10 days), the phosphorylation of Akt (Ser-473), mTOR (Ser-2481), 4E-BP1 (Thr-37/46), p70S6K (Thr-389), and S6 (Ser-240/244) increased. Thus, proteins in the mTOR/p70S6K pathway are modulated in chronic MK-801 animal models. These findings may suggest that repeated MK-801 treatment activates the signal transduction pathways involved in the initiation of protein synthesis in the rat frontal cortex.
Collapse
Affiliation(s)
- Se Chang Yoon
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|