1
|
Adon T, Bhattacharya S, Madhunapantula SV, Kumar HY. Structural requirements of isoform-specific inhibitors of Akt: Implications in the development of effective cancer treatment strategies. Eur J Med Chem 2025; 287:117334. [PMID: 39904143 DOI: 10.1016/j.ejmech.2025.117334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/18/2024] [Accepted: 01/25/2025] [Indexed: 02/06/2025]
Abstract
Akt, also known as protein kinase-B, is an important therapeutic target in the treatment of cancer due to its pivotal roles in the signaling pathways that regulate various hall-mark features of cancer cells such as cell growth, survival, migration, differentiation, and metabolism. The three closely related isoforms of Akt viz., Akt1, Akt2, and Akt3 exhibit distinct physiological roles that affect cellular behavior and tumor development, making isoform selectivity a crucial driving factor in the design and development of inhibitors. This review outlines key amino acids and their structural traits in Akt isoforms, potentially dictating isoform selectivity. We present an analysis of existing structure-activity relationship data of covalent-allosteric Akt inhibitors to shed light on isoform selectivity. Additionally, a brief review of potential predictive biomarkers in enhancing the therapeutic efficacy of Akt inhibitors is presented. Identifying biomarkers that can reliably predict patient response to treatment is crucial for personalizing cancer therapies and improving overall treatment outcomes. By integrating predictive biomarker identification with the ongoing development of isoform-selective Akt inhibitors, it is plausible to establish a foundation for more precise and efficacious interventions in cancer therapy.
Collapse
Affiliation(s)
- Tenzin Adon
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India; Computer Aided Drug Design Lab, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India
| | - Sanyukta Bhattacharya
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India; Computer Aided Drug Design Lab, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India
| | - SubbaRao V Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR, A DST-FIST Supported Center and ICMR-Collaborating Center of Excellence), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India; Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India; University Sophisticated Instrumentation Centre (USIC) [Supported by DST-PURSE & DBT-BUILDER], JSS Academy of Higher Education & Research (JSS AHER), Mysuru, 570015, Karnataka, India
| | - Honnavalli Yogish Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India; Computer Aided Drug Design Lab, Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, Karnataka, India; University Sophisticated Instrumentation Centre (USIC) [Supported by DST-PURSE & DBT-BUILDER], JSS Academy of Higher Education & Research (JSS AHER), Mysuru, 570015, Karnataka, India.
| |
Collapse
|
2
|
Li P, Ma X, Gu X. The essential roles of lncRNAs/PI3K/AKT axis in gastrointestinal tumors. Front Cell Dev Biol 2024; 12:1442193. [PMID: 39161590 PMCID: PMC11330846 DOI: 10.3389/fcell.2024.1442193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
The role of long noncoding RNA (lncRNA) in tumors, particularly in gastrointestinal tumors, has gained significant attention. Accumulating evidence underscores the interaction between various lncRNAs and diverse molecular pathways involved in cancer progression. One such pivotal pathway is the PI3K/AKT pathway, which serves as a crucial intracellular mechanism maintaining the balance among various cellular physiological processes for normal cell growth and survival. Frequent dysregulation of the PI3K/AKT pathway in cancer, along with aberrant activation, plays a critical role in driving tumorigenesis. LncRNAs modulate the PI3K/AKT signaling pathway through diverse mechanisms, primarily by acting as competing endogenous RNA to regulate miRNA expression and associated genes. This interaction significantly influences fundamental biological behaviors such as cell proliferation, metastasis, and drug resistance. Abnormal expression of numerous lncRNAs in gastrointestinal tumors often correlates with clinical outcomes and pathological features in patients with cancer. Additionally, these lncRNAs influence the sensitivity of tumor cells to chemotherapy in multiple types of gastrointestinal tumors through the abnormal activation of the PI3K/AKT pathway. These findings provide valuable insights into the mechanisms underlying gastrointestinal tumors and potential therapeutic targets. However, gastrointestinal tumors remain a significant global health concern, with increasing incidence and mortality rates of gastrointestinal tumors over recent decades. This review provides a comprehensive summary of the latest research on the interactions of lncRNA and the PI3K/AKT pathway in gastrointestinal tumor development. Additionally, it focuses on the functions of lncRNAs and the PI3K/AKT pathway in carcinogenesis, exploring expression profiles, clinicopathological characteristics, interaction mechanisms with the PI3K/AKT pathway, and potential clinical applications.
Collapse
Affiliation(s)
- Penghui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiao Ma
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
3
|
Wang R, Jia S, Chen H, Luo K, Zhang L, Song Y, Qing C, Liu D, Zhou H. Antiplatelet drug ticagrelor suppresses triple negative breast cancer metastasis by targeting PI3K. Biochem Pharmacol 2024; 226:116408. [PMID: 38969297 DOI: 10.1016/j.bcp.2024.116408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Metastatic recurrence is still a major challenge in breast cancer treatment. Patients with triple negative breast cancer (TNBC) develop early recurrence and relapse more frequently. Due to the lack of specific therapeutic targets, new targeted therapies for TNBC are urgently needed. Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway is one of the active pathways involved in chemoresistance and survival of TNBC, being considered as a potential target for TNBC treatment. Our present study identified ticagrelor, an anti-platelet drug, as a pan-PI3K inhibitor with potent inhibitory activity against four isoforms of class I PI3K. At doses normally used in clinic, ticagrelor showed weak cytotoxicity against a panel of breast cancer cells, but significantly inhibited the migration, invasion and the actin cytoskeleton organization of human TNBC MDA-MB-231 and SUM-159PT cells. Mechanistically, ticagrelor effectively inhibited PI3K downstream mTOR complex 1 (mTORC1) and mTORC2 signaling by targeting PI3K and decreased the protein expression of epithelial-mesenchymal transition (EMT) markers. In vivo, ticagrelor significantly suppressed tumor cells lung metastasis in 4T1 tumor bearing BALB/c mice model and experimental lung metastasis model which was established by tail vein injection of GFP-labeled MDA-MB-231 cells. The above data demonstrated that ticagrelor can inhibit the migration and invasion of TNBC both in vitro and in vivo by targeting PI3K, suggesting that ticagrelor, a pan-PI3K inhibitor, might represent a promising therapeutic agent for the treatment of metastatic TNBC.
Collapse
Affiliation(s)
- Rong Wang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Shutao Jia
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Hongyan Chen
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Kaitao Luo
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Limei Zhang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Yan Song
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Chen Qing
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Dandan Liu
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming 650500, Yunnan, China.
| | - Hongyu Zhou
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, Yunnan, China; Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming 650500, Yunnan, China.
| |
Collapse
|
4
|
Dzhemileva LU, Tuktarova RA, Dzhemilev UM, D’yakonov VA. Natural Acetogenins, Chatenaytrienins-1, -2, -3 and -4, Mitochondrial Potential Uncouplers and Autophagy Inducers-Promising Anticancer Agents. Antioxidants (Basel) 2023; 12:1528. [PMID: 37627523 PMCID: PMC10451668 DOI: 10.3390/antiox12081528] [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: 06/26/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
The present paper details the complete stereoselective synthesis of four natural acetogenins, chatenaytrienins-1, -2, -3 and -4, previously isolated from the roots of fruit trees of the family Annonaceae (A. nutans and A. muricata), as an inseparable mixture. The novel organometallic reactions, developed by the authors, of Ti-catalyzed cross-cyclomagnesiation of O-containing and aliphatic allenes using available Grignard reagents were applied at the key stage of synthesis. We have studied the biological activity of the synthesized individual chatenaytrienins-1, -2, -3 and -4 in vitro, including their cytotoxicity in a panel of tumor lines and their ability to induce apoptosis, affect the cell cycle and mitochondria, and activate the main apoptotic signaling pathways in the cell, applying modern approaches of flow cytometry and multiplex analysis with Luminex xMAP technology. It has been shown that chatenaytrienins affect mitochondria by uncoupling the processes of mitochondrial respiration, causing the accumulation of ROS ions, followed by the initiation of apoptosis. The most likely mechanism for the death of cortical neurons from the consumption of tea from the seeds of Annona fruit is long-term chronic hypoxia, which leads to the development of an atypical form of Parkinson's disease that is characteristic of the indigenous inhabitants of Guam and New Caledonia.
Collapse
Affiliation(s)
- Lilya U. Dzhemileva
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, Moscow 119991, Russia; (R.A.T.); (U.M.D.)
| | | | | | - Vladimir A. D’yakonov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, Moscow 119991, Russia; (R.A.T.); (U.M.D.)
| |
Collapse
|
5
|
Kempska J, Oliveira-Ferrer L, Grottke A, Qi M, Alawi M, Meyer F, Borgmann K, Hamester F, Eylmann K, Rossberg M, Smit DJ, Jücker M, Laakmann E, Witzel I, Schmalfeldt B, Müller V, Legler K. Impact of AKT1 on cell invasion and radiosensitivity in a triple negative breast cancer cell line developing brain metastasis. Front Oncol 2023; 13:1129682. [PMID: 37483521 PMCID: PMC10358765 DOI: 10.3389/fonc.2023.1129682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction The PI3K/AKT pathway is activated in 43-70% of breast cancer (BC)-patients and promotes the metastatic potential of BC cells by increasing cell proliferation, invasion and radioresistance. Therefore, AKT1-inhibition in combination with radiotherapy might be an effective treatment option for triple-negative breast cancer (TNBC)-patients with brain metastases. Methods The impact of AKT1-knockout (AKT1_KO) and AKT-inhibition using Ipatasertib on MDA-MB-231 BR cells was assessed using in vitro cell proliferation and migration assays. AKT1-knockout in MDA-MB-231BR cells was performed using CRISPR/Cas9. The effect of AKT1-knockout on radiosensitivity of MDA-MB-231BR cell lines was determined via colony formation assays after cell irradiation. To detect genomic variants in AKT1_KO MDA-MB-231BR cells, whole-genome sequencing (WGS) was performed. Results Pharmacological inhibition of AKT with the pan-AKT inhibitor Ipatasertib led to a significant reduction of cell viability but did not impact cell migration. Moreover, only MDA-MB-231BR cells were sensitized following Ipatasertib-treatment. Furthermore, specific AKT1-knockout in MDA-MB-231BR showed reduced cell viability in comparison to control cells, with significant effect in one of two analyzed clones. Unexpectedly, AKT1 knockout led to increased cell migration and clonogenic potential in both AKT1_KO clones. RNAseq-analysis revealed the deregulation of CTSO, CYBB, GPR68, CEBPA, ID1, ID4, METTL15, PBX1 and PTGFRN leading to the increased cell migration, higher clonogenic survival and decreased radiosensitivity as a consequence of the AKT1 knockout in MDA-MB-231BR. Discussion Collectively, our results demonstrate that Ipatasertib leads to radiosensitization and reduced cell proliferation of MDA-MB-231BR. AKT1-inhibition showed altered gene expression profile leading to modified cell migration, clonogenic survival and radioresistance in MDA-MB-231BR. We conclude, that AKT1-inhibition in combination with radiotherapy contribute to novel treatment strategies for breast cancer brain metastases.
Collapse
Affiliation(s)
- Joanna Kempska
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Astrid Grottke
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minyue Qi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Meyer
- Laboratory of Radiobiology & Experimental Radio Oncology, Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Borgmann
- Laboratory of Radiobiology & Experimental Radio Oncology, Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabienne Hamester
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kathrin Eylmann
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maila Rossberg
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel J. Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elena Laakmann
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabell Witzel
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karen Legler
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Liu G, Wang Y, Zheng Y, Lv J, Li Y, Liu N, Gao H, Ran H, Tang H, Jiang Z. PHB2 binds to ERβ to induce the autophagy of porcine ovarian granulosa cells through mTOR phosphorylation. Theriogenology 2023; 198:114-122. [PMID: 36580849 DOI: 10.1016/j.theriogenology.2022.12.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
Autophagy of ovarian granulosa cells is one of the reasons which results in follicular atresia. PHB2 regulates many fundamental biological processes and is pivotal in the mitophagy of cells; nevertheless, the autophagy in the porcine ovary and how PHB2 regulates the follicular cells are unknown. Here we report a protein complex that induces autophagy in porcine granulosa cells (PGCs) through the direct interaction of ERβ and PHB2. In this study, we aimed to elucidate the autophagy and the role of PHB2 in porcine ovaries using porcine primary ovarian granulosa cells (PGCs). The results showed that PHB2 induces PGCs autophagy because of the change in related genes and protein expression levels. In addition, the results of Co-IP and the distribution of the combination of PHB2 and ERβ showed that this complex is also indicated as an essential role of PHB2 in PGCs autophagy. Based on our results, it can be concluded that PHB2 combined with ERβ induces PGCs autophagy by targeting the mTOR pathway. This study pinpoints a novel regulatory mechanism of autophagy and demonstrates the existence of a protein complex that may underlie its roles in autophagy in PGCs.
Collapse
Affiliation(s)
- Guangyu Liu
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China
| | - Yifan Wang
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China
| | - Yuxin Zheng
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China
| | - Jing Lv
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China
| | - Yuanyou Li
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China
| | - Ning Liu
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China
| | - Huimin Gao
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China
| | - Haohan Ran
- College of Animal Science and Animal Medicine, Tianjin Agricultural University, Tianjin, China
| | - Hong Tang
- State Key Laboratory for Sheep Genetic Improvement and Healthy Production/Institute of Animal Husbandry and Veterinary Medicine, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihezi, 832000, China.
| | - Zhongliang Jiang
- College of Animal Science and Technology, Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, Northwestern A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
7
|
Derakhshani A, Rostami Z, Safarpour H, Shadbad MA, Nourbakhsh NS, Argentiero A, Taefehshokr S, Tabrizi NJ, Kooshkaki O, Astamal RV, Singh PK, Taefehshokr N, Alizadeh N, Silvestris N, Baradaran B. From Oncogenic Signaling Pathways to Single-Cell Sequencing of Immune Cells: Changing the Landscape of Cancer Immunotherapy. Molecules 2021; 26:2278. [PMID: 33920054 PMCID: PMC8071039 DOI: 10.3390/molecules26082278] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
Over the past decade, there have been remarkable advances in understanding the signaling pathways involved in cancer development. It is well-established that cancer is caused by the dysregulation of cellular pathways involved in proliferation, cell cycle, apoptosis, cell metabolism, migration, cell polarity, and differentiation. Besides, growing evidence indicates that extracellular matrix signaling, cell surface proteoglycans, and angiogenesis can contribute to cancer development. Given the genetic instability and vast intra-tumoral heterogeneity revealed by the single-cell sequencing of tumoral cells, the current approaches cannot eliminate the mutating cancer cells. Besides, the polyclonal expansion of tumor-infiltrated lymphocytes in response to tumoral neoantigens cannot elicit anti-tumoral immune responses due to the immunosuppressive tumor microenvironment. Nevertheless, the data from the single-cell sequencing of immune cells can provide valuable insights regarding the expression of inhibitory immune checkpoints/related signaling factors in immune cells, which can be used to select immune checkpoint inhibitors and adjust their dosage. Indeed, the integration of the data obtained from the single-cell sequencing of immune cells with immune checkpoint inhibitors can increase the response rate of immune checkpoint inhibitors, decrease the immune-related adverse events, and facilitate tumoral cell elimination. This study aims to review key pathways involved in tumor development and shed light on single-cell sequencing. It also intends to address the shortcomings of immune checkpoint inhibitors, i.e., their varied response rates among cancer patients and increased risk of autoimmunity development, via applying the data from the single-cell sequencing of immune cells.
Collapse
Affiliation(s)
- Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Zeinab Rostami
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 97178-53577, Iran;
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| | | | | | - Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Neda Jalili Tabrizi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Omid Kooshkaki
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Reza Vaezi Astamal
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Pankaj Kumar Singh
- Principal Research Technologist, Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA;
| | - Nima Taefehshokr
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| |
Collapse
|
8
|
D’yakonov VA, Makarov AA, Dzhemileva LU, Ramazanov IR, Makarova EK, Dzhemilev UM. Natural Trienoic Acids as Anticancer Agents: First Stereoselective Synthesis, Cell Cycle Analysis, Induction of Apoptosis, Cell Signaling and Mitochondrial Targeting Studies. Cancers (Basel) 2021; 13:cancers13081808. [PMID: 33920067 PMCID: PMC8070279 DOI: 10.3390/cancers13081808] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/30/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Currently, the whole world is acutely concerned with the selection of effective treatment regimens for oncological diseases. This problem is becoming more and more catastrophic every year due to the phenomenon of multidrug resistance, the consequence of which is the loss of the effectiveness of drugs against tumor cells. One of the solutions to the problem described above is the synthesis of new low molecular weight compounds that can effectively affect molecular cellular targets, for example, enzymes of the cell cycle, and, as a consequence, interrupt DNA synthesis, contributing to tumor death. Within the framework of this article, we carried out the Z-stereoselective synthesis of natural unsaturated acids containing a 1Z,5Z,9Z-triene moiety, for which it was shown that they are effective inhibitors of human topoisomerase I, and also affect mitochondria. At the same time, using multiplex analysis, the activation of signaling pathways was studied and a probable mechanism of the antitumor action of the synthesized trienoic acids was proposed. Abstract The first Z-stereoselective method was developed for the synthesis of unsaturated acids containing a 1Z,5Z,9Z-triene moiety in 61–64% yields using the new Ti-catalyzed cross-coupling of oxygen-containing and aliphatic 1,2-dienes as the key synthetic step. It was shown for the first time that trienoic acids with non-methylene-interrupted Z-double bonds show moderate cytotoxic activities against tumor cell lines (Jurkat, K562, U937, HL60, HeLa), human embryonic kidney cells (Hek293), normal fibroblasts and human topoisomerase I (hTop1) inhibitory activity in vitro. The synthesized acids efficiently initiate apoptosis of Jurkat tumor cells, with the cell death mechanism being activated by the mitochondrial pathway. A probable mechanism of topoisomerase I inhibition was also hypothesized on the basis of in silico studies resorting to docking. The activation and inhibition of the most versatile intracellular signaling pathways (CREB, JNK, NFkB, p38, ERK1/2, Akt, p70S6K, STAT3 and STAT5 tyrosine kinases) responsible for cell proliferation and for initiation of apoptosis were studied by multiplex assay technology (Luminex xMAP).
Collapse
|
9
|
Alexander JI, Vendramini-Costa DB, Francescone R, Luong T, Franco-Barraza J, Shah N, Gardiner JC, Nicolas E, Raghavan KS, Cukierman E. Palladin isoforms 3 and 4 regulate cancer-associated fibroblast pro-tumor functions in pancreatic ductal adenocarcinoma. Sci Rep 2021; 11:3802. [PMID: 33589694 PMCID: PMC7884442 DOI: 10.1038/s41598-021-82937-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 01/27/2021] [Indexed: 02/04/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) has a five-year survival under 10%. Treatment is compromised due to a fibrotic-like stromal remodeling process, known as desmoplasia, which limits therapeutic perfusion, supports tumor progression, and establishes an immunosuppressive microenvironment. These processes are driven by cancer-associated fibroblasts (CAFs), functionally activated through transforming growth factor beta1 (TGFβ1). CAFs produce a topographically aligned extracellular matrix (ECM) that correlates with reduced overall survival. Paradoxically, ablation of CAF populations results in a more aggressive disease, suggesting CAFs can also restrain PDAC progression. Thus, unraveling the mechanism(s) underlying CAF functions could lead to therapies that reinstate the tumor-suppressive features of the pancreatic stroma. CAF activation involves the f-actin organizing protein palladin. CAFs express two palladin isoforms (iso3 and iso4) which are up-regulated in response to TGFβ1. However, the roles of iso3 and iso4 in CAF functions remain elusive. Using a CAF-derived ECM model, we uncovered that iso3/iso4 are required to sustain TGFβ1-dependent CAF activation, secrete immunosuppressive cytokines, and produce a pro-tumoral ECM. Findings demonstrate a novel role for CAF palladin and suggest that iso3/iso4 regulate both redundant and specific tumor-supportive desmoplastic functions. This study highlights the therapeutic potential of targeting CAFs to restore fibroblastic anti-tumor activity in the pancreatic microenvironment.
Collapse
Affiliation(s)
- J I Alexander
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
- Molecular, Cellular Biology and Genetics Program, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - D B Vendramini-Costa
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - R Francescone
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - T Luong
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - J Franco-Barraza
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - N Shah
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - J C Gardiner
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - E Nicolas
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - K S Raghavan
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
- Molecular, Cellular Biology and Genetics Program, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - E Cukierman
- Cancer Biology and the Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Chan S, Smith E, Gao Y, Kwan J, Blum BC, Tilston-Lunel AM, Turcinovic I, Varelas X, Cardamone MD, Monti S, Emili A, Perissi V. Loss of G-Protein Pathway Suppressor 2 Promotes Tumor Growth Through Activation of AKT Signaling. Front Cell Dev Biol 2021; 8:608044. [PMID: 33490071 PMCID: PMC7817781 DOI: 10.3389/fcell.2020.608044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
G Protein Suppressor 2 (GPS2) is a multifunctional protein that exerts important roles in inflammation and metabolism in adipose, liver, and immune cells. GPS2 has recently been identified as a significantly mutated gene in breast cancer and other malignancies and proposed to work as a putative tumor suppressor. However, molecular mechanisms by which GPS2 prevents cancer development and/or progression are largely unknown. Here, we have profiled the phenotypic changes induced by GPS2 depletion in MDA-MB-231 triple negative breast cancer cells and investigated the underlying molecular mechanisms. We found that GPS2-deleted MDA-MB-231 cells exhibited increased proliferative, migratory, and invasive properties in vitro, and conferred greater tumor burden in vivo in an orthotopic xenograft mouse model. Transcriptomic, proteomic and phospho-proteomic profiling of GPS2-deleted MBA-MB-231 revealed a network of altered signals that relate to cell growth and PI3K/AKT signaling. Overlay of GPS2-regulated gene expression with MDA-MB-231 cells modified to express constitutively active AKT showed significant overlap, suggesting that sustained AKT activation is associated with loss of GPS2. Accordingly, we demonstrate that the pro-oncogenic phenotypes associated with GPS2 deletion are rescued by pharmacological inhibition of AKT with MK2206. Collectively, these observations confirm a tumor suppressor role for GPS2 and reveal that loss of GPS2 promotes breast cancer cell proliferation and tumor growth through uncontrolled activation of AKT signaling. Moreover, our study points to GPS2 as a potential biomarker for a subclass of breast cancers that would be responsive to PI3K-class inhibitor drugs.
Collapse
Affiliation(s)
- Stefanie Chan
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Emma Smith
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Yuan Gao
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Julian Kwan
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Benjamin C. Blum
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | | | - Isabella Turcinovic
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Maria Dafne Cardamone
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Stefano Monti
- Division of Computational Biology, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Valentina Perissi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
11
|
Merckaert T, Zwaenepoel O, Gevaert K, Gettemans J. An AKT2-specific nanobody that targets the hydrophobic motif induces cell cycle arrest, autophagy and loss of focal adhesions in MDA-MB-231 cells. Biomed Pharmacother 2020; 133:111055. [PMID: 33378961 DOI: 10.1016/j.biopha.2020.111055] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/21/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
The AKT kinase family is a high-profile target for cancer therapy. Despite their high degree of homology the three AKT isoforms (AKT1, AKT2 and AKT3) are non-redundant and can even have opposing functions. Small-molecule AKT inhibitors affect all three isoforms which severely limits their usefulness as research tool or therapeutic. Using AKT2-specific nanobodies we examined the function of endogenous AKT2 in breast cancer cells. Two AKT2 nanobodies (Nb8 and Nb9) modulate AKT2 and reduce MDA-MB-231 cell viability/proliferation. Nb8 binds the AKT2 hydrophobic motif and reduces IGF-1-induced phosphorylation of this site. This nanobody also affects the phosphorylation and/or expression levels of a wide range of proteins downstream of AKT, resulting in a G0/G1 cell cycle arrest, the induction of autophagy, a reduction in focal adhesion count and loss of stress fibers. While cell cycle progression is likely to be regulated by more than one isoform, our results indicate that both the effects on autophagy and the cytoskeleton are specific to AKT2. By using an isoform-specific nanobody we were able to map a part of the AKT2 pathway. Our results confirm AKT2 and the hydrophobic motif as targets for cancer therapy. Nb8 can be used as a research tool to study AKT2 signalling events and aid in the design of an AKT2-specific inhibitor.
Collapse
Affiliation(s)
- Tijs Merckaert
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| | - Olivier Zwaenepoel
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| | - Kris Gevaert
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| |
Collapse
|
12
|
Merckaert T, Zwaenepoel O, Gevaert K, Gettemans J. Development and characterization of protein kinase B/AKT isoform-specific nanobodies. PLoS One 2020; 15:e0240554. [PMID: 33045011 PMCID: PMC7549812 DOI: 10.1371/journal.pone.0240554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/28/2020] [Indexed: 11/23/2022] Open
Abstract
The serine/threonine protein kinase AKT is frequently over-activated in cancer and is associated with poor prognosis. As a central node in the PI3K/AKT/mTOR pathway, which regulates various processes considered to be hallmarks of cancer, this kinase has become a prime target for cancer therapy. However, AKT has proven to be a highly complex target as it comes in three isoforms (AKT1, AKT2 and AKT3) which are highly homologous, yet non-redundant. The isoform-specific functions of the AKT kinases can be dependent on context (i.e. different types of cancer) and even opposed to one another. To date, there is no isoform-specific inhibitor available and no alternative to genetic approaches to study the function of a single AKT isoform. We have developed and characterized nanobodies that specifically interact with the AKT1 or AKT2 isoforms. These new tools should enable future studies of AKT1 and AKT2 isoform-specific functions. Furthermore, for both isoforms we obtained a nanobody that interferes with the AKT-PIP3-interaction, an essential step in the activation of the kinase. The nanobodies characterized in this study are a new stepping stone towards unravelling AKT isoform-specific signalling.
Collapse
Affiliation(s)
- Tijs Merckaert
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Kris Gevaert
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
13
|
Makarov AA, Dzhemileva LU, Salimova AR, Makarova EK, Ramazanov IR, D'yakonov VA, Dzhemilev UM. New synthetic analogues of natural 5Z,9Z-dienoic acids: Stereoselective synthesis and study of the anticancer activity. Bioorg Chem 2020; 104:104303. [PMID: 33011528 DOI: 10.1016/j.bioorg.2020.104303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/05/2020] [Accepted: 09/20/2020] [Indexed: 12/23/2022]
Abstract
A stereoselective method was developed for the synthesis of synthetic analogues of natural 5Z,9Z-dienoic acids by esterification of aliphatic and aromatic alcohols and carboxylic acids with (5Z,9Z)-1,14-tetradeca-5,9-dienedioic acid and (5Z,9Z)-1,14-tetradeca-5,9-dienediol, synthesized by Ti-catalyzed homo-cyclomagnesiation of the tetrahydropyran ether of hepta-5,6-dien-1-ol with Grignard reagents. In order to establish the effect of molecular structure on the antitumor activity, the obtained 5Z,9Z-dienoic acids were tested for the inhibitory activity against human topoisomerase I, the cytotoxic activity in vitro against several cancer and normal cell lines (Jurkat, HL-60, K562, U937, fibroblasts), the effect on the cell cycle, and apoptosis-inducing ability using flow cytofluorometry. In addition, the effect of the synthesized acids on the cancer cell production of some phosphorylated and unphosphorylated proteins responsible for proliferation and apoptosis was studied by a new multiplex assay technology, MAGPIX.
Collapse
Affiliation(s)
- Alexey A Makarov
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russian Federation
| | - Lilya U Dzhemileva
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russian Federation.
| | - Alfiya R Salimova
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russian Federation
| | - Elina Kh Makarova
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russian Federation
| | - Ilfir R Ramazanov
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russian Federation
| | - Vladimir A D'yakonov
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russian Federation.
| | - Usein M Dzhemilev
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, 141 Prospekt Oktyabrya, Ufa 450075, Russian Federation
| |
Collapse
|
14
|
Cui H, Cheng Y, He Y, Cheng W, Zhao W, Zhao H, Zhou FH, Wang L, Dong J, Cai S. The AKT inhibitor MK2206 suppresses airway inflammation and the pro‑remodeling pathway in a TDI‑induced asthma mouse model. Mol Med Rep 2020; 22:3723-3734. [PMID: 33000187 PMCID: PMC7533517 DOI: 10.3892/mmr.2020.11450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
The cellular and molecular mechanisms via which MK2206, an AKT inhibitor, prevents the activation of AKT in toluene diisocyanate (TDI)‑induced asthma remain unclear. Thus, the present study aimed to evaluate the potential effects of MK2206 on airway AKT activation, inflammation and remodeling in a TDI‑induced mouse model of asthma. A total of 24 BALB/c mice were selected and randomly divided into untreated (AOO), asthma (TDI), MK2206 (TDI + MK2206), and dexamethasone (TDI + DEX) groups. Phosphorylated AKT (p‑AKT), total AKT, airway remodeling indices, α‑smooth muscle actin (α‑SMA) and collagen I levels in pulmonary tissue were measured using western blotting. Airway inflammation factors, including interleukin (IL)‑4, ‑5, ‑6, and ‑13 in bronchoalveolar lavage fluid (BALF) and IgE in serum, were determined using ELISA. Additionally, the airway hyperresponsiveness (AHR) and pulmonary pathology of all groups were evaluated. The results of the present study demonstrated that p‑AKT levels in lung protein lysate were upregulated, and neutrophil, eosinophil and lymphocyte counts were increased in the lungs obtained from the asthma group compared with the AOO group. Both MK2206 and DEX treatment in TDI‑induced mice resulted not only in the attenuation of AKT phosphorylation, but also reductions in neutrophil, eosinophil and lymphocyte counts in the lungs of mice in the asthma group. Consistently, increases in the levels of the inflammatory cytokines IL‑4, ‑5, ‑6 and ‑13 analyzed in BALF, and serum IgE in the TDI group were demonstrated to be attenuated in the TDI + MK2206 and TDI + DEX groups. Furthermore, α‑SMA and AHR were significantly attenuated in the TDI + MK2206 group compared with the TDI group. These results revealed that MK2206 not only inhibited AKT activation, but also served a role in downregulating airway inflammation and airway remodeling in chemical‑induced asthma. Therefore, the findings of the present study may provide important insight into further combination therapy.
Collapse
Affiliation(s)
- Haiyan Cui
- Department of Respiratory and Critical Care Medicine, Chronic Airway Disease Laboratory, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yuanxiong Cheng
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Yi He
- Department of Immunology Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Weiying Cheng
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Wenqu Zhao
- Department of Respiratory and Critical Care Medicine, Chronic Airway Disease Laboratory, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Haijin Zhao
- Department of Respiratory and Critical Care Medicine, Chronic Airway Disease Laboratory, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Fiona H Zhou
- UniSA Clinical and Health Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Liping Wang
- UniSA Clinical and Health Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Jianghui Dong
- UniSA Clinical and Health Sciences, UniSA Cancer Research Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Shaoxi Cai
- Department of Respiratory and Critical Care Medicine, Chronic Airway Disease Laboratory, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
15
|
Dzhemileva LU, D'yakonov VA, Islamov II, Yunusbaeva MM, Dzhemilev UM. New 1Z,5Z-diene macrodiolides: Catalytic synthesis, anticancer activity, induction of mitochondrial apoptosis, and effect on the cell cycle. Bioorg Chem 2020; 99:103832. [PMID: 32315897 DOI: 10.1016/j.bioorg.2020.103832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 02/03/2023]
Abstract
An original scheme was developed for the synthesis of previously undescribed unsaturated macrodiolides containing a 1Z,5Z-diene moiety in 44-80% yields and with high stereoselectivity (>95%) based on the intermolecular esterification of α,ω-diols with α,ω-alka-nZ,(n + 4)Z-dienedicarboxylic acids (1,12-dodeca-4Z,8Z-dienedicarboxylic acid, 1,14-tetradeca-5Z,9Z-dienedicarboxylic acid, 1,18-octadeca-7Z,11Z-dienedicarboxylic acid) catalyzed by hafnium triflate [Hf(OTf)4]. The unsaturated dicarboxylic acids were prepared via homo-cyclomagnesiation of tetrahydropyran ethers of O-containing 1,2-dienes with EtMgBr in the presence of Mg metal and the Cp2TiCl2 catalyst (10 mol.%) and the subsequent Jones oxidation of pyran ethers formed after the acid hydrolysis of magnesacyclopentanes. The thus prepared macrodiolides exhibit high cytotoxic activity in vitro against Jurkat, K562, U937, Hek293 and HeLa cancer cell lines. It was found that induction of the programmed cell death in Jurkat cells by macrodiolides corresponds to the mitochondrial apoptosis pathway. Also, it was shown that the prepared macrodiolides efficiently suppress phosphorylation of Akt and p38 kinases and CREB transcription factor in cancer cells.
Collapse
Affiliation(s)
- Lilya U Dzhemileva
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation.
| | - Vladimir A D'yakonov
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation.
| | - Ilgiz I Islamov
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation
| | - Milyausha M Yunusbaeva
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation
| | - Usein M Dzhemilev
- Institute of Petrochemistry and Catalysis of RAS (IPC RAS), Prospect Oktyabrya, 141, 450075 Ufa, Russian Federation
| |
Collapse
|
16
|
Hinz N, Jücker M. Distinct functions of AKT isoforms in breast cancer: a comprehensive review. Cell Commun Signal 2019; 17:154. [PMID: 31752925 PMCID: PMC6873690 DOI: 10.1186/s12964-019-0450-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AKT, also known as protein kinase B, is a key element of the PI3K/AKT signaling pathway. Moreover, AKT regulates the hallmarks of cancer, e.g. tumor growth, survival and invasiveness of tumor cells. After AKT was discovered in the early 1990s, further studies revealed that there are three different AKT isoforms, namely AKT1, AKT2 and AKT3. Despite their high similarity of 80%, the distinct AKT isoforms exert non-redundant, partly even opposing effects under physiological and pathological conditions. Breast cancer as the most common cancer entity in women, frequently shows alterations of the PI3K/AKT signaling. MAIN CONTENT A plethora of studies addressed the impact of AKT isoforms on tumor growth, metastasis and angiogenesis of breast cancer as well as on therapy response and overall survival in patients. Therefore, this review aimed to give a comprehensive overview about the isoform-specific effects of AKT in breast cancer and to summarize known downstream and upstream mechanisms. Taking account of conflicting findings among the studies, the majority of the studies reported a tumor initiating role of AKT1, whereas AKT2 is mainly responsible for tumor progression and metastasis. In detail, AKT1 increases cell proliferation through cell cycle proteins like p21, p27 and cyclin D1 and impairs apoptosis e.g. via p53. On the downside AKT1 decreases migration of breast cancer cells, for instance by regulating TSC2, palladin and EMT-proteins. However, AKT2 promotes migration and invasion most notably through regulation of β-integrins, EMT-proteins and F-actin. Whilst AKT3 is associated with a negative ER-status, findings about the role of AKT3 in regulation of the key properties of breast cancer are sparse. Accordingly, AKT1 is mutated and AKT2 is amplified in some cases of breast cancer and AKT isoforms are associated with overall survival and therapy response in an isoform-specific manner. CONCLUSIONS Although there are several discussed hypotheses how isoform specificity is achieved, the mechanisms behind the isoform-specific effects remain mostly unrevealed. As a consequence, further effort is necessary to achieve deeper insights into an isoform-specific AKT signaling in breast cancer and the mechanism behind it.
Collapse
Affiliation(s)
- Nico Hinz
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
17
|
Lv D, Song X, Huang B, Yu YZ, Shu F, Wang C, Chen H, Zhang HB, Zhao S. HMGB1 Promotes Prostate Cancer Development and Metastasis by Interacting with Brahma-Related Gene 1 and Activating the Akt Signaling Pathway. Am J Cancer Res 2019; 9:5166-5182. [PMID: 31410208 PMCID: PMC6691575 DOI: 10.7150/thno.33972] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022] Open
Abstract
Background and Aim: We have previously shown that high-mobility group box 1 (HMGB1) is an independent biomarker for shortened survival of prostate cancer (PCa) patients. However, the specific role of HMGB1 in tumor development and progression remains largely unknown. In this study, we investigated the molecular mechanisms of HMGB1 in PCa tumorigenesis. Methods: Gain-of-function and loss-of-function experiments were used to determine the biological functions of HMGB1 both in vitro and in vivo. Bioinformatic analysis, immunoprecipitation, and immunofluorescence assays were applied to discern and examine the relationship between HMGB1 and its potential targets. Specimens from 64 patients with PCa were analyzed for the expression of HMGB1 and its relationship with Brahma-related gene 1 (BRG1) was examined by immunohistochemistry. Results: The results demonstrated that ectopic expression of HMGB1 facilitated growth and metastasis of PCa by enhancing Akt signaling pathway and promoting epithelial-mesenchymal transition (EMT), while silencing of HMGB1 showed the opposite effects. Mechanistically, HMGB1 exerted these functions through its interaction with BRG1 which may augment BRG1 function and activate the Akt signaling pathway thereby promoting EMT. Importantly, both HMGB1 and BRG1 expression was markedly increased in human PCa tissues. Conclusions: Taken together, these findings indicate that upregulation of HMGB1 promotes PCa development via activation of Akt and accelerates metastasis through regulating BRG1-mediated EMT. HMGB1 could be used as a novel potential target for the treatment of PCa.
Collapse
|
18
|
Alwhaibi A, Verma A, Adil MS, Somanath PR. The unconventional role of Akt1 in the advanced cancers and in diabetes-promoted carcinogenesis. Pharmacol Res 2019; 145:104270. [PMID: 31078742 PMCID: PMC6659399 DOI: 10.1016/j.phrs.2019.104270] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/03/2019] [Accepted: 05/08/2019] [Indexed: 12/15/2022]
Abstract
Decades of research have elucidated the critical role of Akt isoforms in cancer as pro-tumorigenic and metastatic regulators through their specific effects on the cancer cells, tumor endothelial cells and the stromal cells. The pro-cancerous role of Akt isoforms through enhanced cell proliferation and suppression of apoptosis in cancer cells and the cells in the tumor microenvironment is considered a dogma. Intriguingly, studies also indicate that the Akt pathway is essential to protect the endothelial-barrier and prevent aberrant vascular permeability, which is also integral to tumor perfusion and metastasis. To complicate this further, a flurry of recent reports strongly indicates the metastasis suppressive role of Akt, Akt1 in particular in various cancer types. These reports emanated from different laboratories have elegantly demonstrated the paradoxical effect of Akt1 on cancer cell epithelial-to-mesenchymal transition, invasion, tumor endothelial-barrier disruption, and cancer metastasis. Here, we emphasize on the specific role of Akt1 in mediating tumor cell-vasculature reciprocity during the advanced stages of cancers and discuss how Akt1 differentially regulates cancer metastasis through mechanisms distinct from its pro-tumorigenic effects. Since Akt is integral for insulin signaling, endothelial function, and metabolic regulation, we also attempt to shed some light on the specific effects of diabetes in modulating Akt pathway in the promotion of tumor growth and metastasis.
Collapse
Affiliation(s)
- Abdulrahman Alwhaibi
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Arti Verma
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Mir S Adil
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Payaningal R Somanath
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and the Charlie Norwood VA Medical Center, Augusta, GA, USA; Department of Medicine, Vascular Biology Center and Cancer Center, Augusta University, USA.
| |
Collapse
|
19
|
Linton MF, Moslehi JJ, Babaev VR. Akt Signaling in Macrophage Polarization, Survival, and Atherosclerosis. Int J Mol Sci 2019; 20:ijms20112703. [PMID: 31159424 PMCID: PMC6600269 DOI: 10.3390/ijms20112703] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022] Open
Abstract
The PI3K/Akt pathway plays a crucial role in the survival, proliferation, and migration of macrophages, which may impact the development of atherosclerosis. Changes in Akt isoforms or modulation of the Akt activity levels in macrophages significantly affect their polarization phenotype and consequently atherosclerosis in mice. Moreover, the activity levels of Akt signaling determine the viability of monocytes/macrophages and their resistance to pro-apoptotic stimuli in atherosclerotic lesions. Therefore, elimination of pro-apoptotic factors as well as factors that antagonize or suppress Akt signaling in macrophages increases cell viability, protecting them from apoptosis, and this markedly accelerates atherosclerosis in mice. In contrast, inhibition of Akt signaling by the ablation of Rictor in myeloid cells, which disrupts mTORC2 assembly, significantly decreases the viability and proliferation of blood monocytes and macrophages with the suppression of atherosclerosis. In addition, monocytes and macrophages exhibit a threshold effect for Akt protein levels in their ability to survive. Ablation of two Akt isoforms, preserving only a single Akt isoform in myeloid cells, markedly compromises monocyte and macrophage viability, inducing monocytopenia and diminishing early atherosclerosis. These recent advances in our understanding of Akt signaling in macrophages in atherosclerosis may have significant relevance in the burgeoning field of cardio-oncology, where PI3K/Akt inhibitors being tested in cancer patients can have significant cardiovascular and metabolic ramifications.
Collapse
Affiliation(s)
- MacRae F Linton
- Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
| | - Javid J Moslehi
- Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
| | - Vladimir R Babaev
- Atherosclerosis Research Unit, Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232-6300, USA.
| |
Collapse
|
20
|
Mierke CT. The matrix environmental and cell mechanical properties regulate cell migration and contribute to the invasive phenotype of cancer cells. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2019; 82:064602. [PMID: 30947151 DOI: 10.1088/1361-6633/ab1628] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The minimal structural unit of a solid tumor is a single cell or a cellular compartment such as the nucleus. A closer look inside the cells reveals that there are functional compartments or even structural domains determining the overall properties of a cell such as the mechanical phenotype. The mechanical interaction of these living cells leads to the complex organization such as compartments, tissues and organs of organisms including mammals. In contrast to passive non-living materials, living cells actively respond to the mechanical perturbations occurring in their microenvironment during diseases such as fibrosis and cancer. The transformation of single cancer cells in highly aggressive and hence malignant cancer cells during malignant cancer progression encompasses the basement membrane crossing, the invasion of connective tissue, the stroma microenvironments and transbarrier migration, which all require the immediate interaction of the aggressive and invasive cancer cells with the surrounding extracellular matrix environment including normal embedded neighboring cells. All these steps of the metastatic pathway seem to involve mechanical interactions between cancer cells and their microenvironment. The pathology of cancer due to a broad heterogeneity of cancer types is still not fully understood. Hence it is necessary to reveal the signaling pathways such as mechanotransduction pathways that seem to be commonly involved in the development and establishment of the metastatic and mechanical phenotype in several carcinoma cells. We still do not know whether there exist distinct metastatic genes regulating the progression of tumors. These metastatic genes may then be activated either during the progression of cancer by themselves on their migration path or in earlier stages of oncogenesis through activated oncogenes or inactivated tumor suppressor genes, both of which promote the metastatic phenotype. In more detail, the adhesion of cancer cells to their surrounding stroma induces the generation of intracellular contraction forces that deform their microenvironments by alignment of fibers. The amplitude of these forces can adapt to the mechanical properties of the microenvironment. Moreover, the adhesion strength of cancer cells seems to determine whether a cancer cell is able to migrate through connective tissue or across barriers such as the basement membrane or endothelial cell linings of blood or lymph vessels in order to metastasize. In turn, exposure of adherent cancer cells to physical forces, such as shear flow in vessels or compression forces around tumors, reinforces cell adhesion, regulates cell contractility and restructures the ordering of the local stroma matrix that leads subsequently to secretion of crosslinking proteins or matrix degrading enzymes. Hence invasive cancer cells alter the mechanical properties of their microenvironment. From a mechanobiological point-of-view, the recognized physical signals are transduced into biochemical signaling events that guide cellular responses such as cancer progression after the malignant transition of cancer cells from an epithelial and non-motile phenotype to a mesenchymal and motile (invasive) phenotype providing cellular motility. This transition can also be described as the physical attempt to relate this cancer cell transitional behavior to a T1 phase transition such as the jamming to unjamming transition. During the invasion of cancer cells, cell adaptation occurs to mechanical alterations of the local stroma, such as enhanced stroma upon fibrosis, and therefore we need to uncover underlying mechano-coupling and mechano-regulating functional processes that reinforce the invasion of cancer cells. Moreover, these mechanisms may also be responsible for the awakening of dormant residual cancer cells within the microenvironment. Physicists were initially tempted to consider the steps of the cancer metastasis cascade as single events caused by a single mechanical alteration of the overall properties of the cancer cell. However, this general and simple view has been challenged by the finding that several mechanical properties of cancer cells and their microenvironment influence each other and continuously contribute to tumor growth and cancer progression. In addition, basement membrane crossing, cell invasion and transbarrier migration during cancer progression is explained in physical terms by applying physical principles on living cells regardless of their complexity and individual differences of cancer types. As a novel approach, the impact of the individual microenvironment surrounding cancer cells is also included. Moreover, new theories and models are still needed to understand why certain cancers are malignant and aggressive, while others stay still benign. However, due to the broad variety of cancer types, there may be various pathways solely suitable for specific cancer types and distinct steps in the process of cancer progression. In this review, physical concepts and hypotheses of cancer initiation and progression including cancer cell basement membrane crossing, invasion and transbarrier migration are presented and discussed from a biophysical point-of-view. In addition, the crosstalk between cancer cells and a chronically altered microenvironment, such as fibrosis, is discussed including the basic physical concepts of fibrosis and the cellular responses to mechanical stress caused by the mechanically altered microenvironment. Here, is highlighted how biophysical approaches, both experimentally and theoretically, have an impact on classical hallmarks of cancer and fibrosis and how they contribute to the understanding of the regulation of cancer and its progression by sensing and responding to the physical environmental properties through mechanotransduction processes. Finally, this review discusses various physical models of cell migration such as blebbing, nuclear piston, protrusive force and unjamming transition migration modes and how they contribute to cancer progression. Moreover, these cellular migration modes are influenced by microenvironmental perturbances such as fibrosis that can induce mechanical alterations in cancer cells, which in turn may impact the environment. Hence, the classical hallmarks of cancer need to be refined by including biomechanical properties of cells, cell clusters and tissues and their microenvironment to understand mechano-regulatory processes within cancer cells and the entire organism.
Collapse
|
21
|
Marcial-Medina C, Ordoñez-Moreno A, Gonzalez-Reyes C, Cortes-Reynosa P, Perez Salazar E. Oleic acid induces migration through a FFAR1/4, EGFR and AKT-dependent pathway in breast cancer cells. Endocr Connect 2019; 8:252-265. [PMID: 30721135 PMCID: PMC6410766 DOI: 10.1530/ec-18-0543] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 12/25/2022]
Abstract
Free fatty acids (FFAs) are an energy source, and induce activation of signal transduction pathways that mediate several biological processes. In breast cancer cells, oleic acid (OA) induces proliferation, matrix metalloproteinase-9 (MMP-9) secretion, migration and invasion. However, the signal transduction pathways that mediate migration and invasion induced by OA in breast cancer cells have not been studied in detail. We demonstrate here that FFAR1 and FFAR4 mediate migration induced by OA in MDA-MB-231 and MCF-7 breast cancer cells. Moreover, OA induces migration, invasion, AKT1 and AKT2 activation, 12-LOX secretion and an increase of NFκB-DNA binding activity in breast cancer cells. Cell migration requires FFAR1, FFAR4, EGFR, AKT and PI3K activity, whereas invasion is mediated though a PI3K/Akt-dependent pathway. Furthermore, OA promotes relocalization of paxillin to focal contacts and it requires PI3K and EGFR activity, whereas NFκB-DNA binding activity requires PI3K and AKT activity.
Collapse
Affiliation(s)
| | | | | | | | - Eduardo Perez Salazar
- Departamento de Biologia Celular, Cinvestav-IPN, Mexico City, Mexico
- Correspondence should be addressed to E Perez Salazar:
| |
Collapse
|
22
|
Akt1 inhibition promotes breast cancer metastasis through EGFR-mediated β-catenin nuclear accumulation. Cell Commun Signal 2018; 16:82. [PMID: 30445978 PMCID: PMC6240210 DOI: 10.1186/s12964-018-0295-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022] Open
Abstract
Background Knockdown of Akt1 promotes Epithelial-to-Mesenchymal Transition in breast cancer cells. However, the mechanisms are not completely understood. Methods Western blotting, immunofluorescence, luciferase assay, real time PCR, ELISA and Matrigel invasion assay were used to investigate how Akt1 inhibition promotes breast cancer cell invasion in vitro. Mouse model of lung metastasis was used to measure in vivo efficacy of Akt inhibitor MK2206 and its combination with Gefitinib. Results Knockdown of Akt1 stimulated β-catenin nuclear accumulation, resulting in breast cancer cell invasion. β-catenin nuclear accumulation induced by Akt1 inhibition depended on the prolonged activation of EGFR signaling pathway in breast cancer cells. Mechanistic experiments documented that knockdown of Akt1 inactivates PIKfyve via dephosphorylating of PIKfyve at Ser318 site, resulting in a decreased degradation of EGFR signaling pathway. Inhibition of Akt1 using MK2206 could induce an increase in the expression of EGFR and β-catenin in breast cancer cells. In addition, MK2206 at a low dosage enhance breast cancer metastasis in a mouse model of lung metastasis, while an inhibitor of EGFR tyrosine kinase Gefitinib could potentially suppress breast cancer metastasis induced by Akt1 inhibition. Conclusion EGFR-mediated β-catenin nuclear accumulation is critical for Akt1 inhibition-induced breast cancer metastasis.
Collapse
|
23
|
Vilardell J, Girardi C, Marin O, Cozza G, Pinna LA, Ruzzene M. The importance of negative determinants as modulators of CK2 targeting. The lesson of Akt2 S131. PLoS One 2018; 13:e0193479. [PMID: 29494643 PMCID: PMC5832243 DOI: 10.1371/journal.pone.0193479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/12/2018] [Indexed: 01/26/2023] Open
Abstract
CK2 is a pleiotropic S/T protein kinase (formerly known as casein kinase 2) which is attracting increasing interest as therapeutic target, and the identification of its substrates is a crucial step in determining its involvement in different pathological conditions. We recently found that S131 of Akt2 (homologous to the well established CK2 target S129 of Akt1) is not phosphorylated by CK2 either in vitro or in vivo, although the consensus sequence recognized by CK2 (S/T-x-x-E/D/pS/pT) is conserved in it. Here, by exploiting synthetic peptides, in cell transfection experiments, and computational analysis, we show that a single sequence element, a T at position n+1, hampers phosphorylation, causing an α-helix structure organization which prevents the recognition of its own consensus by CK2. Our results highlight the role of negative determinants as crucial modulators of CK2 targeting and corroborate the concept that Akt1 and Akt2 display isoform specific features. Experiments with synthetic peptides suggest that Akt2 S131 could be phosphorylated by kinases of the Plk (Polo-like kinase) family, which are insensitive to the presence of the n+1 T. The low phylogenetic conservation of the Akt2 sequence around S131, as opposed to the extremely well-conserved Akt1 homologous sequence, would indicate a dominant positive role in the selective pressure only for the Akt1 phosphoacceptor site committed to undergo phosphorylation by CK2. By contrast, Akt2 S131 may mediate the response to specific physio/pathological conditions, being consequently shielded against basal CK2 targeting.
Collapse
Affiliation(s)
- Jordi Vilardell
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Cristina Girardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Oriano Marin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giorgio Cozza
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Lorenzo A. Pinna
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CNR Neuroscience Institute, Padova, Italy
| | - Maria Ruzzene
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CNR Neuroscience Institute, Padova, Italy
- * E-mail:
| |
Collapse
|
24
|
Linnerth-Petrik NM, Santry LA, Moorehead R, Jücker M, Wootton SK, Petrik J. Akt isoform specific effects in ovarian cancer progression. Oncotarget 2018; 7:74820-74833. [PMID: 27533079 PMCID: PMC5342704 DOI: 10.18632/oncotarget.11204] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/27/2016] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer remains a significant therapeutic problem and novel, effective therapies are needed. Akt is a serine-threonine kinase that is overexpressed in numerous cancers, including ovarian. Mammalian cells express three Akt isoforms which are encoded by distinct genes. Although there are several Akt inhibitors in clinical trials, most indiscriminately target all isoforms. Current in vitro data and animal knockout experiments suggest that the Akt isoforms may have divergent roles. In this paper, we determined the isoform-specific functions of Akt in ovarian cancer cell proliferation in vitro and in ovarian cancer progression in vivo. For in vitro experiments, murine and human ovarian cancer cells were treated with Akt inhibitors and cell viability was assessed. We used two different in vivo approaches to identify the roles of Akt isoforms in ovarian cancer progression and their influence on the primary tumor and tumor microenvironment. In one experiment, wild-type C57Bl6 mice were orthotopically injected with ID8 cells with stable knockdown of Akt isoforms. In a separate experiment, mice null for Akt 1-3 were orthotopically injected with WT ID8 cells (Figure 1). Our data show that inhibition of Akt1 significantly reduced ovarian cancer cell proliferation and inhibited tumor progression in vivo. Conversely, disruption of Akt2 increased tumor growth. Inhibition of Akt3 had an intermediate phenotype, but also increased growth of ovarian cancer cells. These data suggest that there is minimal redundancy between the Akt isoforms in ovarian cancer progression. These findings have important implications in the design of Akt inhibitors for the effective treatment of ovarian cancer.
Collapse
Affiliation(s)
| | - Lisa A Santry
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Roger Moorehead
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Manfred Jücker
- Center of Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah K Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
25
|
Jin Y, Xie Y, Ostriker AC, Zhang X, Liu R, Lee MY, Leslie KL, Tang W, Du J, Lee SH, Wang Y, Sessa WC, Hwa J, Yu J, Martin KA. Opposing Actions of AKT (Protein Kinase B) Isoforms in Vascular Smooth Muscle Injury and Therapeutic Response. Arterioscler Thromb Vasc Biol 2017; 37:2311-2321. [PMID: 29025710 PMCID: PMC5699966 DOI: 10.1161/atvbaha.117.310053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/26/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Drug-eluting stent delivery of mTORC1 (mechanistic target of rapamycin complex 1) inhibitors is highly effective in preventing intimal hyperplasia after coronary revascularization, but adverse effects limit their use for systemic vascular disease. Understanding the mechanism of action may lead to new treatment strategies. We have shown that rapamycin promotes vascular smooth muscle cell differentiation in an AKT2-dependent manner in vitro. Here, we investigate the roles of AKT (protein kinase B) isoforms in intimal hyperplasia. APPROACH AND RESULTS We found that germ-line-specific or smooth muscle-specific deletion of Akt2 resulted in more severe intimal hyperplasia compared with control mice after arterial denudation injury. Conversely, smooth muscle-specific Akt1 knockout prevented intimal hyperplasia, whereas germ-line Akt1 deletion caused severe thrombosis. Notably, rapamycin prevented intimal hyperplasia in wild-type mice but had no therapeutic benefit in Akt2 knockouts. We identified opposing roles for AKT1 and AKT2 isoforms in smooth muscle cell proliferation, migration, differentiation, and rapamycin response in vitro. Mechanistically, rapamycin induced MYOCD (myocardin) mRNA expression. This was mediated by AKT2 phosphorylation and nuclear exclusion of FOXO4 (forkhead box O4), inhibiting its binding to the MYOCD promoter. CONCLUSIONS Our data reveal opposing roles for AKT isoforms in smooth muscle cell remodeling. AKT2 is required for rapamycin's therapeutic inhibition of intimal hyperplasia, likely mediated in part through AKT2-specific regulation of MYOCD via FOXO4. Because AKT2 signaling is impaired in diabetes mellitus, this work has important implications for rapamycin therapy, particularly in diabetic patients.
Collapse
MESH Headings
- Animals
- Binding Sites
- Cell Cycle Proteins
- Cell Differentiation/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Forkhead Transcription Factors
- Gene Expression Regulation
- Genetic Predisposition to Disease
- Humans
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neointima
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-akt/deficiency
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Sirolimus/pharmacology
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transfection
- Vascular System Injuries/enzymology
- Vascular System Injuries/genetics
- Vascular System Injuries/pathology
- Vascular System Injuries/prevention & control
Collapse
Affiliation(s)
- Yu Jin
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Yi Xie
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Allison C Ostriker
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Xinbo Zhang
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Renjing Liu
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Monica Y Lee
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Kristen L Leslie
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Waiho Tang
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Jing Du
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Seung Hee Lee
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Yingdi Wang
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - William C Sessa
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - John Hwa
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Jun Yu
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - Kathleen A Martin
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (Y.J., Y.X., A.C.O., K.L.L., W.T., J.D., S.H.L., Y.W., J.H., K.A.M.) and Department of Pharmacology (Y.J., Y.X., A.C.O., M.Y.L., K.L.L., W.C.S., K.A.M.), Yale University, New Haven, CT; Section of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, CT (X.Z.); Agnes Ginges Laboratory for Diseases of the Aorta, Centenary Institute, University of Sydney, Camperdown, Australia (R.L.); Sydney Medical School, University of Sydney, Sydney, Australia (R.L.); and Department of Physiology and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.).
| |
Collapse
|
26
|
Palladin is a novel microtubule-associated protein responsible for spindle orientation. Sci Rep 2017; 7:11806. [PMID: 28924223 PMCID: PMC5603589 DOI: 10.1038/s41598-017-12051-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/25/2017] [Indexed: 11/26/2022] Open
Abstract
Mitotic spindles, which consist of microtubules (MTs) and associated proteins, play critical roles in controlling cell division and maintaining tissue homeostasis. The orientation of the mitotic spindle is closely related with the duration of mitosis. However, the molecular mechanism in regulating the orientation of the mitotic spindles is largely undefined. In this study, we found that Palladin is a novel MT-associated protein and regulator of spindle orientation, which maintains proper spindle orientation by stabilizing astral MTs. Palladin depletion distorted spindle orientation, prolonged the metaphase, and impaired proliferation of HeLa cells. Results showed that Palladin depletion-induced spindle misorientation and astral MT instability could be rescued by constitutively active AKT1 or dominant negative GSK3β. Our findings revealed that Palladin regulates spindle orientation and mitotic progression mainly through the AKT1–GSK3β pathway.
Collapse
|
27
|
Wang L, Huang D, Jiang Z, Luo Y, Norris C, Zhang M, Tian X, Tang Y. Akt3 is responsible for the survival and proliferation of embryonic stem cells. Biol Open 2017; 6:850-861. [PMID: 28483982 PMCID: PMC5483023 DOI: 10.1242/bio.024505] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/24/2017] [Indexed: 12/21/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/Akt) pathway plays an important role in regulating cell proliferation, metabolism, and survival. However, the distinct roles of Akt isoforms (Akt1, Akt2, and Akt3) in pluripotent stem cell maintenance are not fully defined. Using mouse embryonic stem cells (ESCs), we show that direct inhibition of Akt activity leads to ESC apoptosis. The Akt3, but not Akt1 or Akt2, activity specifically regulates this effect. Inhibiting Akt3 also leads to a cell cycle arrest at G1 phase. These regulatory roles of Akt3 are dependent on its kinase activity. Blocking the expression of Akt1 plus Akt2 in ESCs does not affect cell survival or proliferation, although blocking Akt1 aggravates the apoptotic effect induced by depletion of Akt3. We further show that blocking Akt3 in ESCs results in significant nuclear accumulation of p53, as well as the activation of its downstream targets, such as Mdm2, p21, and Fas. Inhibiting p53 and its downstream targets partially rescued the effects caused by Akt3-depletion. Our results revealed an Akt3 isoform-specific mechanism for ESC survival and proliferation involving the control of p53 activity.
Collapse
Affiliation(s)
- Ling Wang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Delun Huang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
- Animal Reproduction Institute, Guangxi University, Nanning, 530004, People's Republic of China
| | - Zongliang Jiang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Yan Luo
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Carol Norris
- Center for Open Research Resources and Equipment, University of Connecticut, Storrs, CT 06269, USA
| | - Ming Zhang
- Animal Reproduction Institute, Guangxi University, Nanning, 530004, People's Republic of China
| | - Xiuchun Tian
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Young Tang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
28
|
Vitting-Seerup K, Sandelin A. The Landscape of Isoform Switches in Human Cancers. Mol Cancer Res 2017; 15:1206-1220. [PMID: 28584021 DOI: 10.1158/1541-7786.mcr-16-0459] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/17/2017] [Accepted: 05/25/2017] [Indexed: 11/16/2022]
Abstract
Alternative usage of transcript isoforms from the same gene has been hypothesized as an important feature in cancers. However, differential usage of gene transcripts between conditions (isoform switching) has not been comprehensively characterized in and across cancer types. To this end, we developed methods for identification and visualization of isoform switches with predicted functional consequences. Using these methods, we characterized isoform switching in RNA-seq data from >5,500 cancer patients covering 12 solid cancer types. Isoform switches with potential functional consequences were common, affecting approximately 19% of multiple transcript genes. Among these, isoform switches leading to loss of DNA sequence encoding protein domains were more frequent than expected, particularly in pancancer switches. We identified several isoform switches as powerful biomarkers: 31 switches were highly predictive of patient survival independent of cancer types. Our data constitute an important resource for cancer researchers, available through interactive web tools. Moreover, our methods, available as an R package, enable systematic analysis of isoform switches from other RNA-seq datasets.Implications: This study indicates that isoform switches with predicted functional consequences are common and important in dysfunctional cells, which in turn means that gene expression should be analyzed at the isoform level. Visual Overview: http://mcr.aacrjournals.org/content/molcanres/15/9/1206/F1.large.jpg.Mol Cancer Res; 15(9); 1206-20. ©2017 AACR.
Collapse
Affiliation(s)
- Kristoffer Vitting-Seerup
- The Bioinformatics Centre, Department of Biology and Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Albin Sandelin
- The Bioinformatics Centre, Department of Biology and Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Olsen SN, Wronski A, Castaño Z, Dake B, Malone C, De Raedt T, Enos M, DeRose YS, Zhou W, Guerra S, Loda M, Welm A, Partridge AH, McAllister SS, Kuperwasser C, Cichowski K. Loss of RasGAP Tumor Suppressors Underlies the Aggressive Nature of Luminal B Breast Cancers. Cancer Discov 2017; 7:202-217. [PMID: 27974415 PMCID: PMC6461361 DOI: 10.1158/2159-8290.cd-16-0520] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 12/31/2022]
Abstract
Luminal breast cancers are typically estrogen receptor-positive and generally have the best prognosis. However, a subset of luminal tumors, namely luminal B cancers, frequently metastasize and recur. Unfortunately, the causal events that drive their progression are unknown, and therefore it is difficult to identify individuals who are likely to relapse and should receive escalated treatment. Here, we identify a bifunctional RasGAP tumor suppressor whose expression is lost in almost 50% of luminal B tumors. Moreover, we show that two RasGAP genes are concomitantly suppressed in the most aggressive luminal malignancies. Importantly, these genes cooperatively regulate two major oncogenic pathways, RAS and NF-κB, through distinct domains, and when inactivated drive the metastasis of luminal tumors in vivo Finally, although the cooperative effects on RAS drive invasion, NF-κB activation triggers epithelial-to-mesenchymal transition and is required for metastasis. Collectively, these studies reveal important mechanistic insight into the pathogenesis of luminal B tumors and provide functionally relevant prognostic biomarkers that may guide treatment decisions. SIGNIFICANCE The lack of insight into mechanisms that underlie the aggressive behavior of luminal B breast cancers impairs treatment decisions and therapeutic advances. Here, we show that two RasGAP tumor suppressors are concomitantly suppressed in aggressive luminal B tumors and demonstrate that they drive metastasis by activating RAS and NF-κB. Cancer Discov; 7(2); 202-17. ©2016 AACR.See related commentary by Sears and Gray, p. 131This article is highlighted in the In This Issue feature, p. 115.
Collapse
Affiliation(s)
- Sarah Naomi Olsen
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ania Wronski
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - Zafira Castaño
- Harvard Medical School, Boston, Massachusetts
- Hematology Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Broad Institute of Harvard and MIT, Cambridge, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Benjamin Dake
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - Clare Malone
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Thomas De Raedt
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Miriam Enos
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Wenhui Zhou
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - Stephanie Guerra
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Massimo Loda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alana Welm
- Huntsman Cancer Institute, Salt Lake City, Utah
| | - Ann H Partridge
- Harvard Medical School, Boston, Massachusetts
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sandra S McAllister
- Harvard Medical School, Boston, Massachusetts
- Hematology Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Broad Institute of Harvard and MIT, Cambridge, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Charlotte Kuperwasser
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - Karen Cichowski
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
- Huntsman Cancer Institute, Salt Lake City, Utah
| |
Collapse
|
30
|
Cui X, Guo W, Sun Y, Sun B, Hu S, Sun D, Lam RHW. A microfluidic device for isolation and characterization of transendothelial migrating cancer cells. BIOMICROFLUIDICS 2017; 11:014105. [PMID: 28798840 PMCID: PMC5533502 DOI: 10.1063/1.4974012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/02/2017] [Indexed: 05/30/2023]
Abstract
Transendothelial migration of cancer cells is a critical stage in cancer, including breast cancer, as the migrating cells are generally believed to be highly metastatic. However, it is still challenging for many existing platforms to achieve a fully covering endothelium and to ensure transendothelial migration capability of the extracted cancer cells for analyses with high specificity. Here, we report a microfluidic device containing multiple independent cell collection microchambers underneath an embedded endothelium such that the transendothelial-migrated cells can be selectively collected from only the microchambers with full coverage of an endothelial layer. In this work, we first optimize the pore size of a microfabricated supporting membrane for the endothelium formation. We quantify transendothelial migration rates of a malignant human breast cell type (MDA-MB-231) under different shear stress levels. We investigate characteristics of the migrating cells including morphology, cytoskeletal structures, and migration (speed and persistence). Further implementation of this endothelium-embedded microfluidic device can provide important insights into migration and intracellular characteristics related to cancer metastasis and strategies for effective cancer therapy.
Collapse
Affiliation(s)
- Xin Cui
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Weijin Guo
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01002, USA
| | - Baoce Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Shuhuan Hu
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | | | | |
Collapse
|
31
|
Linton MF, Babaev VR, Huang J, Linton EF, Tao H, Yancey PG. Macrophage Apoptosis and Efferocytosis in the Pathogenesis of Atherosclerosis. Circ J 2016; 80:2259-2268. [PMID: 27725526 DOI: 10.1253/circj.cj-16-0924] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Macrophage apoptosis and the ability of macrophages to clean up dead cells, a process called efferocytosis, are crucial determinants of atherosclerosis lesion progression and plaque stability. Environmental stressors initiate endoplasmic reticulum (ER) stress and activate the unfolded protein response (UPR). Unresolved ER stress with activation of the UPR initiates apoptosis. Macrophages are resistant to apoptotic stimuli, because of activity of the PI3K/Akt pathway. Macrophages express 3 Akt isoforms, Akt1, Akt2 and Akt3, which are products of distinct but homologous genes. Akt displays isoform-specific effects on atherogenesis, which vary with different vascular cell types. Loss of macrophage Akt2 promotes the anti-inflammatory M2 phenotype and reduces atherosclerosis. However, Akt isoforms are redundant with regard to apoptosis. c-Jun NH2-terminal kinase (JNK) is a pro-apoptotic effector of the UPR, and the JNK1 isoform opposes anti-apoptotic Akt signaling. Loss of JNK1 in hematopoietic cells protects macrophages from apoptosis and accelerates early atherosclerosis. IκB kinase α (IKKα, a member of the serine/threonine protein kinase family) plays an important role in mTORC2-mediated Akt signaling in macrophages, and IKKα deficiency reduces macrophage survival and suppresses early atherosclerosis. Efferocytosis involves the interaction of receptors, bridging molecules, and apoptotic cell ligands. Scavenger receptor class B type I is a critical mediator of macrophage efferocytosis via the Src/PI3K/Rac1 pathway in atherosclerosis. Agonists that resolve inflammation offer promising therapeutic potential to promote efferocytosis and prevent atherosclerotic clinical events. (Circ J 2016; 80: 2259-2268).
Collapse
Affiliation(s)
- MacRae F Linton
- Atherosclerosis Research Unit, Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
| | | | | | | | | | | |
Collapse
|
32
|
Regulation of PtdIns(3,4,5)P3/Akt signalling by inositol polyphosphate 5-phosphatases. Biochem Soc Trans 2016; 44:240-52. [DOI: 10.1042/bst20150214] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The phosphoinositide 3-kinase (PI3K) generated lipid signals, PtdIns(3,4,5)P3 and PtdIns(3,4)P2, are both required for the maximal activation of the serine/threonine kinase proto-oncogene Akt. The inositol polyphosphate 5-phosphatases (5-phosphatases) hydrolyse the 5-position phosphate from the inositol head group of PtdIns(3,4,5)P3 to yield PtdIns(3,4)P2. Extensive work has revealed several 5-phosphatases inhibit PI3K-driven Akt signalling, by decreasing PtdIns(3,4,5)P3 despite increasing cellular levels of PtdIns(3,4)P2. The roles that 5-phosphatases play in suppressing cell proliferation and transformation are slow to emerge; however, the 5-phosphatase PIPP [proline-rich inositol polyphosphate 5-phosphatase; inositol polyphosphate 5-phosphatase (INPP5J)] has recently been identified as a putative tumour suppressor in melanoma and breast cancer and SHIP1 [SH2 (Src homology 2)-containing inositol phosphatase 1] inhibits haematopoietic cell proliferation. INPP5E regulates cilia stability and INPP5E mutations have been implicated ciliopathy syndromes. This review will examine 5-phosphatase regulation of PI3K/Akt signalling, focussing on the role PtdIns(3,4,5)P3 5-phosphatases play in developmental diseases and cancer.
Collapse
|
33
|
Grottke A, Ewald F, Lange T, Nörz D, Herzberger C, Bach J, Grabinski N, Gräser L, Höppner F, Nashan B, Schumacher U, Jücker M. Downregulation of AKT3 Increases Migration and Metastasis in Triple Negative Breast Cancer Cells by Upregulating S100A4. PLoS One 2016; 11:e0146370. [PMID: 26741489 PMCID: PMC4704820 DOI: 10.1371/journal.pone.0146370] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/16/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Treatment of breast cancer patients with distant metastases represents one of the biggest challenges in today's gynecological oncology. Therefore, a better understanding of mechanisms promoting the development of metastases is of paramount importance. The serine/threonine kinase AKT was shown to drive cancer progression and metastasis. However, there is emerging data that single AKT isoforms (i.e. AKT1, AKT2 and AKT3) have different or even opposing functions in the regulation of cancer cell migration in vitro, giving rise to the hypothesis that inhibition of distinct AKT isoforms might have undesirable effects on cancer dissemination in vivo. METHODS The triple negative breast cancer cell line MDA-MB-231 was used to investigate the functional roles of AKT in migration and metastasis. AKT single and double knockdown cells were generated using isoform specific shRNAs. Migration was analyzed using live cell imaging, chemotaxis and transwell assays. The metastatic potential of AKT isoform knockdown cells was evaluated in a subcutaneous xenograft mouse model in vivo. RESULTS Depletion of AKT3, but not AKT1 or AKT2, resulted in increased migration in vitro. This effect was even more prominent in AKT2,3 double knockdown cells. Furthermore, combined downregulation of AKT2 and AKT3, as well as AKT1 and AKT3 significantly increased metastasis formation in vivo. Screening for promigratory proteins revealed that downregulation of AKT3 increases the expression of S100A4 protein. In accordance, depletion of S100A4 by siRNA approach reverses the increased migration induced by knockdown of AKT3. CONCLUSIONS We demonstrated that knockdown of AKT3 can increase the metastatic potential of triple negative breast cancer cells. Therefore, our results provide a rationale for the development of AKT isoform specific inhibitors.
Collapse
Affiliation(s)
- Astrid Grottke
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Florian Ewald
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Tobias Lange
- Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Dominik Nörz
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Christiane Herzberger
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Johanna Bach
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Nicole Grabinski
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Lareen Gräser
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Frank Höppner
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Björn Nashan
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Udo Schumacher
- Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Manfred Jücker
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- * E-mail:
| |
Collapse
|
34
|
Cohen DPA, Martignetti L, Robine S, Barillot E, Zinovyev A, Calzone L. Mathematical Modelling of Molecular Pathways Enabling Tumour Cell Invasion and Migration. PLoS Comput Biol 2015; 11:e1004571. [PMID: 26528548 PMCID: PMC4631357 DOI: 10.1371/journal.pcbi.1004571] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 09/29/2015] [Indexed: 02/07/2023] Open
Abstract
Understanding the etiology of metastasis is very important in clinical perspective, since it is estimated that metastasis accounts for 90% of cancer patient mortality. Metastasis results from a sequence of multiple steps including invasion and migration. The early stages of metastasis are tightly controlled in normal cells and can be drastically affected by malignant mutations; therefore, they might constitute the principal determinants of the overall metastatic rate even if the later stages take long to occur. To elucidate the role of individual mutations or their combinations affecting the metastatic development, a logical model has been constructed that recapitulates published experimental results of known gene perturbations on local invasion and migration processes, and predict the effect of not yet experimentally assessed mutations. The model has been validated using experimental data on transcriptome dynamics following TGF-β-dependent induction of Epithelial to Mesenchymal Transition in lung cancer cell lines. A method to associate gene expression profiles with different stable state solutions of the logical model has been developed for that purpose. In addition, we have systematically predicted alleviating (masking) and synergistic pairwise genetic interactions between the genes composing the model with respect to the probability of acquiring the metastatic phenotype. We focused on several unexpected synergistic genetic interactions leading to theoretically very high metastasis probability. Among them, the synergistic combination of Notch overexpression and p53 deletion shows one of the strongest effects, which is in agreement with a recent published experiment in a mouse model of gut cancer. The mathematical model can recapitulate experimental mutations in both cell line and mouse models. Furthermore, the model predicts new gene perturbations that affect the early steps of metastasis underlying potential intervention points for innovative therapeutic strategies in oncology.
Collapse
Affiliation(s)
- David P. A. Cohen
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Loredana Martignetti
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Sylvie Robine
- Institut Curie, Paris, France
- CNRS UMR144, Paris, France
| | - Emmanuel Barillot
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Andrei Zinovyev
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
| | - Laurence Calzone
- Institut Curie, Paris, France
- INSERM, U900, Paris, France
- Mines ParisTech, Fontainebleau, Paris, France
- * E-mail:
| |
Collapse
|
35
|
Fallah S, Korani M, Hajimirza M, Seifi M. Association Between Genetic Variants of Akt1 and Endometrial Cancer. Biochem Genet 2015; 53:281-90. [PMID: 26296520 DOI: 10.1007/s10528-015-9690-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 08/16/2015] [Indexed: 12/29/2022]
Abstract
Akt isoforms have critical roles in the cause and regulation of cancer cells invasive, migration, and metastatic dissemination. In the present study, the association between Akt1 polymorphisms and endometrial cancer was investigated in patients with endometrial cancer and controls. Thirty premenopaused patients diagnosed with endometrial cancer and 30 premenopaused women with no clinically documented abnormalities of the endometrium undergoing hysterectomy were included in this study. Genotypes were determined by polymerase chain reaction and restriction fragment length polymorphism. There was no significant difference between Akt1 gene polymorphisms of patients (SNP1, SNP2 and SNP3) with endometrial cancer and controls (p > 0.05). Difference between alleles frequency of SNP1, SNP2, SNP3 of patients with endometrial cancer and controls was not significant (p > 0.05). SNPs (rs72715985), (rs2494750), and (rs74090038) of Akt1 gene are not associated with endometrial cancer in Iranian subjects.
Collapse
Affiliation(s)
- Soudabeh Fallah
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, 14155-5983, Tehran, Iran. .,Research Center of Pediatric Infectious Disease, Rasool Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohsen Korani
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Milad Hajimirza
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, 14155-5983, Tehran, Iran
| | - Morteza Seifi
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
36
|
Guidetti GF, Canobbio I, Torti M. PI3K/Akt in platelet integrin signaling and implications in thrombosis. Adv Biol Regul 2015; 59:36-52. [PMID: 26159296 DOI: 10.1016/j.jbior.2015.06.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/04/2015] [Accepted: 06/04/2015] [Indexed: 01/09/2023]
Abstract
Blood platelets are anucleated circulating cells that play a critical role in hemostasis and are also implicated in arterial thrombosis, a major cause of death worldwide. The biological function of platelets strongly relies in their reactiveness to a variety of extracellular agonists that regulate their adhesion to extracellular matrix at the site of vascular injury and their ability to form rapidly growing cell aggregates. Among the membrane receptors expressed on the cell surface, integrins are crucial for both platelet activation, adhesion and aggregation. Integrin affinity for specific ligands is regulated by intracellular signaling pathways activated in stimulated platelets, and, once engaged, integrins themselves generate and propagate signals inside the cells to reinforce and consolidate platelet response and thrombus formation. Phosphatidylinositol 3-Kinases (PI3Ks) have emerged as crucial players in platelet activation, and they are directly implicated in the regulation of integrin function. This review will discuss the contribution of PI3Ks in platelet integrin signaling, focusing on the role of specific members of class I PI3Ks and their downstream effector Akt on both integrin inside-out and outside-in signaling. The contribution of the PI3K/Akt pathways stimulated by integrin engagement and platelet activation in thrombus formation and stabilization will also be discussed in order to highlight the possibility to target these enzymes in effective anti-thrombotic therapeutic strategies.
Collapse
Affiliation(s)
- Gianni F Guidetti
- Department of Biology and Biotechnology, Laboratories of Biochemistry, University of Pavia, Pavia, Italy
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, Laboratories of Biochemistry, University of Pavia, Pavia, Italy
| | - Mauro Torti
- Department of Biology and Biotechnology, Laboratories of Biochemistry, University of Pavia, Pavia, Italy.
| |
Collapse
|
37
|
Abstract
Aberrant activation of fundamental cellular processes, such as proliferation, migration and survival, underlies the development of numerous human pathophysiologies, including cancer. One of the most frequently hyperactivated pathways in cancer is the phosphoinositide 3-kinase (PI3K)/Akt signalling cascade. Three isoforms of the serine/threonine protein kinase Akt (Akt1, Akt2 and Akt3) function to regulate cell survival, growth, proliferation and metabolism. Strikingly, non-redundant and even opposing functions of Akt isoforms in the regulation of phenotypes associated with malignancy in humans have been described. However, the mechanisms by which Akt isoform-specificity is conferred are largely unknown. In the present review, we highlight recent findings that have contributed to our understanding of the complexity of Akt isoform-specific signalling and discussed potential mechanisms by which this isoform-specificity is conferred. An understanding of the mechanisms of Akt isoform-specificity has important implications for the development of isoform-specific Akt inhibitors and will be critical to finding novel targets to treat disease.
Collapse
|
38
|
Xie Y, Jin Y, Merenick BL, Ding M, Fetalvero KM, Wagner RJ, Mai A, Gleim S, Tucker DF, Birnbaum MJ, Ballif BA, Luciano AK, Sessa WC, Rzucidlo EM, Powell RJ, Hou L, Zhao H, Hwa J, Yu J, Martin KA. Phosphorylation of GATA-6 is required for vascular smooth muscle cell differentiation after mTORC1 inhibition. Sci Signal 2015; 8:ra44. [PMID: 25969542 DOI: 10.1126/scisignal.2005482] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Vascular smooth muscle cells (VSMCs) undergo transcriptionally regulated reversible differentiation in growing and injured blood vessels. This dedifferentiation also contributes to VSMC hyperplasia after vascular injury, including that caused by angioplasty and stenting. Stents provide mechanical support and can contain and release rapamycin, an inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1). Rapamycin suppresses VSMC hyperplasia and promotes VSMC differentiation. We report that rapamycin-induced differentiation of VSMCs required the transcription factor GATA-6. Inhibition of mTORC1 stabilized GATA-6 and promoted the nuclear accumulation of GATA-6, its binding to DNA, its transactivation of promoters encoding contractile proteins, and its inhibition of proliferation. These effects were mediated by phosphorylation of GATA-6 at Ser(290), potentially by Akt2, a kinase that is activated in VSMCs when mTORC1 is inhibited. Rapamycin induced phosphorylation of GATA-6 in wild-type mice, but not in Akt2(-/-) mice. Intimal hyperplasia after arterial injury was greater in Akt2(-/-) mice than in wild-type mice, and the exacerbated response in Akt2(-/-) mice was rescued to a greater extent by local overexpression of the wild-type or phosphomimetic (S290D) mutant GATA-6 than by that of the phosphorylation-deficient (S290A) mutant. Our data indicated that GATA-6 and Akt2 are involved in the mTORC1-mediated regulation of VSMC proliferation and differentiation. Identifying the downstream transcriptional targets of mTORC1 may provide cell type-specific drug targets to combat cardiovascular diseases associated with excessive proliferation of VSMCs.
Collapse
Affiliation(s)
- Yi Xie
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Yu Jin
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Bethany L Merenick
- Department of Pharmacology and Toxicology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA. Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Min Ding
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA. Department of Pharmacology and Toxicology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA. Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Kristina M Fetalvero
- Department of Pharmacology and Toxicology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA. Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Robert J Wagner
- Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Alice Mai
- Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Scott Gleim
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - David F Tucker
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Morris J Birnbaum
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT 05405, USA
| | - Amelia K Luciano
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - William C Sessa
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Eva M Rzucidlo
- Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Richard J Powell
- Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Lin Hou
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA. Department of Pharmacology and Toxicology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Jun Yu
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Vascular Biology and Therapeutics Program, and Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT 06511, USA. Department of Pharmacology and Toxicology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA. Department of Surgery, Section of Vascular Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA.
| |
Collapse
|
39
|
Mok KW, Chen H, Lee WM, Cheng CY. rpS6 regulates blood-testis barrier dynamics through Arp3-mediated actin microfilament organization in rat sertoli cells. An in vitro study. Endocrinology 2015; 156:1900-13. [PMID: 25714812 PMCID: PMC4398761 DOI: 10.1210/en.2014-1791] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the seminiferous epithelium of rat testes, preleptotene spermatocytes residing in the basal compartment are transported across the blood-testis barrier (BTB) to enter the adluminal compartment at stage VIII of the epithelial cycle. This process involves redistribution of tight junction (TJ) proteins via reorganization of actin cytoskeleton in Sertoli cells that serves as attachment site for adhesion protein complexes. Ribosomal protein S6 (rpS6), a downstream molecule of mTORC1 (mammalian target of rapamycin complex 1), participates in this process via a yet-to-be defined mechanism. Here, we constructed an rpS6 quadruple phosphomimetic mutant by converting Ser residues at 235, 236, 240, and 244 to Glu via site-directed mutagenesis, making this mutant constitutively active. When this rpS6 mutant was overexpressed in Sertoli cells cultured in vitro with an established TJ barrier mimicking the BTB in vivo, it perturbed the TJ permeability by down-regulating and redistributing TJ proteins at the cell-cell interface. These changes are mediated by a reorganization of actin microfilaments, which was triggered by a redistribution of activated actin-related protein 3 (Arp3) as well as changes in Arp3-neuronal Wiskott-Aldrich Syndrome protein (N-WASP) interaction. This in turn induced reorganization of actin microfilaments, converting them from a "bundled" to an "unbundled/branched" configuration, concomitant with a reduced actin bundling activity, thereby destabilizing the TJ-barrier function. These changes were mediated by Akt (transforming oncogene of v-akt), because an Akt knockdown by RNA interference was able to mimic the phenotypes of rpS6 mutant overexpression at the Sertoli cell BTB. In summary, this study illustrates a mechanism by which mTORC1 signal complex regulates BTB function through rpS6 downstream by modulating actin organization via the Arp2/3 complex, which may be applicable to other tissue barriers.
Collapse
Affiliation(s)
- Ka-Wai Mok
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (K.-W.M., H.C., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065; and School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
40
|
Borges S, Perez EA, Thompson EA, Radisky DC, Geiger XJ, Storz P. Effective Targeting of Estrogen Receptor-Negative Breast Cancers with the Protein Kinase D Inhibitor CRT0066101. Mol Cancer Ther 2015; 14:1306-16. [PMID: 25852060 DOI: 10.1158/1535-7163.mct-14-0945] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/02/2015] [Indexed: 12/11/2022]
Abstract
Invasive ductal carcinomas (IDC) of the breast are associated with altered expression of hormone receptors (HR), amplification or overexpression of HER2, or a triple-negative phenotype. The most aggressive cases of IDC are characterized by a high proliferation rate, a great propensity to metastasize, and their ability to resist to standard chemotherapy, hormone therapy, or HER2-targeted therapy. Using progression tissue microarrays, we here demonstrate that the serine/threonine kinase protein kinase D3 (PKD3) is highly upregulated in estrogen receptor (ER)-negative (ER(-)) tumors. We identify direct binding of the ER to the PRKD3 gene promoter as a mechanism of inhibition of PKD3 expression. Loss of ER results in upregulation of PKD3, leading to all hallmarks of aggressive IDC, including increased cell proliferation, migration, and invasion. This identifies ER(-) breast cancers as ideal for treatment with the PKD inhibitor CRT0066101. We show that similar to a knockdown of PKD3, treatment with this inhibitor targets all tumorigenic processes in vitro and decreases growth of primary tumors and metastasis in vivo. Our data strongly support the development of PKD inhibitors for clinical use for ER(-) breast cancers, including the triple-negative phenotype.
Collapse
Affiliation(s)
- Sahra Borges
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Edith A Perez
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida. Department of Hematology/Oncology, Mayo Clinic, Jacksonville, Florida
| | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | | | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
41
|
Abstract
Cancer is driven by genetic and epigenetic alterations that allow cells to overproliferate and escape mechanisms that normally control their survival and migration. Many of these alterations map to signaling pathways that control cell growth and division, cell death, cell fate, and cell motility, and can be placed in the context of distortions of wider signaling networks that fuel cancer progression, such as changes in the tumor microenvironment, angiogenesis, and inflammation. Mutations that convert cellular proto-oncogenes to oncogenes can cause hyperactivation of these signaling pathways, whereas inactivation of tumor suppressors eliminates critical negative regulators of signaling. An examination of the PI3K-Akt and Ras-ERK pathways illustrates how such alterations dysregulate signaling in cancer and produce many of the characteristic features of tumor cells.
Collapse
Affiliation(s)
- Richard Sever
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | - Joan S Brugge
- Harvard Medical School, Department of Cell Biology, Boston, Massachusetts 02115
| |
Collapse
|
42
|
Nowak DG, Cho H, Herzka T, Watrud K, DeMarco DV, Wang VMY, Senturk S, Fellmann C, Ding D, Beinortas T, Kleinman D, Chen M, Sordella R, Wilkinson JE, Castillo-Martin M, Cordon-Cardo C, Robinson BD, Trotman LC. MYC Drives Pten/Trp53-Deficient Proliferation and Metastasis due to IL6 Secretion and AKT Suppression via PHLPP2. Cancer Discov 2015; 5:636-51. [PMID: 25829425 DOI: 10.1158/2159-8290.cd-14-1113] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/26/2015] [Indexed: 01/24/2023]
Abstract
UNLABELLED We have recently recapitulated metastasis of human PTEN/TP53-mutant prostate cancer in the mouse using the RapidCaP system. Surprisingly, we found that this metastasis is driven by MYC, and not AKT, activation. Here, we show that cell-cell communication by IL6 drives the AKT-MYC switch through activation of the AKT-suppressing phosphatase PHLPP2, when PTEN and p53 are lost together, but not separately. IL6 then communicates a downstream program of STAT3-mediated MYC activation, which drives cell proliferation. Similarly, in tissues, peak proliferation in Pten/Trp53-mutant primary and metastatic prostate cancer does not correlate with activated AKT, but with STAT3/MYC activation instead. Mechanistically, MYC strongly activates the AKT phosphatase PHLPP2 in primary cells and prostate cancer metastasis. We show genetically that Phlpp2 is essential for dictating the proliferation of MYC-mediated AKT suppression. Collectively, our data reveal competition between two proto-oncogenes, MYC and AKT, which ensnarls the Phlpp2 gene to facilitate MYC-driven prostate cancer metastasis after loss of Pten and Trp53. SIGNIFICANCE Our data identify IL6 detection as a potential causal biomarker for MYC-driven metastasis after loss of PTEN and p53. Second, our finding that MYC then must supersede AKT to drive cell proliferation points to MYC inhibition as a critical part of PI3K pathway therapy in lethal prostate cancer.
Collapse
Affiliation(s)
- Dawid G Nowak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Hyejin Cho
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Tali Herzka
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Kaitlin Watrud
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | | | | - Serif Senturk
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | | - David Ding
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | | - David Kleinman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Muhan Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | | - John E Wilkinson
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | - Carlos Cordon-Cardo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brian D Robinson
- Department of Pathology, NewYork-Presbyterian Hospital, Weill Cornell Medical College, New York, New York
| | - Lloyd C Trotman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York.
| |
Collapse
|
43
|
Special AT-rich sequence-binding protein 2 suppresses invadopodia formation in HCT116 cells via palladin inhibition. Exp Cell Res 2015; 332:78-88. [DOI: 10.1016/j.yexcr.2014.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/28/2014] [Accepted: 12/05/2014] [Indexed: 11/18/2022]
|
44
|
Manna P, Jain SK. Phosphatidylinositol-3,4,5-triphosphate and cellular signaling: implications for obesity and diabetes. Cell Physiol Biochem 2015; 35:1253-75. [PMID: 25721445 DOI: 10.1159/000373949] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2015] [Indexed: 12/26/2022] Open
Abstract
Phosphatidylinositol-3,4,5-triphosphate (PtdIns(3,4,5)P₃) is one of the most important phosphoinositides and is capable of activating a wide range of proteins through its interaction with their specific binding domains. Localization and activation of these effector proteins regulate a number of cellular functions, including cell survival, proliferation, cytoskeletal rearrangement, intracellular vesicle trafficking, and cell metabolism. Phosphoinositides have been investigated as an important agonist-dependent second messenger in the regulation of diverse physiological events depending upon the phosphorylation status of their inositol group. Dysregulation in formation as well as metabolism of phosphoinositides is associated with various pathophysiological disorders such as inflammation, allergy, cardiovascular diseases, cancer, and metabolic diseases. Recent studies have demonstrated that the impaired metabolism of PtdIns(3,4,5)P₃ is a prime mediator of insulin resistance associated with various metabolic diseases including obesity and diabetes. This review examines the current status of the role of PtdIns(3,4,5)P₃ signaling in the regulation of various cellular functions and the implications of dysregulated PtdIns(3,4,5)P₃ signaling in obesity, diabetes, and their associated complications.
Collapse
Affiliation(s)
- Prasenjit Manna
- Department of Pediatrics, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | |
Collapse
|
45
|
Halacli SO, Dogan AL. FOXP1 regulation via the PI3K/Akt/p70S6K signaling pathway in breast cancer cells. Oncol Lett 2015; 9:1482-1488. [PMID: 25663935 PMCID: PMC4315073 DOI: 10.3892/ol.2015.2885] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/07/2015] [Indexed: 02/01/2023] Open
Abstract
Loss of Forkhead box P1 (FOXP1) protein expression confers a poor prognosis in sporadic and familial breast cancer patients, and the FOXP1 gene maps to a tumor suppressor locus at chromosome 3p14. Although correlation studies have indicated that FOXP1 has a role in tumor suppression, determination of the regulatory mechanism of FOXP1 is required to establish its function in breast cancer. It has previously been identified that FOXP1 is regulated by estrogen in breast cancer and that treatment with bisphenol A is effective for regulating the transformation of the normal human breast epithelial cell line, MCF-10F. In addition, FOXO-regulated activation of FOXP1 inhibits the apoptosis of MCF-10F cells following tamoxifen and Akt inhibitor VIII administration. The present study indicates that FOXP1 regulation occurs via a PI3K/Akt/p70S6 kinase (p70S6K) signaling pathway. Following treatment with wortmannin, an inhibitor of phosphatidylinositol 3-kinase (PI3K)/Akt, MCF7 and MDA-MB-231 breast cancer cells demonstrated decreased FOXP1 protein expression levels; this result was also observed in the small interfering (si)RNA silencing of Akt. By contrast, overexpression of Akt resulted in increased FOXP1 protein expression levels in the MDA-MB-231 cells compared with the control cell lysates. Furthermore, treatment with rapamycin, a specific inhibitor of the mammalian target of rapamycin/p70S6K cascade, resulted in decreased FOXP1 expression in the MCF7 cells, but not in the MDA-MB-231 cells, which were resistant to rapamycin-induced inhibition. In addition, silencing of p70S6K using siRNA produced a marked decrease in FOXP1 expression. These data indicate that FOXP1 protein expression is regulated by a PI3K/Akt/p70S6K signaling cascade in breast cancer.
Collapse
Affiliation(s)
- Sevil Oskay Halacli
- Pediatric Immunology Unit, Institute of Children's Health, Hacettepe University, Ankara 06100, Turkey
| | - Ayse Lale Dogan
- Department of Basic Oncology, Institute of Oncology, Hacettepe University, Ankara 06100, Turkey
| |
Collapse
|
46
|
Gungor-Ordueri NE, Celik-Ozenci C, Cheng CY. Fascin 1 is an actin filament-bundling protein that regulates ectoplasmic specialization dynamics in the rat testis. Am J Physiol Endocrinol Metab 2014; 307:E738-53. [PMID: 25159326 PMCID: PMC4216949 DOI: 10.1152/ajpendo.00113.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the testis, spermatids are polarized cells, with their heads pointing toward the basement membrane during maturation. This polarity is crucial to pack the maximal number of spermatids in the seminiferous epithelium so that millions of sperms can be produced daily. A loss of spermatid polarity is detected after rodents are exposed to toxicants (e.g., cadmium) or nonhormonal male contraceptives (e.g., adjudin), which is associated with a disruption on the expression and/or localization of polarity proteins. In the rat testis, fascin 1, an actin-bundling protein found in mammalian cells, was expressed by Sertoli and germ cells. Fascin 1 was a component of the ectoplasmic specialization (ES), a testis-specific anchoring junction known to confer spermatid adhesion and polarity. Its expression in the seminiferous epithelium was stage specific. Fascin 1 was localized to the basal ES at the Sertoli cell-cell interface of the blood-testis barrier in all stages of the epithelial cycle, except it diminished considerably at late stage VIII. Fascin 1 was highly expressed at the apical ES at stage VII-early stage VIII and restricted to the step 19 spermatids. Its knockdown by RNAi that silenced fascin 1 by ~70% in Sertoli cells cultured in vitro was found to perturb the tight junction-permeability barrier via a disruption of F-actin organization. Knockdown of fascin 1 in vivo by ~60-70% induced defects in spermatid polarity, which was mediated by a mislocalization and/or downregulation of actin-bundling proteins Eps8 and palladin, thereby impeding F-actin organization and disrupting spermatid polarity. In summary, these findings provide insightful information on spermatid polarity regulation.
Collapse
Affiliation(s)
- N Ece Gungor-Ordueri
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York; and
| | - Ciler Celik-Ozenci
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York; and
| |
Collapse
|
47
|
Qian X, Mruk DD, Cheng YH, Cheng CY. Actin cross-linking protein palladin and spermatogenesis. SPERMATOGENESIS 2014; 3:e23473. [PMID: 23687615 PMCID: PMC3644046 DOI: 10.4161/spmg.23473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the seminiferous epithelium of the mammalian testis, the most distinctive ultrastructure is the extensive bundles of actin filaments that lie near the Sertoli-spermatid interface and the Sertoli-Sertoli cell interface known as the apical ectoplasmic specialization (apical ES) and the basal ES, respectively. These actin filament bundles not only confer strong adhesion at these sites, they are uniquely found in the testis. Recent studies have shown that ES also confers spermatid and Sertoli cell polarity in the seminiferous epithelium during the epithelial cycle. While these junctions were first described in the 1970s, there are few functional studies in the literature to examine the regulation of these actin filament bundles. It is conceivable that these actin filament bundles at the ES undergo extensive re-organization to accommodate changes in location of developing spermatids during spermiogenesis as spermatids are transported across the seminiferous epithelium. Additionally, these actin filaments are rapidly reorganized during BTB restructuring to accommodate the transit of preleptotene spermatocytes across the barrier at stage VIII of the epithelial cycle. Thus, actin binding and regulatory proteins are likely involved in these events to confer changes in F-actin organization at these sites. Interestingly, there are no reports in the field to study these regulatory proteins until recently. Herein, we summarize some of the latest findings in the field regarding a novel actin cross-linker and actin-bundling protein called palladin. We also discuss in this opinion article the likely role of palladin in regulating actin filament bundles at the ES during spermatogenesis, highlighting the significant of palladin and how this protein is plausibly working in concert with other actin-binding/regulatory proteins and components of polarity proteins to regulate the cyclic events of actin organization and re-organization during the epithelial cycle of spermatogenesis. We also propose a hypothetic model by which palladin regulates ES restructuring during the epithelial cycle of spermatogenesis.
Collapse
Affiliation(s)
- Xiaojing Qian
- The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; Population Council; New York, NY USA ; School of Basic Medicine; Peking Union Medical College; Beijing, China
| | | | | | | |
Collapse
|
48
|
Hu B, Emdad L, Bacolod MD, Kegelman TP, Shen XN, Alzubi MA, Das SK, Sarkar D, Fisher PB. Astrocyte elevated gene-1 interacts with Akt isoform 2 to control glioma growth, survival, and pathogenesis. Cancer Res 2014; 74:7321-32. [PMID: 25304263 DOI: 10.1158/0008-5472.can-13-2978] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The oncogene astrocyte elevated gene-1 (AEG-1; MTDH) is highly expressed in glioblastoma multiforme (GBM) and many other types of cancer, where it activates multiple signaling pathways that drive proliferation, invasion, angiogenesis, chemoresistance, radioresistance, and metastasis. AEG-1 activates the Akt signaling pathway and Akt and c-Myc are positive regulators of AEG-1 transcription, generating a positive feedback loop between AEG-1 and Akt in regulating tumorigenesis. Here, we describe in GBM cells a direct interaction between an internal domain of AEG-1 and the PH domain of Akt2, a major driver in GBM. Expression and interaction of AEG-1 and Akt2 are elevated in GBM and contribute to tumor cell survival, proliferation, and invasion. Clinically, in silico gene expression and immunohistochemical analyses of patient specimens showed that AEG-1 and Akt2 expression correlated with GBM progression and reduced patient survival. AEG-1-Akt2 interaction prolonged stabilization of Akt2 phosphorylation at S474, regulating downstream signaling cascades that enable cell proliferation and survival. Disrupting AEG-1-Akt2 interaction by competitive binding of the Akt2-PH domain led to reduced cell viability and invasion. When combined with AEG-1 silencing, conditional expression of Akt2-PH markedly increased survival in an orthotopic mouse model of human GBM. Our study uncovers a novel molecular mechanism by which AEG-1 augments glioma progression and offers a rationale to block AEG-1-Akt2 signaling function as a novel GBM treatment.
Collapse
Affiliation(s)
- Bin Hu
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Manny D Bacolod
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Timothy P Kegelman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Xue-Ning Shen
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Mohammad A Alzubi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia. VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia.
| |
Collapse
|
49
|
Babaev VR, Hebron KE, Wiese CB, Toth CL, Ding L, Zhang Y, May JM, Fazio S, Vickers KC, Linton MF. Macrophage deficiency of Akt2 reduces atherosclerosis in Ldlr null mice. J Lipid Res 2014; 55:2296-308. [PMID: 25240046 PMCID: PMC4617132 DOI: 10.1194/jlr.m050633] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Macrophages play crucial roles in the formation of atherosclerotic lesions. Akt, a serine/threonine protein kinase B, is vital for cell proliferation, migration, and survival. Macrophages express three Akt isoforms, Akt1, Akt2, and Akt3, but the roles of Akt1 and Akt2 in atherosclerosis in vivo remain unclear. To dissect the impact of macrophage Akt1 and Akt2 on early atherosclerosis, we generated mice with hematopoietic deficiency of Akt1 or Akt2. After 8 weeks on Western diet, Ldlr−/− mice reconstituted with Akt1−/− fetal liver cells (Akt1−/−→Ldlr−/−) had similar atherosclerotic lesion areas compared with control mice transplanted with WT cells (WT→Ldlr−/−). In contrast, Akt2−/−→Ldlr−/− mice had dramatically reduced atherosclerotic lesions compared with WT→Ldlr−/− mice of both genders. Similarly, in the setting of advanced atherosclerotic lesions, Akt2−/−→Ldlr−/− mice had smaller aortic lesions compared with WT→Ldlr−/− and Akt1−/−→Ldlr−/− mice. Importantly, Akt2−/−→Ldlr−/− mice had reduced numbers of proinflammatory blood monocytes expressing Ly-6Chi and chemokine C-C motif receptor 2. Peritoneal macrophages isolated from Akt2−/− mice were skewed toward an M2 phenotype and showed decreased expression of proinflammatory genes and reduced cell migration. Our data demonstrate that loss of Akt2 suppresses the ability of macrophages to undergo M1 polarization reducing both early and advanced atherosclerosis.
Collapse
Affiliation(s)
- Vladimir R Babaev
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Katie E Hebron
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Carrie B Wiese
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Cynthia L Toth
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Lei Ding
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Youmin Zhang
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - James M May
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Sergio Fazio
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Pathology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Kasey C Vickers
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN 37232
| | - MacRae F Linton
- Atherosclerosis Research Unit, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
50
|
Tang Y, Jiang Z, Luo Y, Zhao X, Wang L, Norris C, Tian XC. Differential effects of Akt isoforms on somatic cell reprogramming. J Cell Sci 2014; 127:3998-4008. [PMID: 25037569 DOI: 10.1242/jcs.150029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Akt plays an important role in cell growth, proliferation and survival. The specific roles of the three Akt isoforms in somatic cell reprogramming have not been investigated. Here we report that, during iPSC generation, enhanced Akt1 activity promotes complete reprogramming mainly through increased activation of Stat3 in concert with leukemia inhibitory factor (LIF) and, to a lesser extent, through promotion of colony formation. Akt1 augments Stat3 activity through activation of mTOR and upregulation of LIF receptor expression. Similarly, enhanced Akt2 or Akt3 activation also promotes reprogramming and coordinates with LIF to activate Stat3. Blocking Akt1 or Akt3 but not Akt2 expression prohibits cell proliferation and reprogramming. Furthermore, the halt in cell proliferation and reprogramming caused by mTOR and Akt inhibitors can be reversed by inhibition of GSK3. Finally, we found that expressing the GSK3β target Esrrb overrides inhibition of Akt and restores reprogramming. Our data demonstrated that during reprogramming, Akt promotes establishment of pluripotency through co-stimulation of Stat3 activity with LIF. Akt1 and Akt3 are essential for the proliferation of reprogrammed cells, and Esrrb supports cell proliferation and complete reprogramming during Akt signaling.
Collapse
Affiliation(s)
- Yong Tang
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Zongliang Jiang
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Yan Luo
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Xueming Zhao
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Ling Wang
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Carol Norris
- Department of Molecular and Cellular Biology/Biotechnology and Bioservices Center, University of Connecticut, Storrs, CT 06269, USA
| | - Xiuchun Cindy Tian
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|