301
|
Liu D, Wang X, Qin W, Chen J, Wang Y, Zhuang M, Sun B. Suppressive effect of exogenous carbon monoxide on endotoxin-stimulated platelet over-activation via the glycoprotein-mediated PI3K-Akt-GSK3β pathway. Sci Rep 2016; 6:23653. [PMID: 27020460 PMCID: PMC4810323 DOI: 10.1038/srep23653] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 03/10/2016] [Indexed: 12/15/2022] Open
Abstract
Platelet activation is an important event involved in the pathophysiological processes of the coagulation system. Clinical evidence has shown that platelets undergo distinctive pathological processes during sepsis. Unfortunately, how platelets physiologically respond to inflammation or sepsis is not well understood. In this study, we used a lipopolysaccharide (LPS)-stimulated platelet model to systemically investigate alterations in membrane glycoprotein expression, molecular signaling, morphology and critical functions of platelets. We found that platelet adhesion, aggregation, secretion, and spreading on immobilized fibrinogen and the expression of platelet membrane glycoproteins were significantly increased by LPS stimulation, and these changes were accompanied by a significant decrease in cGMP levels and an abnormal distribution of platelet α-granules. Exogenous CO reversed these alterations. Profound morphological changes in LPS-stimulated platelets were observed using atomic force microscopy and phase microscopy. Furthermore, the elevated activities of PI3Ks, AKt and GSK-3β were effectively suppressed by exogenous CO, leading to the improvement of platelet function. Together, these results provide evidence that platelet over-activation persists under LPS-stimulation and that exogenous CO plays an important role in suppressing platelet activation via the glycoprotein-mediated PI3K-Akt-GSK3β pathway.
Collapse
Affiliation(s)
- Dadong Liu
- Department of Critical Care Medicine, Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Xu Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Weiting Qin
- Department of Burns and Plastic Surgery, Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Jingjia Chen
- Department of Burns and Plastic Surgery, Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yawei Wang
- School of Science, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Mingfeng Zhuang
- Department of Burns and Plastic Surgery, Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Bingwei Sun
- Department of Burns and Plastic Surgery, Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu Province, China
| |
Collapse
|
302
|
Wu T, Wang C, Ding L, Shen Y, Cui H, Wang M, Wang H. Arginine Relieves the Inflammatory Response and Enhances the Casein Expression in Bovine Mammary Epithelial Cells Induced by Lipopolysaccharide. Mediators Inflamm 2016; 2016:9618795. [PMID: 27110069 PMCID: PMC4821974 DOI: 10.1155/2016/9618795] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/24/2016] [Indexed: 12/31/2022] Open
Abstract
As one of functional active amino acids, L-arginine holds a key position in immunity. However, the mechanism that arginine modulates cow mammary inflammatory response in ruminant is unclear. Therefore, this study was conducted to investigate the effects of L-arginine on inflammatory response and casein expression after challenging the bovine mammary epithelial cells (BMECs) with lipopolysaccharide (LPS). The cells were divided into four groups, stimulated with or without LPS (10 μg/mL) and treated with or without arginine (100 μg/mL) for 12 h. The concentration of proinflammatory cytokines, inducible nitric oxide synthase (iNOS), mammalian target of rapamycin (mTOR), and Toll-like receptor 4 (TLR4) signaling pathways as well as the casein was determined. The results showed that arginine reduced the LPS-induced production like IL-1β, IL-6, TNF-α, and iNOS. Though the expression of NF-κB was attenuated and the mTOR signaling pathway was upregulated, arginine had no effect on TLR4 expression. In addition, our results show that the content of β-casein and the total casein were enhanced after arginine was supplemented in LPS-induced BMECs. In conclusion, arginine could relieve the inflammatory reaction induced by LPS and enhance the concentration of β-casein and the total casein in bovine mammary epithelial cells.
Collapse
Affiliation(s)
- Tianyou Wu
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Chao Wang
- Cell Signaling Group, School of Pathology and Laboratory Medicine, The University of Western Australia, M Block QEII Medical Center, Monash Avenue, Nedlands, WA 6009, Australia
| | - Luoyang Ding
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yizhao Shen
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Huihui Cui
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
303
|
Sudano MJ, Rascado TDS, Tata A, Belaz KRA, Santos VG, Valente RS, Mesquita FS, Ferreira CR, Araújo JP, Eberlin MN, Landim-Alvarenga FDC. Lipidome signatures in early bovine embryo development. Theriogenology 2016; 86:472-484.e1. [PMID: 27107972 DOI: 10.1016/j.theriogenology.2016.03.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Mammalian preimplantation embryonic development is a complex, conserved, and well-orchestrated process involving dynamic molecular and structural changes. Understanding membrane lipid profile fluctuation during this crucial period is fundamental to address mechanisms governing embryogenesis. Therefore, the aim of the present work was to perform a comprehensive assessment of stage-specific lipid profiles during early bovine embryonic development and associate with the mRNA abundance of lipid metabolism-related genes (ACSL3, ELOVL5, and ELOVL6) and with the amount of cytoplasmic lipid droplets. Immature oocytes were recovered from slaughterhouse-derived ovaries, two-cell embryos, and eight- to 16-cell embryos, morula, and blastocysts that were in vitro produced under different environmental conditions. Lipid droplets content and mRNA transcript levels for ACSL3, ELOVL5, and ELOVL6, monitored by lipid staining and quantitative polymerase chain reaction, respectively, increased at morula followed by a decrease at blastocyst stage. Relative mRNA abundance changes of ACSL3 were closely related to cytoplasmic lipid droplet accumulation. Characteristic dynamic changes of phospholipid profiles were observed during early embryo development and related to unsaturation level, acyl chain length, and class composition. ELOVL5 and ELOVL6 mRNA levels were suggestive of overexpression of membrane phospholipids containing elongated fatty acids with 16, 18, and 20 carbons. In addition, putative biomarkers of key events of embryogenesis, embryo lipid accumulation, and elongation were identified. This study provides a comprehensive description of stage-specific lipidome signatures and proposes a mechanism to explain its potential relationship with the fluctuation of both cytoplasmic lipid droplets content and mRNA levels of lipid metabolism-related genes during early bovine embryo development.
Collapse
Affiliation(s)
- Mateus J Sudano
- School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, RS, Brazil.
| | - Tatiana D S Rascado
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, SP, Brazil
| | - Alessandra Tata
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas, Campinas, SP, Brazil
| | - Katia R A Belaz
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas, Campinas, SP, Brazil
| | - Vanessa G Santos
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas, Campinas, SP, Brazil
| | - Roniele S Valente
- School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Fernando S Mesquita
- School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Christina R Ferreira
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas, Campinas, SP, Brazil
| | - João P Araújo
- Department of Microbiology and Immunology, Biosciences Institute, São Paulo State University, Botucatu, SP, Brazil
| | - Marcos N Eberlin
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas, Campinas, SP, Brazil
| | - Fernanda D C Landim-Alvarenga
- Department of Animal Reproduction and Veterinary Radiology, School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu, SP, Brazil
| |
Collapse
|
304
|
Xie G, Yu X, Liang H, Chen J, Tang X, Wu S, Liao C. Pristimerin overcomes adriamycin resistance in breast cancer cells through suppressing Akt signaling. Oncol Lett 2016; 11:3111-3116. [PMID: 27123073 DOI: 10.3892/ol.2016.4335] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 02/05/2016] [Indexed: 11/06/2022] Open
Abstract
Breast cancer remains a major public health problem worldwide. Chemotherapy serves an important role in the treatment of breast cancer. However, resistance to chemotherapeutic agents, in particular, multi-drug resistance (MDR), is a major cause of treatment failure in cancer. Agents that can either enhance the effects of chemotherapeutics or overcome chemoresistance are urgently needed for the treatment of breast cancer. Pristimerin, a quinonemethide triterpenoid compound isolated from Celastraceae and Hippocrateaceae, has been shown to possess antitumor, anti-inflammatory, antioxidant and insecticidal properties. The aim of the present study was to investigate whether pristimerin can override chemoresistance in MCF-7/adriamycin (ADR)-resistant human breast cancer cells. The results demonstrated that pristimerin indeed displayed potent cytocidal effect on multidrug-resistant MCF-7/ADR breast cancer cells, and that these effects occurred through the suppression of Akt signaling, which in turn led to the downregulation of antiapoptotic effectors and increased apoptosis. These findings indicate that use of pristimerin may represent a potentially promising approach for the treatment of ADR-resistant breast cancer.
Collapse
Affiliation(s)
- Gui'e Xie
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Xinpei Yu
- Cancer Center, Southern Medical University, Guangzhou, Guangdong 510315, P.R. China; Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, Guangdong 510315, P.R. China
| | - Huichao Liang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Jingsong Chen
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Xuewei Tang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Shaoqing Wu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Can Liao
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| |
Collapse
|
305
|
Bommarito D, Martin A, Forcade E, Nastke MD, Ritz J, Bellucci R. Enhancement of tumor cell susceptibility to natural killer cell activity through inhibition of the PI3K signaling pathway. Cancer Immunol Immunother 2016; 65:355-66. [PMID: 26883876 PMCID: PMC4862590 DOI: 10.1007/s00262-016-1804-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/29/2016] [Indexed: 01/09/2023]
Abstract
Natural killer (NK) cells are the primary effectors of the innate immune response against virus-infected cells or cells that have undergone malignant transformation. NK cells recognize their targets through a complex array of activating and inhibitory receptors, which regulate the intensity of the effector response against individual target cells. However, many studies have shown that tumor cells can escape immune cell recognition through a variety of mechanisms, developing resistance to NK cell killing. Using a lentiviral shRNA library, we previously demonstrated that several common signaling pathways modulate susceptibility of tumor cells to NK cell activity. In this study, we focused on one of the genes (PI3KCB), identified in this genetic screen. The PI3KCB gene encodes an isoform of the catalytic subunit of PI3K called P110β. The PI3K pathway has been linked to diverse cellular functions, but has never been associated with susceptibility to NK cell activity. Gene silencing of PI3KCB resulted in increased susceptibility of several tumor cell lines to NK cell lytic activity and induced increased IFN-γ secretion by NK cells. Treatment of primary tumor cells with two different PI3K inhibitors also increased target cell susceptibility to NK cell activity. These effects are due, at least in part, to modulation of several activating and inhibitory ligands and appear to be correlated with PI3K signaling pathway inhibition. These findings identify a new and important role of PI3KCB in modulating tumor cell susceptibility to NK cells and open the way to future combined target immunotherapies.
Collapse
Affiliation(s)
- Davide Bommarito
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, M526, Boston, MA, 02215, USA
| | - Allison Martin
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, M526, Boston, MA, 02215, USA
| | - Edouard Forcade
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, M526, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Maria-Dorothea Nastke
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, M526, Boston, MA, 02215, USA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, M526, Boston, MA, 02215, USA.
- Cancer Vaccine Center, Dana-Farber Cancer Institute, 450 Brookline Ave, M530, Boston, MA, 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Roberto Bellucci
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, M526, Boston, MA, 02215, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
306
|
Kim MJ, Koo JE, Han GY, Kim B, Lee YS, Ahn C, Kim CW. Dual-Blocking of PI3K and mTOR Improves Chemotherapeutic Effects on SW620 Human Colorectal Cancer Stem Cells by Inducing Differentiation. J Korean Med Sci 2016; 31:360-70. [PMID: 26955235 PMCID: PMC4779859 DOI: 10.3346/jkms.2016.31.3.360] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/19/2015] [Indexed: 12/28/2022] Open
Abstract
Cancer stem cells (CSCs) have tumor initiation, self-renewal, metastasis and chemo-resistance properties in various tumors including colorectal cancer. Targeting of CSCs may be essential to prevent relapse of tumors after chemotherapy. Phosphatidylinositol-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) signals are central regulators of cell growth, proliferation, differentiation, and apoptosis. These pathways are related to colorectal tumorigenesis. This study focused on PI3K and mTOR pathways by inhibition which initiate differentiation of SW620 derived CSCs and investigated its effect on tumor progression. By using rapamycin, LY294002, and NVP-BEZ235, respectively, PI3K and mTOR signals were blocked independently or dually in colorectal CSCs. Colorectal CSCs gained their differentiation property and lost their stemness properties most significantly in dual-blocked CSCs. After treated with anti-cancer drug (paclitaxel) on the differentiated CSCs cell viability, self-renewal ability and differentiation status were analyzed. As a result dual-blocking group has most enhanced sensitivity for anti-cancer drug. Xenograft tumorigenesis assay by using immunodeficiency mice also shows that dual-inhibited group more effectively increased drug sensitivity and suppressed tumor growth compared to single-inhibited groups. Therefore it could have potent anti-cancer effects that dual-blocking of PI3K and mTOR induces differentiation and improves chemotherapeutic effects on SW620 human colorectal CSCs.
Collapse
Affiliation(s)
- Min-Jung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
- Ministry of Food and Drug Safety, Cheongju, Korea
| | - Jeong-Eun Koo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Gi-Yeon Han
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Buyun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Yoo-Sun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Chiyoung Ahn
- Ministry of Food and Drug Safety, Cheongju, Korea
| | - Chan-Wha Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| |
Collapse
|
307
|
Kalsi N, Gopalakrishnan C, Rajendran V, Purohit R. Biophysical aspect of phosphatidylinositol 3-kinase and role of oncogenic mutants (E542K & E545K). J Biomol Struct Dyn 2016; 34:2711-2721. [DOI: 10.1080/07391102.2015.1127774] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
308
|
Josephs DH, Sarker D. Pharmacodynamic Biomarker Development for PI3K Pathway Therapeutics. TRANSLATIONAL ONCOGENOMICS 2016; 7:33-49. [PMID: 26917948 PMCID: PMC4762492 DOI: 10.4137/tog.s30529] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 11/08/2015] [Accepted: 11/10/2015] [Indexed: 12/11/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway is integral to many essential cell processes, including cell growth, differentiation, proliferation, motility, and metabolism. Somatic mutations and genetic amplifications that result in activation of the pathway are frequently detected in cancer. This has led to the development of rationally designed therapeutics targeting key members of the pathway. Critical to the successful development of these drugs are pharmacodynamic biomarkers that aim to define the degree of target and pathway inhibition. In this review, we discuss the pharmacodynamic biomarkers that have been utilized in early-phase clinical trials of PI3K pathway inhibitors. We focus on the challenges related to development and interpretation of these assays, their optimal integration with pharmacokinetic and predictive biomarkers, and future strategies to ensure successful development of PI3K pathway inhibitors within a personalized medicine paradigm for cancer.
Collapse
Affiliation(s)
- Debra H Josephs
- Department of Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| | - Debashis Sarker
- Department of Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
309
|
Abstract
Buparlisib (formerly BKM 120), an oral 2,6-dimorpholino pyrimidine derivative is a potent pan-PI3K inhibitor causing inhibition of PI3K downstream signaling including downregulation of p-Akt and p-S6R and apoptosis of cancer cells. Buparlisib is rapidly absorbed, has more than 90% bioavailability, good blood-brain barrier penetration and half-life of 40 h. Phase I trials have shown good disease control rate with tolerable toxicity profile at the recommended doses of 100 mg. The most common adverse events noted with buparlisib are rash, hyperglycemia, derangement of liver functions and psychiatric events. Several clinical trials with buparlisib alone or in combination with chemotherapy and targeted therapies are underway. Buparlisib has not yet been approved for regular use. Further randomized trials are required before buparlisib is approved for treatment of breast cancer.
Collapse
Affiliation(s)
- Bhawna Sirohi
- Department of Medical Oncology, Mazumdar Shaw Cancer Centre, Narayana Health, Bangalore, India
| | | | | |
Collapse
|
310
|
Li Z, Yao J, Xie Y, Geng X, Liu Z. Phosphoinositide 3-kinase family in channel catfish and their regulated expression after bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2016; 49:364-373. [PMID: 26772478 DOI: 10.1016/j.fsi.2016.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/30/2015] [Accepted: 01/03/2016] [Indexed: 06/05/2023]
Abstract
The phosphoinositide-3-kinase (PI3Ks) family of lipid kinases is widely conserved from yeast to mammals. In this work, we identified a total of 14 members of the PI3Ks from the channel catfish genome and transcriptome and conducted phylogenetic and syntenic analyses of these genes. The expression profiles after infection with Edwardsiella ictaluri and Flavobacterium columnare were examined to determine the involvement of PI3Ks in immune responses after bacterial infection in catfish. The results indicated that PI3Ks genes including all of the catalytic subunit and several regulatory subunits genes were widely regulated after bacterial infection. The expression patterns were quite different when challenged with different bacteria. The PI3Ks were up-regulated rapidly at the early stage after ESC infection, but their induced expression was much slower, at the middle stage after columnaris infection. RNA-Seq datasets indicated that PI3K genes may be expressed at different levels in different catfish differing in their resistance levels against columnaris. Future studies are required to confirm and validate these observations. Taken together, this study indicated that PI3K genes may be involved as a part of the defense responses of catfish after infections, and they could be one of the determinants for disease resistance.
Collapse
Affiliation(s)
- Zhaoxia Li
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA; Marine Science and Engineering College, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Jun Yao
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Yangjie Xie
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Xin Geng
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Zhanjiang Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
311
|
Caspase-3 is Involved in Aluminum-Induced Impairment of Long-Term Potentiation in Rats Through the Akt/GSK-3β Pathway. Neurotox Res 2016; 29:484-94. [DOI: 10.1007/s12640-016-9597-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/15/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
|
312
|
Ho CY, Choy CH, Botelho RJ. Radiolabeling and Quantification of Cellular Levels of Phosphoinositides by High Performance Liquid Chromatography-coupled Flow Scintillation. J Vis Exp 2016. [PMID: 26780479 DOI: 10.3791/53529] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Phosphoinositides (PtdInsPs) are essential signaling lipids responsible for recruiting specific effectors and conferring organelles with molecular identity and function. Each of the seven PtdInsPs varies in their distribution and abundance, which are tightly regulated by specific kinases and phosphatases. The abundance of PtdInsPs can change abruptly in response to various signaling events or disturbance of the regulatory machinery. To understand how these events lead to changes in the amount of PtdInsPs and their resulting impact, it is important to quantify PtdInsP levels before and after a signaling event or between control and abnormal conditions. However, due to their low abundance and similarity, quantifying the relative amounts of each PtdInsP can be challenging. This article describes a method for quantifying PtdInsP levels by metabolically labeling cells with (3)H-myo-inositol, which is incorporated into PtdInsPs. Phospholipids are then precipitated and deacylated. The resulting soluble (3)H-glycero-inositides are further extracted, separated by high-performance liquid chromatography (HPLC), and detected by flow scintillation. The labeling and processing of yeast samples is described in detail, as well as the instrumental setup for the HPLC and flow scintillator. Despite losing structural information regarding acyl chain content, this method is sensitive and can be optimized to concurrently quantify all seven PtdInsPs in cells.
Collapse
Affiliation(s)
- Cheuk Y Ho
- Department of Chemistry and Biology, Program in Molecular Science, Ryerson University
| | - Christopher H Choy
- Department of Chemistry and Biology, Program in Molecular Science, Ryerson University
| | - Roberto J Botelho
- Department of Chemistry and Biology, Program in Molecular Science, Ryerson University;
| |
Collapse
|
313
|
The inhibition of PI3K and NFκB promoted curcumin-induced cell cycle arrest at G2/M via altering polyamine metabolism in Bcl-2 overexpressing MCF-7 breast cancer cells. Biomed Pharmacother 2015; 77:150-60. [PMID: 26796279 DOI: 10.1016/j.biopha.2015.12.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 11/06/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022] Open
Abstract
Bcl-2 protein has been contributed with number of genes which are involved in oncogenesis. Among the many targets of Bcl-2, NFκB have potential role in induction of cell cycle arrest. Curcumin has potential therapeutic effects against breast cancer through multiple signaling pathways. In this study, we investigated the role of curcumin in induction of cell cycle arrest via regulating of NFκB and polyamine biosynthesis in wt and Bcl-2+ MCF-7 cells. To examine the effect of curcumin on cell cycle regulatory proteins, PI3K/Akt, NFκB pathways and polyamine catabolism, we performed immunoblotting assay. In addition, cell cycle analysis was performed by flow cytometry. The results indicated that curcumin induced cell cycle arrest at G2/M phase by downregulation of cyclin B1 and Cdc2 and inhibited colony formation in MCF-7wt cells. However, Bcl-2 overexpression prevented the inhibition of cell cycle associated proteins after curcumin treatment. The combination of LY294002, PI3K inhibitor, and curcumin induced cell cycle arrest by decreasing CDK4, CDK2 and cyclin E2 in Bcl-2+ MCF-7 cells. Moreover, LY294002 further inhibited the phosphorylation of Akt in Bcl-2+ MCF-7 cells. Curcumin could suppress the nuclear transport of NFκB through decreasing the interaction of P-IκB-NFκB. The combination of wedelolactone, NFκB inhibitor, and curcumin acted different on SSAT expression in wt MCF-7 and Bcl-2+ MCF-7 cells. NFκB inhibition increased the SSAT after curcumin treatment in Bcl-2 overexpressed MCF-7 cells. Inhibition of NFκB activity as well as suppression of ROS generation with NAC resulted in the partial relief of cells from G2/M checkpoint after curcumin treatment in wt MCF-7 cells. In conclusion, the potential role of curcumin in induction of cell cycle arrest is related with NFκB-regulated polyamine biosynthesis.
Collapse
|
314
|
Hensler M, Vančurová I, Becht E, Palata O, Strnad P, Tesařová P, Čabiňaková M, Švec D, Kubista M, Bartůňková J, Špíšek R, Sojka L. Gene expression profiling of circulating tumor cells and peripheral blood mononuclear cells from breast cancer patients. Oncoimmunology 2015; 5:e1102827. [PMID: 27141386 PMCID: PMC4839342 DOI: 10.1080/2162402x.2015.1102827] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/15/2015] [Accepted: 09/26/2015] [Indexed: 01/26/2023] Open
Abstract
Circulating tumor cells (CTCs) are cancer cells that are released from a tumor into the bloodstream. The presence of CTCs in peripheral blood has been associated with metastasis formation in patients with breast cancer. Therefore, the molecular characterization of CTCs may improve diagnostics and support treatment decisions. We performed gene expression profiling to evaluate the enriched CTCs and peripheral blood mononuclear cells (PBMCs) of breast cancer patients using an expression panel of 55 breast cancer-associated genes. The study revealed several significantly differentially expressed genes in the CTC-positive samples, including a few that were exclusively expressed in these cells. However, the expression of these genes was barely detectable in the PBMC samples. Some genes were differentially expressed in PBMCs, and the expression of these genes was correlated with tumor grade and the formation of metastasis. In this study, we have shown that the enriched CTCs of breast cancer patients overexpress genes involved in proteolytic degradation of the extracellular matrix (ECM) as well as genes that play important roles in the epithelial-mesenchymal transition (EMT) process that may occur in these cells.
Collapse
Affiliation(s)
| | - Irena Vančurová
- Sotio, a.s., Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Etienne Becht
- Laboratory of Cancer, Immune Control and Escape, UMRS 1138 INSERM, Cordeliers Research Center , Paris, France
| | | | - Pavel Strnad
- Department of Gynecology and Obstetrics, Second Faculty of Medicine, Charles University and University Hospital Motol , Prague, Czech Republic
| | - Petra Tesařová
- Oncology Clinic, First Faculty of Medicine, Charles University , Prague, Czech Republic
| | - Michaela Čabiňaková
- Oncology Clinic, First Faculty of Medicine, Charles University , Prague, Czech Republic
| | - David Švec
- TATAA Biocenter, Göteborg, Sweden; Laboratory of Gene Expression, Institute of Biotechnology, Academy of Sciences of the Czech Republic, v.v.i., Prague, Czech Republic
| | - Mikael Kubista
- TATAA Biocenter, Göteborg, Sweden; Laboratory of Gene Expression, Institute of Biotechnology, Academy of Sciences of the Czech Republic, v.v.i., Prague, Czech Republic
| | - Jiřina Bartůňková
- Sotio, a.s., Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Radek Špíšek
- Sotio, a.s., Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Luděk Sojka
- Sotio, a.s., Prague, Czech Republic; Department of Immunology, Charles University, Second Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
315
|
Carnagarin R, Dharmarajan AM, Dass CR. Molecular aspects of glucose homeostasis in skeletal muscle--A focus on the molecular mechanisms of insulin resistance. Mol Cell Endocrinol 2015; 417:52-62. [PMID: 26362689 DOI: 10.1016/j.mce.2015.09.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 09/02/2015] [Indexed: 01/14/2023]
Abstract
Among all the varied actions of insulin, regulation of glucose homeostasis is the most critical and intensively studied. With the availability of glucose from nutrient metabolism, insulin action in muscle results in increased glucose disposal via uptake from the circulation and storage of excess, thereby maintaining euglycemia. This major action of insulin is executed by redistribution of the glucose transporter protein, GLUT4 from intracellular storage sites to the plasma membrane and storage of glucose in the form of glycogen which also involves modulation of actin dynamics that govern trafficking of all the signal proteins of insulin signal transduction. The cellular mechanisms responsible for these trafficking events and the defects associated with insulin resistance are largely enigmatic, and this review provides a consolidated overview of the various molecular mechanisms involved in insulin-dependent glucose homeostasis in skeletal muscle, as insulin resistance at this major peripheral site impacts whole body glucose homeostasis.
Collapse
Affiliation(s)
- Revathy Carnagarin
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Pharmacy, Curtin University, Bentley 6102, Australia
| | - Arun M Dharmarajan
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Biomedical Science, Curtin University, Bentley 6102, Australia
| | - Crispin R Dass
- Curtin Biosciences Research Precinct, Bentley 6102, Australia; School of Pharmacy, Curtin University, Bentley 6102, Australia.
| |
Collapse
|
316
|
Peng W, Tu ZC, Long ZJ, Liu Q, Lu G. Discovery of 2-(2-aminopyrimidin-5-yl)-4-morpholino-N-(pyridin-3-yl)quinazolin-7-amines as novel PI3K/mTOR inhibitors and anticancer agents. Eur J Med Chem 2015; 108:644-654. [PMID: 26731167 DOI: 10.1016/j.ejmech.2015.11.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/19/2015] [Accepted: 11/21/2015] [Indexed: 10/22/2022]
Abstract
In this study, a series of novel 7 or 8-substituted 4-morpholine-quinazoline derivatives was designed and synthesized. Their PI3Kα inhibitory activities, antiproliferative activities against seven cancer cell lines, namely, PC-3, DU145, MCF-7, BT474, SK-BR-3, U937 and A431, were evaluated in vitro. Compound 17f proved to be a potential drug candidate with high PI3Kα inhibition activity (IC50 = 4.2 nM) and good antiproliferative activity. Compound 17f was also tested for its inhibitory activities against other kinases, such as PI3Kβ, PI3Kγ, PI3Kδ and mTOR, its effects on p-Akt (S473) and cell cycle. These results suggested that compound 17f could significantly inhibit the PI3K/Akt/mTOR pathway as a potent PI3K inhibitor and anticancer agent.
Collapse
Affiliation(s)
- Wei Peng
- Institute of Drug Synthesis and Pharmaceutical Process, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Zheng-Chao Tu
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, PR China
| | - Zi-Jie Long
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | - Quentin Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | - Gui Lu
- Institute of Drug Synthesis and Pharmaceutical Process, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China; Institute of Human Virology, Sun Yat-sen University, Guangzhou 510080, PR China.
| |
Collapse
|
317
|
Munster P, Aggarwal R, Hong D, Schellens JHM, van der Noll R, Specht J, Witteveen PO, Werner TL, Dees EC, Bergsland E, Agarwal N, Kleha JF, Durante M, Adams L, Smith DA, Lampkin TA, Morris SR, Kurzrock R. First-in-Human Phase I Study of GSK2126458, an Oral Pan-Class I Phosphatidylinositol-3-Kinase Inhibitor, in Patients with Advanced Solid Tumor Malignancies. Clin Cancer Res 2015; 22:1932-9. [PMID: 26603258 DOI: 10.1158/1078-0432.ccr-15-1665] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/12/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE GSK2126458 (GSK458) is a potent inhibitor of PI3K (α, β, γ, and δ), with preclinical studies demonstrating broad antitumor activity. We performed a first-in-human phase I study in patients with advanced solid tumors. MATERIALS AND METHODS Patients received oral GSK458 once or twice daily in a dose-escalation design to define the maximum tolerated dose (MTD). Expansion cohorts evaluated pharmacodynamics, pharmacokinetics, and clinical activity in histologically and molecularly defined cohorts. RESULTS One hundred and seventy patients received doses ranging from 0.1 to 3 mg once or twice daily. Dose-limiting toxicities (grade 3 diarrhea,n= 4; fatigue and rash,n= 1) occurred in 5 patients (n= 3 at 3 mg/day). The MTD was 2.5 mg/day (MTD with twice daily dosing undefined). The most common grade ≥3 treatment-related adverse events included diarrhea (8%) and skin rash (5%). Pharmacokinetic analyses demonstrated increased duration of drug exposure above target level with twice daily dosing. Fasting insulin and glucose levels increased with dose and exposure of GSK458. Durable objective responses (ORs) were observed across multiple tumor types (sarcoma, kidney, breast, endometrial, oropharyngeal, and bladder cancer). Responses were not associated withPIK3CAmutations (OR rate: 5% wild-type vs. 6% mutant). CONCLUSIONS Although the MTD of GSK458 was 2.5 mg once daily, twice-daily dosing may increase duration of target inhibition. Fasting insulin and glucose levels served as pharmacodynamic markers of drug exposure. Select patients achieved durable responses; however,PIK3CAmutations were neither necessary nor predictive of response. Combination treatment strategies and novel biomarkers may be needed to optimally target PI3K.
Collapse
Affiliation(s)
| | | | - David Hong
- MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | - Theresa L Werner
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - E Claire Dees
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina
| | | | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | | | | | - Laurel Adams
- GlaxoSmithKline, Research Triangle Park, North Carolina
| | | | | | | | | |
Collapse
|
318
|
Zhang N, Chen X. Potential role of O-GlcNAcylation and involvement of PI3K/Akt1 pathway in the expression of oncogenic phenotypes of gastric cancer cells in vitro. Biotechnol Appl Biochem 2015; 63:841-851. [PMID: 26333304 DOI: 10.1002/bab.1441] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/26/2015] [Indexed: 12/22/2022]
Abstract
O-GlcNAcylation is a monosaccharide modification by a residue of N-acetylglucosamine (GlcNAc) attached to serine or threonine moieties on nuclear and cytoplasmic proteins. O-GlcNAcylation is dynamically regulated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Increasing evidence suggests that O-GlcNAcylation is involved in a variety of human cancers. However, the exact role of O-GlcNAcylation in tumor progression remains unclear. Here, we show that O-GlcNAcylation accelerates oncogenic phenotypes of gastric cancer. First, cell models with increased or decreased O-GlcNAcylation were constructed by OGT overexpression, downregulation of OGA activity with specific inhibitor Thiamet-G, or silence of OGT. MTT assays indicated that O-GlcNAcylation increased proliferation of gastric cancer cells. Soft agar assay and Transwell assays showed that O-GlcNAcylation significantly enhanced cellular colony formation, migration, and invasion in vitro. Akt1 activity was stimulated by upregulation of phosphorylation at Ser473 mediated by elevated O-GlcNAcylation. The enhanced cell invasion by Thiamet-G treatment was suppressed by PI3K inhibitor LY294002. Although the cell invasion induced by Thiamet-G was reduced by Akt1 shRNA, it was still higher in comparison with that to the control (cells with Akt1 shRNA alone). And Akt1 overexpression promoted Thiamet-G-induced cell invasion. These results suggested that O-GlcNAcylation enhanced oncogenic phenotypes possibly partially involving PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Nuobei Zhang
- Department of Gastroenterology, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi, People's Republic of China
| | - Xin Chen
- Department of Nuclear Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi, People's Republic of China
| |
Collapse
|
319
|
Zhang P, Wang C, Ma T, You S. O-GlcNAcylation enhances the invasion of thyroid anaplastic cancer cells partially by PI3K/Akt1 pathway. Onco Targets Ther 2015; 8:3305-13. [PMID: 26635480 PMCID: PMC4646590 DOI: 10.2147/ott.s82845] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background The PI3K family participates in multiple signaling pathways to regulate cellular functions. PI3K/Akt signaling pathway plays an important role in tumorigenesis and development. O-GlcNAcylation, a posttranslational modification, is thought to modulate a wide range of biological processes, such as transcription, cell growth, signal transduction, and cell motility. O-GlcNAcylation is catalyzed by the nucleocytoplasmic enzymes, OGT and OGA, which adds or removes O-GlcNAc moieties, respectively. Abnormal O-GlcNAcylation has been implicated in a variety of human diseases. However, the role of O-GlcNAcylation in tumorigenesis and progression of cancer is still under-investigated. Understanding the O-GlcNAc-associated molecular mechanism might be significant for diagnosis and therapy of cancer. Methods Human thyroid anaplastic cancer 8305C cells were used to evaluate the role of O-GlcNAcylation in tumorigenesis and progression of cancer. The global O-GlcNAc level of intracellular proteins was up-regulated by OGA inhibitor Thiamet-G treatment or OGT over-expression. Cell proliferation was assessed by MTT assay. Invasion in vitro was determined by Transwell assay, and phosphorylation of Akt1 at Ser473 was assessed by Western blot for activity of Akt1. PI3K-specific inhibitor LY294002 and RNA interference of Akt1 were used to investigate the impact of PI3K/Akt signaling on the regulation of O-GlcNAcylation during tumor progression. Results Cell models with remarkably up-regulated O-GlcNAcylation were constructed, and then cell proliferation and invasion were determined. The results indicated that the proliferation was not affected by OGA inhibition or OGT overexpression, while the invasion of 8305C cells with OGA inhibition or OGT overexpression was obviously increased. Akt1 activity was stimulated by elevated O-GlcNAcylation by mediating phosphorylation at Ser473. The enhanced invasion of thyroid cancer cells by Thiamet-G treatment or OGT overexpression was significantly depressed by PI3K inhibitor LY294002. Moreover, silence of Akt1 remarkably attenuated the increase of cell invasion induced by Thiamet-G treatment, but the invasion was still higher compared to Akt1-silenced only cells. In other words, Thiamet-G restored the invasion of Akt1-silenced thyroid cancer cells, but it was still lower relative to Thiamet-G-treated only cells. Conclusion Taken together, our findings suggested that O-GlcNAcylation enhanced the invasion of thyroid anaplastic cancer cells partially by PI3K/Akt signaling, which might be a potential target for the diagnosis and treatment of thyroid anaplastic cancer.
Collapse
Affiliation(s)
- Peng Zhang
- General Surgery Department, General Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Chunli Wang
- General Surgery Department, General Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Tao Ma
- General Surgery Department, General Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Shengyi You
- General Surgery Department, General Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
320
|
Pérez-Ramírez C, Cañadas-Garre M, Molina MÁ, Faus-Dáder MJ, Calleja-Hernández MÁ. PTEN and PI3K/AKT in non-small-cell lung cancer. Pharmacogenomics 2015; 16:1843-62. [DOI: 10.2217/pgs.15.122] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of cancer deaths worldwide. In the last years, the identification of activating EGFR mutations, conferring increased sensitivity and disease response to tyrosine kinase inhibitors, has changed the prospect of NSCLC patients. The PTEN/PI3K/AKT pathway regulates multiple cellular functions, including cell growth, differentiation, proliferation, survival, motility, invasion and intracellular trafficking. Alterations in this pathway, mainly PTEN inactivation, have been associated with resistance to EGFR-tyrosine kinase inhibitor therapy and lower survival in NSCLC patients. In this review, we will briefly discuss the main PTEN/PI3K/AKT pathway alterations found in NSCLC, as well as the cell processes regulated by PTEN/PI3K/AKT leading to tumorigenesis.
Collapse
Affiliation(s)
- Cristina Pérez-Ramírez
- Pharmacogenetics Unit. UGC Provincial de Farmacia de Granada. Instituto de Investigación Biosanitaria de Granada. Complejo Hospitalario Universitario de Granada. Avda. Fuerzas Armadas, 2. 18014 Granada, Spain
- Department of Biochemistry. Faculty of Pharmacy. University of Granada Campus Universitario de Cartuja, s/n. 18071 Granada, Spain
| | - Marisa Cañadas-Garre
- Pharmacogenetics Unit. UGC Provincial de Farmacia de Granada. Instituto de Investigación Biosanitaria de Granada. Complejo Hospitalario Universitario de Granada. Avda. Fuerzas Armadas, 2. 18014 Granada, Spain
| | - Miguel Ángel Molina
- PANGAEA BIOTECH, S.L. Hospital Universitario Quirón Dexeus. C/Sabino Arana, 5-19. 08028 Barcelona
| | - María José Faus-Dáder
- Department of Biochemistry. Faculty of Pharmacy. University of Granada Campus Universitario de Cartuja, s/n. 18071 Granada, Spain
| | - Miguel Ángel Calleja-Hernández
- Pharmacogenetics Unit. UGC Provincial de Farmacia de Granada. Instituto de Investigación Biosanitaria de Granada. Complejo Hospitalario Universitario de Granada. Avda. Fuerzas Armadas, 2. 18014 Granada, Spain
- Department of Pharmacology. Faculty of Pharmacy. University of Granada. Campus Universitario de Cartuja, s/n. 18071 Granada, Spain
| |
Collapse
|
321
|
Haidar M, Whitworth J, Noé G, Liu WQ, Vidal M, Langsley G. TGF-β2 induces Grb2 to recruit PI3-K to TGF-RII that activates JNK/AP-1-signaling and augments invasiveness of Theileria-transformed macrophages. Sci Rep 2015; 5:15688. [PMID: 26511382 PMCID: PMC4625156 DOI: 10.1038/srep15688] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/28/2015] [Indexed: 01/09/2023] Open
Abstract
Theileria-infected macrophages display many features of cancer cells such as heightened invasive capacity; however, the tumor-like phenotype is reversible by killing the parasite. Moreover, virulent macrophages can be attenuated by multiple in vitro passages and so provide a powerful model to elucidate mechanisms related to transformed macrophage virulence. Here, we demonstrate that in two independent Theileria-transformed macrophage cell lines Grb2 expression is down-regulated concomitant with loss of tumor virulence. Using peptidimer-c to ablate SH2 and SH3 interactions of Grb2 we identify TGF-receptor II and the p85 subunit of PI3-K, as Grb2 partners in virulent macrophages. Ablation of Grb2 interactions reduces PI3-K recruitment to TGF-RII and decreases PIP3 production, and dampens JNK phosphorylation and AP-1-driven transcriptional activity down to levels characteristic of attenuated macrophages. Loss of TGF-R>PI3-K>JNK>AP-1 signaling negatively impacts on virulence traits such as reduced JAM-L/ITG4A and Fos-B/MMP9 expression that contribute to virulent macrophage adhesion and invasiveness.
Collapse
Affiliation(s)
- Malak Haidar
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médicine, Université Paris Descartes - Sorbonne Paris Cité, France.,Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris, 75014 France
| | - Jessie Whitworth
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médicine, Université Paris Descartes - Sorbonne Paris Cité, France.,Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris, 75014 France
| | - Gaelle Noé
- UF Pharmacocinétique et pharmacochimie Hôpital Cochin, Paris, France Assistance Publique Hôpitaux de Paris.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Wang Qing Liu
- UF Pharmacocinétique et pharmacochimie Hôpital Cochin, Paris, France Assistance Publique Hôpitaux de Paris.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Michel Vidal
- UF Pharmacocinétique et pharmacochimie Hôpital Cochin, Paris, France Assistance Publique Hôpitaux de Paris.,UMR8638 CNRS, Faculté de Pharmacie, Université Paris Descartes, PRES Sorbonne Paris Cité, Paris, France
| | - Gordon Langsley
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médicine, Université Paris Descartes - Sorbonne Paris Cité, France.,Inserm U1016, Cnrs UMR8104, Cochin Institute, Paris, 75014 France
| |
Collapse
|
322
|
Lv D, Yang H, Wang W, Xie Y, Hu W, Ye M, Chen X. High PHLPP expression is associated with better prognosis in patients with resected lung adenocarcinoma. BMC Cancer 2015; 15:687. [PMID: 26463718 PMCID: PMC4604720 DOI: 10.1186/s12885-015-1711-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 10/08/2015] [Indexed: 01/19/2023] Open
Abstract
Background PH domain Leucine-rich-repeats protein phosphatase (PHLPP) is a novel family of Ser/Thr protein dephosphatases that play a critical role in maintaining the balance in cell signaling. PHLPP negatively regulates PI3K/Akt and RAF/RAS/′ signaling activation, which is crucial in development, growth, and proliferation of lung cancer. The aim of this study was to investigate the association of PHLPP expression with biological behavior and prognosis of lung adenocarcinoma. Methods One hundred and fifty eight patients with pathologically documented stage I, II or IIIA lung adenocarcinoma were recruited in this study. Expression of PHLPP, p-AKT and p-ERK were evaluated by immunohistochemistry (IHC) in paraffin-embedded resected specimens. The correlation of their expression, which was dichotomized to low expression (a score of 0, 1) versus high expression (a score of 2, 3), with the clinicopathological parameters and prognosis of the patients also analyzed. Results High PHLPP expression rate in lung adenocarcinoma was 23.4 %. PHLPP expression level was significantly associated with tumor differentiation (p = 0.025) and tumor stage (p = 0.024). Patients with high expression of PHLPP showed significantly longer average survival time and higher 3 years survival rate than those with low expression of PHLPP (45 months versus 38 months, 85.8 % versus 73.5 % respectively) (Log rank test x2 = 7.086, p =0.008). A significant inverse correlation was observed between PHLPP expression and p-AKT (r = −0.523, p = 0.000) or p-ERK (r = −0.530, p = 0.000). Conclusion Our results suggest that high levels of PHLPP might reflect a less aggressive lung adenocarcinoma phenotype and predict better survival in patients with lung adenocarcinoma. PHLPP can be a potential prognostic marker to screen patients for favorable prognoses.
Collapse
Affiliation(s)
- Dongqing Lv
- Laboratory of Cellular and Molecular Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China. .,Department of Pulmonary Medicine, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China.
| | - Haihua Yang
- Laboratory of Cellular and Molecular Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China. .,Department of Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China.
| | - Wei Wang
- Laboratory of Cellular and Molecular Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China. .,Department of Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China.
| | - Youyou Xie
- Laboratory of Cellular and Molecular Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China. .,Department of Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China.
| | - Wei Hu
- Laboratory of Cellular and Molecular Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China. .,Department of Radiation Oncology, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China.
| | - Minhua Ye
- Department of Thoracic Surgery, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China.
| | - Xiaofeng Chen
- Enze Medical Research Center, Taizhou Hospital, Wenzhou Medical University, Zhejiang Province, 317000, China.
| |
Collapse
|
323
|
Neuroleukin/Autocrine Motility Factor Receptor Pathway Promotes Proliferation of Articular Chondrocytes through Activation of AKT and Smad2/3. Sci Rep 2015; 5:15101. [PMID: 26459914 PMCID: PMC4602231 DOI: 10.1038/srep15101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 09/15/2015] [Indexed: 12/12/2022] Open
Abstract
Cartilage defect is an intractable clinical problem. Therapeutic strategies for cartilage repair are far from optimal due to poor proliferation capacity of chondrocytes. Autologous chondrocyte implantation is a cell based therapy that uses in vitro amplified healthy chondrocytes from the patient. However, chondrocyte dedifferentiation during in vitro culture limits its application. Neuroleukin (NLK) is a multifunctional protein that stimulates cell growth and migration, together with its receptor autocrine motility factor receptor (AMFR, also called gp78). We investigated expression of NLK and AMFR/gp78 during cartilage development in vivo and in cultured articular chondrocytes in vitro, and found the pair associates with chondrocyte proliferation and differentiation. While applied to isolated articular chondrocytes, NLK promotes cell proliferation and secretion of type II collagen, a marker of proliferating chondrocytes. Further work demonstrates that NLK up regulates pAKT and pSmad2/3, but down regulates pSmad1/5. In animals, NLK treatment also promotes chondrocyte proliferation while inhibits terminal differentiation, leading to expanded proliferating zone but decreased prehypertrophic and hypertrophic zones in the growth plate region. NLK is therefore a candidate factor that can be applied in the treatment of cartilage defects.
Collapse
|
324
|
Sheen MR, Warner SL, Fields JL, Conejo-Garcia JR, Fiering S. Myristoylated p110α Causes Embryonic Death Due to Developmental and Vascular Defects. Open Life Sci 2015; 10:461-478. [PMID: 27482546 PMCID: PMC4966669 DOI: 10.1515/biol-2015-0048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway regulates many important cellular functions. The functional impact of deregulating the PIK3CA gene, encoding the p110α catalytic subunit of PI3K, is validated by frequent gain of function mutations in a range of human cancers. We generated a mouse model with an inducible constitutively active form of PI3K. In this model Cre recombinase activates expression of a myristoylated form of p110α (myr-p110α). The myristoylated version of p110α brings the protein to the cytoplasmic side of the cell membrane, which mimics the normal activation mechanism for the p110α catalytic subunit and activates the PI3K enzyme. Constitutively activated PI3K signaling induced by myr-p110α in all cells of the developing mouse caused lethality during embryonic development. Transgenic Cre;myr-p110α heterozygous embryos displayed morphological malformation and poor vascular development with extremely dilated blood vessels and hemorrhage in the embryo and the extraembryonic yolk sac. Previous studies demonstrated that loss of p110α during embryonic development causes angiogenic disruption and here we show that constitutive activation of p110α by gain of function mutation during development also disrupts vasculogenesis/angiogenesis in what appears to be a similar manner. These finding demonstrate the importance of tight regulation of PI3K signaling during embryonic vasculogenesis/angiogenesis..
Collapse
Affiliation(s)
- Mee Rie Sheen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, United States
| | - Sandra L Warner
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, United States; Norris Cotton Cancer Center, Lebanon, NH 03756, United States
| | - Jennifer L Fields
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, United States; Norris Cotton Cancer Center, Lebanon, NH 03756, United States
| | - Jose R Conejo-Garcia
- umor Microenvironment and Metastasis Program, the Wistar Institute, Philadelphia, PA 19104, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, United States; Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, United States
| |
Collapse
|
325
|
Galli U, Ciraolo E, Massarotti A, Margaria JP, Sorba G, Hirsch E, Tron GC. The Guareschi Pyridine Scaffold as a Valuable Platform for the Identification of Selective PI3K Inhibitors. Molecules 2015; 20:17275-87. [PMID: 26393561 PMCID: PMC6332036 DOI: 10.3390/molecules200917275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/09/2015] [Accepted: 09/14/2015] [Indexed: 11/16/2022] Open
Abstract
A novel series of 4-aryl-3-cyano-2-(3-hydroxyphenyl)-6-morpholino-pyridines have been designed as potential phosphatidylinositol-3-kinase (PI3K) inhibitors. The compounds have been synthesized using the Guareschi reaction to prepare the key 4-aryl-3-cyano-2,6-dihydroxypyridine intermediate. A different selectivity according to the nature of the aryl group has been observed. Compound 9b is a selective inhibitor against the PI3Kα isoform, maintaining a good inhibitory activity. Docking studies were also performed in order to rationalize its profile of selectivity.
Collapse
Affiliation(s)
- Ubaldina Galli
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, Novara 28100, Italy.
| | - Elisa Ciraolo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy.
| | - Alberto Massarotti
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, Novara 28100, Italy.
| | - Jean Piero Margaria
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy.
| | - Giovanni Sorba
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, Novara 28100, Italy.
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy.
| | - Gian Cesare Tron
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale "A. Avogadro", Largo Donegani 2, Novara 28100, Italy.
| |
Collapse
|
326
|
Kriplani N, Hermida MA, Brown ER, Leslie NR. Class I PI 3-kinases: Function and evolution. Adv Biol Regul 2015; 59:53-64. [PMID: 26159297 DOI: 10.1016/j.jbior.2015.05.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 05/25/2015] [Indexed: 06/04/2023]
Abstract
In many human cell types, the class I phosphoinositide 3-kinases play key roles in the control of diverse cellular processes including growth, proliferation, survival and polarity. This is achieved through their activation by many cell surface receptors, leading to the synthesis of the phosphoinositide lipid signal, PIP3, which in turn influences the function of numerous direct PIP3-binding proteins. Here we review PI3K pathway biology and analyse the evolutionary distribution of its components and their functions. The broad phylogenetic distribution of class I PI3Ks in metazoa, amoebozoa and choannoflagellates, implies that these enzymes evolved in single celled organisms and were later co-opted into metazoan intercellular communication. A similar distribution is evident for the AKT and Cytohesin groups of downstream PIP3-binding proteins, with other effectors and pathway components appearing to evolve later. The genomic and functional phylogeny of regulatory systems such as the PI3K pathway provides a framework to improve our understanding of the mechanisms by which key cellular processes are controlled in humans.
Collapse
Affiliation(s)
- Nisha Kriplani
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Miguel A Hermida
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Euan R Brown
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Nicholas R Leslie
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, EH14 4AS, UK.
| |
Collapse
|
327
|
Xie XX, Li H, Wang J, Mao S, Xin MH, Lu SM, Mei QB, Zhang SQ. Synthesis and anticancer effects evaluation of 1-alkyl-3-(6-(2-methoxy-3-sulfonylaminopyridin-5-yl)benzo[d]thiazol-2-yl)urea as anticancer agents with low toxicity. Bioorg Med Chem 2015; 23:6477-85. [PMID: 26321603 DOI: 10.1016/j.bmc.2015.08.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/12/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023]
Abstract
As a PI3K and mTOR dual inhibitor, N-(2-chloro-5-(2-acetylaminobenzo[d]thiazol-6-yl)pyridin-3-yl)-4-fluorophenylsulfonamide displays toxicity when orally administrated. In the present study, alkylurea moiety replaced the acetamide group in the compound and a series of 1-alkyl-3-(6-(2,3-disubstituted pyridin-5-yl)benzo[d]thiazol-2-yl)urea derivatives were synthesized. The antiproliferative activities of the synthesized compounds in vitro were evaluated against HCT116, MCF-7, U87 MG and A549 cell lines. The compounds with potent antiproliferative activity were tested for their acute oral toxicity and inhibitory activity against PI3Ks and mTORC1. The results indicate that the compound attached a 2-(dialkylamino)ethylurea moiety at the 2-positeion of benzothiazole can retain the antiproliferative activity and inhibitory activity against PI3K and mTOR. In addition, their acute oral toxicity reduced dramatically. Moreover, compound 2f can effectively inhibit tumor growth in a mice S180 homograft model. These findings suggest that 1-(2-dialkylaminoethyl)-3-(6-(2-methoxy-3-sulfonylaminopyridin-5-yl)benzo[d]thiazol-2-yl)urea derivatives can serve as potent PI3K inhibitors and anticancer agents with low toxicity.
Collapse
Affiliation(s)
- Xiao-Xiao Xie
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Huan Li
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Juan Wang
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, PR China
| | - Shuai Mao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Min-Hang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - She-Min Lu
- Department of Genetics and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Qi-Bing Mei
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
328
|
A tryptophan derivative TD-26 attenuates thrombus formation by inhibiting both PI3K/Akt signaling and binding of fibrinogen to integrin αIIbβ3. Biochem Biophys Res Commun 2015; 465:516-22. [PMID: 26278818 DOI: 10.1016/j.bbrc.2015.08.051] [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: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 01/08/2023]
Abstract
The incidence and mortality of thrombotic disorders are rapidly increasing worldwide. The existing antithrombotic drugs, however, are associated with side effects, especially bleeding complications. Therefore, there remains a need for the development of more effective and safer antithrombotic agents. In this study, we discovered a new synthetic tryptophan derivative TD-26, producing potent inhibitory effect on platelet aggregation while without causing obvious bleeding risk. It has been shown that TD-26 inhibited platelet aggregation induced by ADP, thrombin, U46619 and collagen in vitro and suppressed the platelet aggregation induced by ADP ex vivo. Mechanism studies indicated that TD-26 inhibited platelet adhesion to fibrinogen-coated surfaces, blocked the binding of fibrinogen to integrin αIIbβ3 and reduced Akt(Ser473) phosphorylation in platelet phosphatidylinositol 3-kinase (PI3K) signaling. Furthermore, TD-26 exhibited potent antithrombotic activity in vivo. In animal models, it decreased death of mice with acute pulmonary thrombosis by 90% and attenuated thrombosis weight by 60.3%, both at a dose of 3 mg/kg. Additionally, TD-26 did not obviously prolong bleeding time in mice. Taken together, our results reveal that TD-26 is a novel antithrombotic compound exhibiting both integrin αIIbβ3 inhibition and PI3K signaling blockage, with a low bleeding risk.
Collapse
|
329
|
Hu XF, Ye HW, Xin SY, He JM, Guo W. Effect of epidermal growth factor on insulin-PI3K signaling pathway and potential mechansim involved. Shijie Huaren Xiaohua Zazhi 2015; 23:3592-3596. [DOI: 10.11569/wcjd.v23.i22.3592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To examine the effect of epidermal growth factor (EGF) on the insulin-PI3K signalling pathway and the potential mechanism involved.
METHODS: HepG2 cells were cultured in Dulbecco's modified Eagle' s medium (DMEM) containing 10% fetal calf serum with the addition of L-glutamine. After overnight serum starvation, cells were stimulated with EGF or insulin for different periods of time. Cell protein expression was determined by Western blot.
RESULTS: Insulin stimulation caused Akt phosphorylation in HepG2 cells. Prestimulation with epidemimal growth factor for 30 min had no significant effect on PI3K signalling. In contrast, prestimulation with EGF for 4 h inhibited insulin induced activation of p-Akt. When cells were transfected with phosphatase and tensin homolog deleted on chromosome 10 (PTEN) mutant C124S or N-terminal deleted p85, EGF induced inhibition of phospho-Akt was reversed.
CONCLUSION: Interaction between EGF and insulin inhibits PI3K-Akt activation. The underlying mechanism involves PTEN and regulatory subunit p85. Mutations of PTEN and p85 reverse the inhibition.
Collapse
|
330
|
Chen J, Li L, Su J, Li B, Chen T, Ling F, Zhang X. Enhancing effect of natural borneol on the cellular uptake of demethoxycurcumin and their combined induction of G2/M arrest in HepG2 cells via ROS generation. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
331
|
Echeverria I, Liu Y, Gabelli SB, Amzel LM. Oncogenic mutations weaken the interactions that stabilize the p110α-p85α heterodimer in phosphatidylinositol 3-kinase α. FEBS J 2015; 282:3528-42. [PMID: 26122737 DOI: 10.1111/febs.13365] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/20/2015] [Accepted: 06/26/2015] [Indexed: 12/20/2022]
Abstract
Phosphatidylinositol 3-kinase (PI3K) α is a heterodimeric lipid kinase that catalyzes the conversion of phosphoinositol-4,5-bisphosphate to phosphoinositol-3,4,5-trisphosphate. The PI3Kα signaling pathway plays an important role in cell growth, proliferation, and survival. This pathway is activated in numerous cancers, where the PI3KCA gene, which encodes for the p110α PI3Kα subunit, is mutated. Its mutation often results in gain of enzymatic activity; however, the mechanism of activation by oncogenic mutations remains unknown. Here, using computational methods, we show that oncogenic mutations that are far from the catalytic site and increase the enzymatic affinity destabilize the p110α-p85α dimer. By affecting the dynamics of the protein, these mutations favor the conformations that reduce the autoinhibitory effect of the p85α nSH2 domain. For example, we determined that, in all of the mutants, the nSH2 domain shows increased positional heterogeneity as compared with the wild-type, as demonstrated by changes in the fluctuation profiles computed by normal mode analysis of coarse-grained elastic network models. Analysis of the interdomain interactions of the wild-type and mutants at the p110α-p85α interface obtained with molecular dynamics simulations suggest that all of the tumor-associated mutations effectively weaken the interactions between p110α and p85α by disrupting key stabilizing interactions. These findings have important implications for understanding how oncogenic mutations change the conformational multiplicity of PI3Kα and lead to increased enzymatic activity. This mechanism may apply to other enzymes and/or macromolecular complexes that play a key role in cell signaling.
Collapse
Affiliation(s)
- Ignacia Echeverria
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy, University of California at San Francisco, CA, USA.,Department of Pharmaceutical Chemistry, School of Pharmacy, University of California at San Francisco, CA, USA.,Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yunlong Liu
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sandra B Gabelli
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Medicine and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - L Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
332
|
Abstract
INTRODUCTION Myelofibrosis (MF) is a myeloproliferative neoplasm associated with significant disease burden composed of splenomegaly, constitutional symptoms and a reduced life expectancy. The advent of targeted treatments has provided new means by which to improve MF associated splenomegaly, symptoms, health-related quality of life and even mortality. AREAS COVERED We discuss the spectrum of targeted treatments currently under investigation for MF. We furthermore compare their effects on improving anemia, reducing fibrosis and splenomegaly and enhancing symptom control. EXPERT OPINION MF is a complex disorder, partly attributable to its heterogeneity. Although the severity of patient symptoms correlates with risk category, high symptom burden may also be observed in low-risk patients. Serial use of PRO tools allows clinicians to objectively evaluate the MF symptom burden, compare efficacy of therapies and adjust medications to improve symptom control. Novel targeted agents have proven superior to historic treatment regimens for symptom management. Promising treatment categories include JAK2 inhibitors, histone deacetylase inhibitors, hypomethylating agents, heat shock protein-90 inhibitors, hedgehog inhibitors, PI3-AKT-mTOR inhibitors, antifibrosing agents and telomerase inhibitors. The majority of therapies remain under investigation, either alone or in combination with other treatments. It is anticipated that these agents will be increasingly integrated into standard treatment algorithms for MF symptom management.
Collapse
Affiliation(s)
- Holly L Geyer
- a 1 Mayo Clinic, Department of Hospital Internal Medicine , 13400 E Shea Blvd, AZ, USA
| | - Ruben A Mesa
- b 2 Mayo Clinic, Division of Hematology and Oncology, Department of Hematology and Medical Oncology , 13400 E Shea Blvd, Scottsdale, AZ, USA +1 480 301 8335 ; +1 480 301 4675 ;
| |
Collapse
|
333
|
Cross-species DNA copy number analyses identifies multiple 1q21-q23 subtype-specific driver genes for breast cancer. Breast Cancer Res Treat 2015; 152:347-56. [PMID: 26109346 PMCID: PMC4491106 DOI: 10.1007/s10549-015-3476-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 06/15/2015] [Indexed: 11/17/2022]
Abstract
A large number of DNA copy number alterations (CNAs) exist in human breast cancers, and thus characterizing the most frequent CNAs is key to advancing therapeutics because it is likely that these regions contain breast tumor ‘drivers’ (i.e., cancer causal genes). This study aims to characterize the genomic landscape of breast cancer CNAs and identify potential subtype-specific drivers using a large set of human breast tumors and genetically engineered mouse (GEM) mammary tumors. Using a novel method called SWITCHplus, we identified subtype-specific DNA CNAs occurring at a 15 % or greater frequency, which excluded many well-known breast cancer-related drivers such as amplification of ERBB2, and deletions of TP53 and RB1. A comparison of CNAs between mouse and human breast tumors identified regions with shared subtype-specific CNAs. Additional criteria that included gene expression-to-copy number correlation, a DawnRank network analysis, and RNA interference functional studies highlighted candidate driver genes that fulfilled these multiple criteria. Numerous regions of shared CNAs were observed between human breast tumors and GEM mammary tumor models that shared similar gene expression features. Specifically, we identified chromosome 1q21-23 as a Basal-like subtype-enriched region with multiple potential driver genes including PI4KB, SHC1, and NCSTN. This step-wise computational approach based on a cross-species comparison is applicable to any tumor type for which sufficient human and model system DNA copy number data exist, and in this instance, highlights that a single region of amplification may in fact harbor multiple driver genes.
Collapse
|
334
|
Beyond conventional chemotherapy: Emerging molecular targeted and immunotherapy strategies in urothelial carcinoma. Cancer Treat Rev 2015; 41:699-706. [PMID: 26138514 DOI: 10.1016/j.ctrv.2015.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/17/2015] [Indexed: 01/20/2023]
Abstract
Advanced urothelial carcinoma is frequently lethal, and improvements in cytotoxic chemotherapy have plateaued. Recent technological advances allows for a comprehensive analysis of genomic alterations in a timely manner. The Cancer Genome Atlas (TCGA) study revealed that there are numerous genomic aberrations in muscle-invasive urothelial carcinoma, such as TP53, ARID1A, PIK3CA, ERCC2, FGFR3, and HER2. Molecular targeted therapies against similar genetic alterations are currently available for other malignancies, but their efficacy in urothelial carcinoma has not been established. This review describes the genomic landscape of malignant urothelial carcinomas, with an emphasis on the potential to prosecute these tumours by deploying novel targeted agents and immunotherapy in appropriately selected patient populations.
Collapse
|
335
|
Ma C, Zhu L, Wang J, He H, Chang X, Gao J, Shumin W, Yan T. Anti-inflammatory effects of water extract of Taraxacum mongolicum hand.-Mazz on lipopolysaccharide-induced inflammation in acute lung injury by suppressing PI3K/Akt/mTOR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2015; 168:349-55. [PMID: 25861954 DOI: 10.1016/j.jep.2015.03.068] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 03/09/2015] [Accepted: 03/13/2015] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Taraxacum mongolicum Hand.-Mazz is a famous medicinal plant in China, has been listed in the Pharmacopoeia of the People's Republic of China and used to treat infection, fever, upper respiratory tract infection, pneumonia, and other infectious diseases. This study aims to evaluate the possible mechanisms responsible for the anti-inflammation effects of water extract of T. mongolicum Hand.-Mazz (WETMHM) on lipopolysaccharide (LPS)-induced inflammatory in acute lung injury. MATERIALS AND METHODS Female BALB/c mice were randomly divided into five groups with 10 mice in each group: (1) control group (saline), (2) LPS group, (3) LPS+dexamethasone (LPS+Dex, 2mg/kg, administered by gavage), (4) LPS+WETMHM (5 g/kg, administered by gavage), (5) LPS+WETMHM (10 g/kg, administered by gavage). The cell counting in the bronchoalveolar lavage fluid (BALF) was measured. The animal lung edema degree was evaluated by wet/dry weight (W/D) ratio. The superoxidase dismutase (SOD) activity and myeloperoxidase (MPO) activity were assayed by SOD and MPO kits, respectively. The levels of inflammation mediators including tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 were assayed by an enzyme-linked immunosorbent assay method. Pathological changes of lung tissues were observed by hematoxylin and eosin (HE) staining. The levels of P-PI3K, PI3K, P-Akt, Akt, P-mTOR and mTOR were measured by Western blotting. RESULTS The data showed that treatment with the WETMHM inhibited LPS-induced inflammation: (1) WETMHM attenuated inflammation cell numbers in the BALF, (2) decreased protein levels of lung PI3K/Akt/mTOR, and (3) improved SOD activity and (4) inhibited MPO activity; (5) histological studies demonstrated that WETMHM substantially inhibited LPS-induced neutrophils in lung tissue. CONCLUSION The results indicated that the WETMHM had a protective effect on LPS-induced ALI in mice.
Collapse
Affiliation(s)
- Chunhua Ma
- Department of Physiology and Pharmacology, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu 210009, PR China
| | - Lingpeng Zhu
- Department of Physiology and Pharmacology, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu 210009, PR China
| | - Jing Wang
- Department of Physiology and Pharmacology, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu 210009, PR China
| | - He He
- Department of Physiology and Pharmacology, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu 210009, PR China
| | - Xiayun Chang
- Department of Physiology and Pharmacology, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu 210009, PR China
| | - Jin Gao
- Department of Physiology and Pharmacology, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu 210009, PR China
| | - Wang Shumin
- Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Tianhua Yan
- Department of Physiology and Pharmacology, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu 210009, PR China.
| |
Collapse
|
336
|
Zhao HF, Wang J, Tony To SS. The phosphatidylinositol 3-kinase/Akt and c-Jun N-terminal kinase signaling in cancer: Alliance or contradiction? (Review). Int J Oncol 2015; 47:429-36. [PMID: 26082006 DOI: 10.3892/ijo.2015.3052] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 04/27/2015] [Indexed: 11/05/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway and c-Jun N-terminal kinase (JNK) pathway are responsible for regulating a variety of cellular processes including cell growth, migration, invasion and apoptosis. These two pathways are essential to the development and progression of tumors. The dual roles of JNK signaling in apoptosis and tumor development determine the different interactions between the PI3K/Akt and JNK pathways. Activation of PI3K/Akt signaling can inhibit stress- and cytokine-induced JNK activation through Akt antagonizing and the formation of the JIP1-JNK module, as well as the activities of upstream kinases ASK1, MKK4/7 and MLK. On the other hand, hyperactivation of Akt and JNK is also found in cancers that harbor EGFR overexpression or loss of PTEN. Understanding the activation mechanism of PI3K/Akt and JNK pathways, as well as the interplays between these two pathways in cancer may contribute to the identification of novel therapeutic targets. In the present report, we summarized the current understanding of the PI3K/Akt and JNK signaling networks, as well as their biological roles in cancers. In addition, the interactions and regulatory network between PI3K/Akt and JNK pathways in cancer were discussed.
Collapse
Affiliation(s)
- Hua-Fu Zhao
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, P.R. China
| | - Jing Wang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, P.R. China
| | - Shing-Shun Tony To
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, P.R. China
| |
Collapse
|
337
|
Jain MV, Jangamreddy JR, Grabarek J, Schweizer F, Klonisch T, Cieślar-Pobuda A, Łos MJ. Nuclear localized Akt enhances breast cancer stem-like cells through counter-regulation of p21(Waf1/Cip1) and p27(kip1). Cell Cycle 2015; 14:2109-20. [PMID: 26030190 DOI: 10.1080/15384101.2015.1041692] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Cancer stem-like cells (CSCs) are a rare subpopulation of cancer cells capable of propagating the disease and causing cancer recurrence. In this study, we found that the cellular localization of PKB/Akt kinase affects the maintenance of CSCs. When Akt tagged with nuclear localization signal (Akt-NLS) was overexpressed in SKBR3 and MDA-MB468 cells, these cells showed a 10-15% increase in the number of cells with CSCs enhanced ALDH activity and demonstrated a CD44(+High)/CD24(-Low) phenotype. This effect was completely reversed in the presence of Akt-specific inhibitor, triciribine. Furthermore, cells overexpressing Akt or Akt-NLS were less likely to be in G0/G1 phase of the cell cycle by inactivating p21(Waf1/Cip1) and exhibited increased clonogenicity and proliferation as assayed by colony-forming assay (mammosphere formation). Thus, our data emphasize the importance the intracellular localization of Akt has on stemness in human breast cancer cells. It also indicates a new robust way for improving the enrichment and culture of CSCs for experimental purposes. Hence, it allows for the development of simpler protocols to study stemness, clonogenic potency, and screening of new chemotherapeutic agents that preferentially target cancer stem cells. SUMMARY The presented data, (i) shows new, stemness-promoting role of nuclear Akt/PKB kinase, (ii) it underlines the effects of nuclear Akt on cell cycle regulation, and finally (iii) it suggests new ways to study cancer stem-like cells.
Collapse
Key Words
- 7-AAD, 7-aminoactinomycin D
- ALDH, aldehyde dehydrogenase
- Akt-NLS
- BPE, bovine pituitary epithelial
- Bcl2, B cell lymphoma 2
- CDK, cyclin-dependent kinase
- CSCs, cancer stem-like cells
- DEAB, diethylaminobenzaldehyde
- FBS, fetal bovine serum
- GAPDH, glucose-6-phosphate dehydrogenase
- GPCR, G-protein-coupled receptor
- GSK3, glycogen synthase kinase-3
- IGF1, insulin like growth factor 1
- JAK, Janus kinase
- NLS, nuclear localization signal
- PDK, phosphoinositide dependent kinase
- PH, pleckstrin-homology
- PI3K
- PI3K, phoshatidylinositol-3-kinase
- PKB, protein kinase B
- PTEN, phosphatase and tensin homolog
- PVDF, polyvinylidene fluoride
- RIPA, radioimmunoprecipitation
- RPMI, Roswell Park Memorial Institute
- RT, room temperature
- RTK, receptor tyrosine kinase
- STAT, signal transducer and activator of transcription
- T-ALL, T-cell acute lymphoblastic leukemia
- WT, wild type
- cancer stem-like cells
- hEGF, human epidermal growth factor
- mTOR
- mTOR, mammalian target of rapamycin
- poly-HEMA, poly-2-hydroxyethyl methacrylate
- stemness
Collapse
Affiliation(s)
- Mayur Vilas Jain
- a Department of Clinical & Experimental Medicine; Division of Cell Biology Integrative Regenerative Med. Center (IGEN); Linköping Univ. ; Linköping , Sweden
| | | | | | | | | | | | | |
Collapse
|
338
|
Gorai S, Bagdi PR, Borah R, Paul D, Santra MK, Khan AT, Manna D. Insights into the inhibitory mechanism of triazole-based small molecules on phosphatidylinositol-4,5-bisphosphate binding pleckstrin homology domain. Biochem Biophys Rep 2015; 2:75-86. [PMID: 29124147 PMCID: PMC5668642 DOI: 10.1016/j.bbrep.2015.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 12/11/2022] Open
Abstract
Background Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is an important regulator of several cellular processes and a precursor for other second messengers which are involved in cell signaling pathways. Signaling proteins preferably interact with PI(4,5)P2 through its pleckstrin homology (PH) domain. Efforts are underway to design small molecule-based antagonist, which can specifically inhibit the PI(4,5)P2/PH-domain interaction to establish an alternate strategy for the development of drug(s) for phosphoinositide signaling pathways. Methods Surface plasmon resonance, molecular docking, circular dichroism, competitive Förster resonance energy transfer, isothermal titration calorimetric analyses and liposome pull down assay were used. Results In this study, we employed 1,2,3-triazol-4-yl methanol containing small molecule (CIPs) as antagonists for PI(4,5)P2/PH-domain interaction and determined their inhibitory effect by using competitive-surface plasmon resonance analysis (IC50 ranges from 53 to 159 nM for PI(4,5)P2/PLCδ1-PH domain binding assay). We also used phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3], phosphatidylinositol 3,4-bisphosphate [PI(3,4)P2], PI(4,5)P2 specific PH-domains to determine binding selectivity of the compounds. Various physicochemical analyses showed that the compounds have weak affect on fluidity of the model membrane but, strongly interact with the phospholipase C δ1 (PLCδ1)-PH domains. The 1,2,3-triazol-4-yl methanol moiety and nitro group of the compounds are essential for their exothermic interaction with the PH-domains. Potent compound can efficiently displace PLCδ1-PH domain from plasma membrane to cytosol in A549 cells. Conclusions Overall, our studies demonstrate that these compounds interact with the PIP-binding PH-domains and inhibit their membrane recruitment. General significance These results suggest specific but differential binding of these compounds to the PLCδ1-PH domain and emphasize the role of their structural differences in binding parameters. These triazole-based compounds could be directly used/further developed as potential inhibitor for PH domain-dependent enzyme activity.
Collapse
Affiliation(s)
- Sukhamoy Gorai
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Prasanta Ray Bagdi
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Rituparna Borah
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Debasish Paul
- National Center for Cell Science, Pune 411007, Maharashtra, India
| | | | - Abu Taleb Khan
- Alia University, DN 18, 8th Floor, Sector V, Kolkata 700091, India
| | - Debasis Manna
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam 781039, India
| |
Collapse
|
339
|
Cho DC. Targeting the PI3K/Akt/mTOR pathway in malignancy: rationale and clinical outlook. BioDrugs 2015; 28:373-81. [PMID: 24567265 DOI: 10.1007/s40259-014-0090-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K) pathway, including major downstream effectors Akt and mammalian target of rapamycin (mTOR), plays a critical role in malignant transformation and subsequent processes of growth, proliferation, and metastases. Not surprisingly, the PI3K/Akt/mTOR pathway has emerged as an attractive drug target and numerous agents directed against various elements of the pathway are currently in clinical development. While early clinical trials with the first generations of these agents have shown limited single-agent efficacy, efforts are now focused on the development of more specific inhibitors, patient selection strategies, and combinational approaches. In this review, we discuss the PI3K/Akt/mTOR pathway in cancer, the rationale for its emergence as a therapeutic target, and progress thus far in the clinical development of inhibitors targeting its various elements.
Collapse
Affiliation(s)
- Daniel C Cho
- NYU Cancer Institute, NYU Langone Medical Center, 160 East 34th Street, New York, NY, 10016, USA,
| |
Collapse
|
340
|
Shehata M, Inokuchi K. Does autophagy work in synaptic plasticity and memory? Rev Neurosci 2015; 25:543-57. [PMID: 24651020 DOI: 10.1515/revneuro-2014-0002] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/17/2014] [Indexed: 12/12/2022]
Abstract
Many studies have reported the roles played by regulated proteolysis in neural plasticity and memory. Within this context, most of the research focused on the ubiquitin-proteasome system and the endosome-lysosome system while giving lesser consideration to another major protein degradation system, namely, autophagy. Although autophagy intersects with many of the pathways known to underlie synaptic plasticity and memory, only few reports related autophagy to synaptic remodeling. These pathways include PI3K-mTOR pathway and endosome-dependent proteolysis. In this review, we will discuss several lines of evidence supporting a physiological role of autophagy in memory processes, and the possible mechanistic scenarios for how autophagy could fulfill this function.
Collapse
|
341
|
The Flavonoid Apigenin Ameliorates Cisplatin-Induced Nephrotoxicity through Reduction of p53 Activation and Promotion of PI3K/Akt Pathway in Human Renal Proximal Tubular Epithelial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:186436. [PMID: 26089934 PMCID: PMC4454761 DOI: 10.1155/2015/186436] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/10/2015] [Indexed: 12/17/2022]
Abstract
Apigenin is a member of the flavone subclass of flavonoids present in fruits and vegetables. Apigenin has long been considered to have various biological activities, such as antioxidant, anti-inflammatory, and antitumorigenic properties, in various cell types. Cisplatin was known to exhibit cytotoxic effect to renal cells by inducing apoptosis through activation of p53. The present study investigated the antiapoptotic effects of apigenin on the cisplatin-treated human renal proximal tubular epithelial (HK-2) cells. HK-2 cells were pretreated with apigenin (5, 10, 20 μM) for 1 h and then treated with 40 μM cisplatin for various times. Apigenin inhibited the cisplatin-induced apoptosis of HK-2 cells. Interestingly, apigenin itself exerted cytostatic activity because of its ability to induce cell cycle arrest. Apigenin inhibited caspase-3 activity and PARP cleavage in cisplatin-treated cells. Apigenin reduced cisplatin-induced phosphorylation and expression of p53, with no significant influence on production of ROS that is known to induce p53 activation. Furthermore, apigenin promoted cisplatin-induced Akt phosphorylation, suggesting that enhanced Akt activation may be involved in cytoprotection. Taken together, these results suggest that apigenin ameliorates cisplatin-induced apoptosis through reduction of p53 activation and promotion of PI3K/Akt pathway in HK-2 cells.
Collapse
|
342
|
De Buck SS, Jakab A, Boehm M, Bootle D, Juric D, Quadt C, Goggin TK. Population pharmacokinetics and pharmacodynamics of BYL719, a phosphoinositide 3-kinase antagonist, in adult patients with advanced solid malignancies. Br J Clin Pharmacol 2015; 78:543-55. [PMID: 24617631 DOI: 10.1111/bcp.12378] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/26/2014] [Indexed: 12/14/2022] Open
Abstract
AIMS The aim was to characterize the population pharmacokinetics of BYL719 in cancer patients and assess the time course of tumour response in relation to drug exposure and dosing schedule. METHODS Plasma samples and longitudinal tumour size measurements were collected from 60 patients with advanced solid malignancies who received oral BYL719 once daily (30-450 mg) or twice daily at 120 mg or 200 mg. Non-linear mixed effect modelling was employed to develop the population pharmacokinetic and pharmacodynamic model. RESULTS The pharmacokinetics were best described by a one compartment disposition model and transit compartments accounting for the lag time in absorption. The typical population oral clearance and volume of distribution estimates with their between-subject variability (BSV) were 10 l h(-1) (BSV 26%) and 108 l (BSV 28%), respectively. The estimated optimal number of transit compartments was 8.1, with a mean transit time to the absorption compartment of 1.28 h (BSV 32%). The between-occasion variability in the rate and extent of absorption was 46% and 26%, respectively. Tumour growth was modelled using a turnover model characterized by a zero order growth rate of 0.581 cm week(1) and a first order death rate of 0.0123 week(-1) . BYL719 inhibited tumour growth with an IC50 of 100 ng ml(-1) (BSV 154%). Model-based predictions showed potential for additional anti-tumour activity of twice daily dosing at total daily dose below 400 mg, but a loss of efficacy if administered less frequently than once daily. CONCLUSIONS The proposed model provides a valuable approach for planning future clinical studies and for designing optimized dosing regimens with BYL719.
Collapse
Affiliation(s)
- Stefan S De Buck
- Oncology Clinical Pharmacology, Novartis Pharmaceuticals A.G., Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
343
|
Ma MY, Bai J, Chang WL, Tao KX. Effect of monoacylglycerol lipase inhibitor JZL184 on apoptosis of colorectal cancer cells. Shijie Huaren Xiaohua Zazhi 2015; 23:2256-2263. [DOI: 10.11569/wcjd.v23.i14.2256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether JZL184, a monoacylglycerol lipase inhibitor, induces apoptosis of colorectal cancer cells and to explore the possible mechanism.
METHODS: SW480 and Lovo cells were treated with JZL184, JZL184 + 5-fluorouracil (5-Fu) or 5-Fu alone for 48 h. Apoptosis was assessed by flow cytometry. The protein levels of p-AKT, p-mTOR, pro-Caspase3 and pro-Caspase8 were assessed by Western blot.
RESULTS: Treatment with JZL184 + 5-Fu increased SW480 and Lovo cell apoptosis more significantly than 5-Fu alone (apoptosis increase in SW480 cells: JZL184 + 500 μmol/L 5-Fu 13.91% ± 9.13%, JZL184 + 200 μmol/L 5-Fu 26.34% ± 13.32%, JZL184 + 100 μmol 5-Fu 43.32% ± 8.04%, JZL184 + 10 μmol 5-Fu 31.4% ± 5.82%; Lovo cells: JZL184 + 500 μmol/L 5-Fu 17.56% ± 8.14%, JZL184 + 200 μmol/L 5-Fu 33.04% ± 9.49%, JZL184 + 100 μmol/L 5-Fu 36.91% ± 16.63%, JZL184 + 10 μmol/L 5-Fu 21.26% ± 11.03%). Treatment with JZL184 significantly decreased the protein levels of p-AKT, p-mTOR, pro-Caspase3 and pro-Caspase8 in colorectal cancer SW480 and Lovo cells (P < 0.05).
CONCLUSION: JZL184 can inhibit the AKT-mTOR pathway and promote pro-Caspase8 and pro-Caspase3 activation to increase the apoptosis of SW480 and Lovo cells treated with 5-Fu.
Collapse
|
344
|
Wen R, Hu S, Xiao Q, Han C, Gan C, Gou H, Liu H, Li L, Xu H, He H, Wang J. Leptin exerts proliferative and anti-apoptotic effects on goose granulosa cells through the PI3K/Akt/mTOR signaling pathway. J Steroid Biochem Mol Biol 2015; 149:70-9. [PMID: 25576904 DOI: 10.1016/j.jsbmb.2015.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/08/2014] [Accepted: 01/03/2015] [Indexed: 01/30/2023]
Abstract
Leptin was known as a pivotal regulator for the control of food intake and energy expenditure. However, leptin has also been found to be involved in the regulation of female reproductive system through interactions with pathways in the hypothalamic-hypophyseal axis and direct action at the ovarian level. In the present study, granulosa cells from goose ovarian preovulatory (F1-F3) follicles were cultured with leptin (0, 1, 10 or 100ng/ml). The proliferative and anti-apoptotic actions of leptin in granulosa cells were revealed by CCK-8, BrdU and TUNEL assays. Quantitative real-time PCR and Western blot analyses further indicated that leptin treatment led to increased expression of cyclin D1, cyclin D2, cyclin D3 and bcl-2, and decreased expression of p21 and caspase-3. The effects were involved in the activation of the PI3K/Akt/mTOR signaling pathway, as leptin treatment enhanced the expression of PI3K, Akt1, Akt2, Raptor, mTOR, S6K and p-S6K. Moreover, blockade of the PI3K/Akt/mTOR pathway attenuated the influences of leptin on proliferation and apoptosis of granulosa cells, considering that activated factors by leptin were inhibited in the presence of either 20μM LY294002 (a PI3K inhibitor) or 10μM rapamycin (an mTOR inhibitor). In addition, leptin had a modulatory effect on the expression of its receptor at the transcriptional and translational levels, and blockade of PI3K/Akt/mTOR inhibited both basal and leptin-induced Lepr gene and protein expression. These findings suggest that leptin exerts its proliferative and anti-apoptotic effects on goose granulosa cells through the PI3K/Akt/mTOR signaling pathway via interaction with its receptor.
Collapse
Affiliation(s)
- Rui Wen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Qihai Xiao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Chao Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Hua Gou
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Hengyong Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, Sichuan 625014, PR China.
| |
Collapse
|
345
|
Balakrishnan K, Peluso M, Fu M, Rosin NY, Burger JA, Wierda WG, Keating MJ, Faia K, O'Brien S, Kutok JL, Gandhi V. The phosphoinositide-3-kinase (PI3K)-delta and gamma inhibitor, IPI-145 (Duvelisib), overcomes signals from the PI3K/AKT/S6 pathway and promotes apoptosis in CLL. Leukemia 2015; 29:1811-22. [PMID: 25917267 DOI: 10.1038/leu.2015.105] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/26/2015] [Accepted: 04/07/2015] [Indexed: 11/10/2022]
Abstract
The functional relevance of the B-cell receptor (BCR) and the evolution of protein kinases as therapeutic targets have recently shifted the paradigm for treatment of B-cell malignancies. Inhibition of p110δ with idelalisib has shown clinical activity in chronic lymphocytic leukemia (CLL). The dynamic interplay of isoforms p110δ and p110γ in leukocytes support the hypothesis that dual blockade may provide a therapeutic benefit. IPI-145, an oral inhibitor of p110δ and p110γ isoforms, sensitizes BCR-stimulated and/or stromal co-cultured primary CLL cells to apoptosis (median 20%, n=57; P<0.0001) including samples with poor prognostic markers, unmutated IgVH (n=28) and prior treatment (n=15; P<0.0001). IPI-145 potently inhibits the CD40L/IL-2/IL-10 induced proliferation of CLL cells with an IC50 in sub-nanomolar range. A corresponding dose-responsive inhibition of pAKT(Ser473) is observed with an IC50 of 0.36 nM. IPI-145 diminishes the BCR-induced chemokines CCL3 and CCL4 secretion to 17% and 37%, respectively. Pre-treatment with 1 μM IPI-145 inhibits the chemotaxis toward CXCL12; reduces pseudoemperipolesis to median 50%, inferring its ability to interfere with homing capabilities of CLL cells. BCR-activated signaling proteins AKT(Ser473), BAD(Ser112), ERK(Thr202/Tyr204) and S6(Ser235/236) are mitigated by IPI-145. Importantly, for clinical development in hematological malignancies, IPI-145 is selective to CLL B cells, sparing normal B- and T-lymphocytes.
Collapse
Affiliation(s)
- K Balakrishnan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Peluso
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - M Fu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - N Y Rosin
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J A Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K Faia
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - S O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J L Kutok
- Infinity Pharmaceuticals Inc., Cambridge, MA, USA
| | - V Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
346
|
Yip PY. Phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin (PI3K-Akt-mTOR) signaling pathway in non-small cell lung cancer. Transl Lung Cancer Res 2015; 4:165-76. [PMID: 25870799 DOI: 10.3978/j.issn.2218-6751.2015.01.04] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 12/30/2014] [Indexed: 12/13/2022]
Abstract
Non-small cell lung cancer (NSCLC) is a devastating disease with poor prognosis. Systemic chemotherapy has been the mainstay of treatment in advanced disease for many decades. Personalized targeted therapy such as epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) and crizotinib has significantly changed the treatment paradigm in NSCLC. The future success of development of molecular targeted therapy relies on the understanding of signal transduction pathways. The PI3K-Akt-mTOR pathway is commonly deregulated in human malignancy including NSCLC. Therefore, this pathway is a target for many therapeutic developments. This review will provide an overview of PI3K-Akt-mTOR signaling pathway, genetic alterations activating the pathway and clinical therapeutic development of pathway inhibitors.
Collapse
Affiliation(s)
- Po Yee Yip
- 1 Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia ; 2 Department of Medical Oncology, Macarthur Cancer Therapy Centre, Campbelltown, NSW, Australia
| |
Collapse
|
347
|
Wang XM, Xu J, Xin MH, Lu SM, Zhang SQ. Design, synthesis and antiproliferative activity evaluation of m -(4-morpholinyl-1,3,5-triazin-2-yl)benzamides in vitro. Bioorg Med Chem Lett 2015; 25:1730-1735. [DOI: 10.1016/j.bmcl.2015.02.067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/21/2015] [Accepted: 02/26/2015] [Indexed: 11/15/2022]
|
348
|
Gocek E, Studzinski GP. The Potential of Vitamin D-Regulated Intracellular Signaling Pathways as Targets for Myeloid Leukemia Therapy. J Clin Med 2015; 4:504-34. [PMID: 26239344 PMCID: PMC4470153 DOI: 10.3390/jcm4040504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/06/2015] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
The current standard regimens for the treatment of acute myeloid leukemia (AML) are curative in less than half of patients; therefore, there is a great need for innovative new approaches to this problem. One approach is to target new treatments to the pathways that are instrumental to cell growth and survival with drugs that are less harmful to normal cells than to neoplastic cells. In this review, we focus on the MAPK family of signaling pathways and those that are known to, or potentially can, interact with MAPKs, such as PI3K/AKT/FOXO and JAK/STAT. We exemplify the recent studies in this field with specific relevance to vitamin D and its derivatives, since they have featured prominently in recent scientific literature as having anti-cancer properties. Since microRNAs also are known to be regulated by activated vitamin D, this is also briefly discussed here, as are the implications of the emerging acquisition of transcriptosome data and potentiation of the biological effects of vitamin D by other compounds. While there are ongoing clinical trials of various compounds that affect signaling pathways, more studies are needed to establish the clinical utility of vitamin D in the treatment of cancer.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| | - George P Studzinski
- Department of Pathology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 17101, USA.
| |
Collapse
|
349
|
Revisiting the estrogen receptor pathway and its role in endocrine therapy for postmenopausal women with estrogen receptor-positive metastatic breast cancer. Breast Cancer Res Treat 2015; 150:231-42. [DOI: 10.1007/s10549-015-3316-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/19/2015] [Indexed: 01/27/2023]
|
350
|
Yu ZL, Wang JN, Wu XH, Xie HJ, Han Y, Guan YT, Qin Y, Jiang JM. Tanshinone IIA Prevents Rat Basilar Artery Smooth Muscle Cells Proliferation by Inactivation of PDK1 During the Development of Hypertension. J Cardiovasc Pharmacol Ther 2015; 20:563-71. [PMID: 25736282 DOI: 10.1177/1074248415574743] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 12/28/2014] [Indexed: 12/13/2022]
Abstract
Basilar vascular smooth muscle cells (BASMCs) hyperplasia is a prominent feature of cerebrovascular remodeling and stroke during the development of hypertension. Tanshinone IIA (Tan) has been reported to exhibit a protective effect against the pathological features of hypertension. Previous studies have shown that phosphoinostitide-3 kinase (PI3K)/3'-phosphoinostitide dependent kinase (PDK1)/AKT pathway is involved in the regulation of proliferation of various cell types. Therefore, there may be a crosstalk between Tan antihypertension processes and PI3K/PDK1/AKT proliferative effect in BASMCs. To test this hypothesis, we used a 2-kidney, 2-clip hypertension model to examine the effect of Tan on PI3K/PDK1/AKT pathway by cellular, molecular, and biochemical approaches. Our results revealed that the abundance of PDK1 in plasma was paralleled with an increase in blood pressure and the cross-sectional area of basilar artery in hypertensive rats. Tan decreased blood pressure and hypertension-induced PDK1 phosphorylation but produced no effect on the phosphorylation of PI3K. Moreover, Tan attenuated endothelin 1 induced the activation of PDK1/AKT pathway in rat BASMCs. Tan could inhibit cell cycle transition by regulating the expression of cyclin D1 and p27, in turn, prevent proliferation of BASMCs. Our study provides a novel mechanism by which Tan prevents cerebrovascular cell proliferation during hypertension, and thus Tan may be a potential therapeutic agent for cerebrovascular remodeling and stroke.
Collapse
Affiliation(s)
- Zhi-Liang Yu
- Department of Neurology, Shanghai Seventh People's Hospital, Shanghai, China
| | - Jie-Ning Wang
- Department of Rehabilitation Medicine, Shanghai Seventh People's Hospital, Shanghai, China
| | - Xiao-Hua Wu
- Department of Neurology, Shanghai Seventh People's Hospital, Shanghai, China
| | - Hui-Jun Xie
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ying Han
- Department of Neurology, Shanghai Seventh People's Hospital, Shanghai, China
| | - Yang-Tai Guan
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yong Qin
- Department of Neurology, Shanghai Seventh People's Hospital, Shanghai, China
| | - Jian-Ming Jiang
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|