1
|
Zhang R, Khare P, Banerjee P, Ivan C, Schneider S, Barbaglio F, Clise-Dwyer K, Jensen VB, Thompson E, Mendoza M, Chiorazzi N, Chen SS, Yan XJJ, Jain N, Ghia P, Caligaris-Cappio F, Mendonsa R, Kasimsetty S, Swoboda R, Bayraktar R, Wierda W, Gandhi V, Calin GA, Keating MJ, Bertilaccio MTS. The DLEU2/miR-15a/miR-16-1 cluster shapes the immune microenvironment of chronic lymphocytic leukemia. Blood Cancer J 2024; 14:168. [PMID: 39438453 PMCID: PMC11496494 DOI: 10.1038/s41408-024-01142-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/25/2024] Open
Abstract
The development and progression of chronic lymphocytic leukemia (CLL) depend on genetic abnormalities and on the immunosuppressive microenvironment. We have explored the possibility that genetic drivers might be responsible for the immune cell dysregulation that shapes the protumor microenvironment. We performed a transcriptome analysis of coding and non-coding RNAs (ncRNAs) during leukemia progression in the Rag2-/-γc-/- MEC1-based xenotransplantation model. The DLEU2/miR-16 locus was found downmodulated in monocytes/macrophages of leukemic mice. To validate the role of this cluster in the tumor immune microenvironment, we generated a mouse model that simultaneously mimics the overexpression of hTCL1 and the germline deletion of the minimal deleted region (MDR) encoding the DLEU2/miR-15a/miR-16-1 cluster. This model provides an innovative and faster CLL system where monocyte differentiation and macrophage polarization are exacerbated, and T-cells are dysfunctional. MDR deletion inversely correlates with the levels of predicted target proteins including BCL2 and PD1/PD-L1 on murine CLL cells and immune cells. The inverse correlation of miR-15a/miR-16-1 with target proteins has been confirmed on patient-derived immune cells. Forced expression of miR-16-1 interferes with monocyte differentiation into tumor-associated macrophages, indicating that selected ncRNAs drive the protumor phenotype of non-malignant immune cells.
Collapse
MESH Headings
- MicroRNAs/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Animals
- Mice
- Tumor Microenvironment/immunology
- Humans
- RNA, Long Noncoding/genetics
- Tumor Suppressor Proteins/genetics
- Multigene Family
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyanka Khare
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyanka Banerjee
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Caris Life Sciences, Irving, TX, USA
| | - Sarah Schneider
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Federica Barbaglio
- Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Karen Clise-Dwyer
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vanessa Behrana Jensen
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erika Thompson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marisela Mendoza
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, USA
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, USA
- Northwell Health Cancer Institute, Lake Success, NY, USA
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Xiao-Jie Joy Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paolo Ghia
- B cell neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Federico Caligaris-Cappio
- Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
- AIRC (Associazione Italiana per la Ricerca sul Cancro), 20123, Milan, Italy
| | | | | | | | - Recep Bayraktar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
2
|
Chandra S, Goswami A, Mandal P. Molecular Heterogeneity of Cervical Cancer Among Different Ethnic/Racial Populations. J Racial Ethn Health Disparities 2022; 9:2441-2450. [PMID: 34741276 DOI: 10.1007/s40615-021-01180-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/29/2022]
Abstract
The study aimed to find differential gene mutation profile and gene expression status among different ethnic/racial human populations relevant for cervical cancer pathogenesis. The study was based on freely available datasets of The Cancer Genome Atlas (TCGA) of cervical cancer samples in Genomic Data Commons (GDC) data portal. We identified that choline metabolism in cancer and Ras signaling pathways were significantly associated with the Hispanic and Latino group of cervical cancer patients. In these pathways, mutations in the PIK3CA gene, especially E545K, were significantly associated with the Hispanic and LATINO group. We found that AFF3 gene mutation was associated with downregulation of its expression only among the White racial category of cervical cancer cases. Additionally, hypomethylation of the CpG position in the S shore region of the PM20D1 gene was associated with overexpression among the Asian category of cervical cancer cases. Heterogeneity of the molecular profile of AFF3 and PM20D1 gene among racial groups reflects the potential of differential targeted therapy of cervical cancer.
Collapse
Affiliation(s)
- Sanchita Chandra
- Biomedical Genetics Laboratory, Department of Zoology, The University of Burdwan, Burdwan, 713104, West Bengal, India
| | - Anindita Goswami
- Biomedical Genetics Laboratory, Department of Zoology, The University of Burdwan, Burdwan, 713104, West Bengal, India
| | - Paramita Mandal
- Biomedical Genetics Laboratory, Department of Zoology, The University of Burdwan, Burdwan, 713104, West Bengal, India.
| |
Collapse
|
3
|
Liao QQ, Dong QQ, Zhang H, Shu HP, Tu YC, Yao LJ. Contributions of SGK3 to transporter-related diseases. Front Cell Dev Biol 2022; 10:1007924. [PMID: 36531961 PMCID: PMC9753149 DOI: 10.3389/fcell.2022.1007924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/09/2022] [Indexed: 02/09/2024] Open
Abstract
Serum- and glucocorticoid-induced kinase 3 (SGK3), which is ubiquitously expressed in mammals, is regulated by estrogens and androgens. SGK3 is activated by insulin and growth factors through signaling pathways involving phosphatidylinositol-3-kinase (PI3K), 3-phosphoinositide-dependent kinase-1 (PDK-1), and mammalian target of rapamycin complex 2 (mTORC2). Activated SGK3 can activate ion channels (TRPV5/6, SOC, Kv1.3, Kv1.5, Kv7.1, BKCa, Kir2.1, Kir2.2, ENaC, Nav1.5, ClC-2, and ClC Ka), carriers and receptors (Npt2a, Npt2b, NHE3, GluR1, GluR6, SN1, EAAT1, EAAT2, EAAT4, EAAT5, SGLT1, SLC1A5, SLC6A19, SLC6A8, and NaDC1), and Na+/K+-ATPase, promoting the transportation of calcium, phosphorus, sodium, glucose, and neutral amino acids in the kidney and intestine, the absorption of potassium and neutral amino acids in the renal tubules, the transportation of glutamate and glutamine in the nervous system, and the transportation of creatine. SGK3-sensitive transporters contribute to a variety of physiological and pathophysiological processes, such as maintaining calcium and phosphorus homeostasis, hydro-salinity balance and acid-base balance, cell proliferation, muscle action potential, cardiac and neural electrophysiological disturbances, bone density, intestinal nutrition absorption, immune function, and multiple substance metabolism. These processes are related to kidney stones, hypophosphorous rickets, multiple syndromes, arrhythmia, hypertension, heart failure, epilepsy, Alzheimer's disease, amyotrophic lateral sclerosis, glaucoma, ataxia idiopathic deafness, and other diseases.
Collapse
Affiliation(s)
- Qian-Qian Liao
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qing-Qing Dong
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Zhang
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hua-Pan Shu
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Chi Tu
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Jun Yao
- Department of Nephrology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Myricetin-induced apoptosis in triple-negative breast cancer cells through inhibition of the PI3K/Akt/mTOR pathway. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:248. [PMID: 36209343 DOI: 10.1007/s12032-022-01856-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/17/2022] [Indexed: 10/10/2022]
Abstract
Breast cancer is still a severe origin of malignant demise in females, and its prevalence is rising worldwide. Triple-negative breast cancer (TNBC) is a diversified aggressive breast tumor distinguished by inadequate prognosis, early recurrence, high invasion, and extremely metastasized disease. Chemotherapy is being used to treat it; however, it has low efficacy. On the other hand, with the growing number of corroborations on subtypes of TNBC and molecular biology of tumors, significant advancement in TNBC targeted treatment has been made. Myricetin (MYR), a polyhydroxyflavonol compound widely found in nature, has been shown to possess anticancer effects in various cancers. Though, the mechanisms and impacts of MYR on metastasis of TNBC remain unclear. Early and late apoptotic cell death and cell proliferation inhibition were observed in MYR-treated TNBC cells. MYR modulated cell cycle, pro-angiogenic, and invasion effects via the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/Protein kinase B (PKB/also known as AKT) signaling pathways. Moreover, it regulates the expression of MAPK, PI3K/AKT/mTOR, IκB/NF-κB, Hippo, STAT3, GSK-3β, Nrf2/HO-1, TLR, eNOS / NO, ACE, and AChE. Here, we review the anticancer effects of MYR for TNBC and target the PI3K/AKT/mTOR pathway as a therapeutic target for the fruitful treatment of TNBC to summarize MYR's therapeutic potential.
Collapse
|
5
|
Zughaibi TA, Suhail M, Tarique M, Tabrez S. Targeting PI3K/Akt/mTOR Pathway by Different Flavonoids: A Cancer Chemopreventive Approach. Int J Mol Sci 2021; 22:12455. [PMID: 34830339 PMCID: PMC8621356 DOI: 10.3390/ijms222212455] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/02/2021] [Accepted: 11/13/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is, globally, one of the main causes of death. Even though various therapies are available, they are still painful because of their adverse side effects. Available treatments frequently fail due to unpromising responses, resistance to classical anticancer drugs, radiation therapy, chemotherapy, and low accessibility to tumor tissues. Developing novel strategies to minimize adverse side effects, improve chemotherapy sensitivity, and control cancer progression is needed. Many studies have suggested small dietary molecules as complementary treatments for cancer patients. Different components of herbal/edible plants, known as flavonoids, have recently garnered attention due to their broad biological properties (e.g., antioxidant, antiviral, antimicrobial, anti-inflammatory, anti-mutagenic, anticancer, hepatoprotective, and cardioprotective). These flavonoids have shown anticancer activity by affecting different signaling cascades. This article summarizes the key progress made in this area and discusses the role of flavonoids by specifically inhibiting the PI3K/Akt/mTOR pathway in various cancers.
Collapse
Affiliation(s)
- Torki A. Zughaibi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Tarique
- Department of Child Health, School of Medicine, University of Missouri, Columbia, MO 65201, USA;
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
6
|
Ohashi Y. Activation Mechanisms of the VPS34 Complexes. Cells 2021; 10:cells10113124. [PMID: 34831348 PMCID: PMC8624279 DOI: 10.3390/cells10113124] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 01/18/2023] Open
Abstract
Phosphatidylinositol-3-phosphate (PtdIns(3)P) is essential for cell survival, and its intracellular synthesis is spatially and temporally regulated. It has major roles in two distinctive cellular pathways, namely, the autophagy and endocytic pathways. PtdIns(3)P is synthesized from phosphatidylinositol (PtdIns) by PIK3C3C/VPS34 in mammals or Vps34 in yeast. Pathway-specific VPS34/Vps34 activity is the consequence of the enzyme being incorporated into two mutually exclusive complexes: complex I for autophagy, composed of VPS34/Vps34-Vps15/Vps15-Beclin 1/Vps30-ATG14L/Atg14 (mammals/yeast), and complex II for endocytic pathways, in which ATG14L/Atg14 is replaced with UVRAG/Vps38 (mammals/yeast). Because of its involvement in autophagy, defects in which are closely associated with human diseases such as cancer and neurodegenerative diseases, developing highly selective drugs that target specific VPS34/Vps34 complexes is an essential goal in the autophagy field. Recent studies on the activation mechanisms of VPS34/Vps34 complexes have revealed that a variety of factors, including conformational changes, lipid physicochemical parameters, upstream regulators, and downstream effectors, greatly influence the activity of these complexes. This review summarizes and highlights each of these influences as well as clarifying key questions remaining in the field and outlining future perspectives.
Collapse
Affiliation(s)
- Yohei Ohashi
- MRC Laboratory of Molecular Biology, Protein and Nucleic Acid Chemistry Division, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
7
|
Jiang F, Liu H, Peng F, Liu Z, Ding K, Song J, Li L, Chen J, Shao Q, Yan S, De Veirman K, Vanderkerken K, Fu R. Complement C3a activates osteoclasts by regulating the PI3K/PDK1/SGK3 pathway in patients with multiple myeloma. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0430. [PMID: 33960177 PMCID: PMC8330530 DOI: 10.20892/j.issn.2095-3941.2020.0430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 11/27/2020] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Myeloma bone disease (MBD) is the most common complication of multiple myeloma (MM). Our previous study showed that the serum levels of C3/C4 in MM patients were significantly positively correlated with the severity of bone disease. However, the mechanism of C3a/C4a in osteoclasts MM patients remains unclear. METHODS The formation and function of osteoclasts were analyzed after adding C3a/C4a in vitro. RNA-seq analysis was used to screen the potential pathways affecting osteoclasts, and the results were verified by Western blot, qRT-PCR, and pathway inhibitors. RESULTS The osteoclast area per view induced by 1 μg/mL (mean ± SD: 50.828 ± 12.984%) and 10 μg/mL (53.663 ± 12.685%) of C3a was significantly increased compared to the control group (0 μg/mL) (34.635 ± 8.916%) (P < 0.001 and P < 0.001, respectively). The relative mRNA expressions of genes, OSCAR/TRAP/RANKL/cathepsin K, induced by 1 μg/mL (median: 5.041, 3.726, 1.638, and 4.752, respectively) and 10 μg/mL (median: 5.140, 3.702, 2.250, and 5.172, respectively) of C3a was significantly increased compared to the control group (median: 3.137, 2.004, 0.573, and 2.257, respectively) (1 μg/mL P = 0.001, P = 0.003, P < 0.001, and P = 0.008, respectively; 10 μg/mL: P < 0.001, P = 0.019, P < 0.001, and P = 0.002, respectively). The absorption areas of the osteoclast resorption pits per view induced by 1 μg/mL (mean ± SD: 51.464 ± 11.983%) and 10 μg/mL (50.219 ± 12.067%) of C3a was also significantly increased (33.845 ± 8.331%) (P < 0.001 and P < 0.001, respectively) compared to the control. There was no difference between the C4a and control groups. RNA-seq analysis showed that C3a promoted the proliferation of osteoclasts using the phosphoinositide 3-kinase (PI3K) signaling pathway. The relative expressions of PIK3CA/phosphoinositide dependent kinase-1 (PDK1)/serum and glucocorticoid inducible protein kinases (SGK3) genes and PI3K/PDK1/p-SGK3 protein in the C3a group were significantly higher than in the control group. The activation role of C3a in osteoclasts of MM patients was reduced by the SGK inhibitor (EMD638683). CONCLUSIONS C3a activated osteoclasts by regulating the PI3K/PDK1/SGK3 pathways in MM patients, which was reduced using a SGK inhibitor. Overall, our results identified potential therapeutic targets and strategies for MBD patients.
Collapse
Affiliation(s)
- Fengjuan Jiang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fengping Peng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jia Song
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jin Chen
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qing Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Siyang Yan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Kim De Veirman
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
8
|
Guerriero I, Monaco G, Coppola V, Orlacchio A. Serum and Glucocorticoid-Inducible Kinase 1 (SGK1) in NSCLC Therapy. Pharmaceuticals (Basel) 2020; 13:ph13110413. [PMID: 33266470 PMCID: PMC7700219 DOI: 10.3390/ph13110413] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the most prevalent and one of the deadliest cancers worldwide. Despite recent success, there is still an urgent need for new therapeutic strategies. It is also becoming increasingly evident that combinatorial approaches are more effective than single modality treatments. This review proposes that the serum and glucocorticoid-inducible kinase 1 (SGK1) may represent an attractive target for therapy of NSCLC. Although ubiquitously expressed, SGK1 deletion in mice causes only mild defects of ion physiology. The frequent overexpression of SGK1 in tumors is likely stress-induced and provides a therapeutic window to spare normal tissues. SGK1 appears to promote oncogenic signaling aimed at preserving the survival and fitness of cancer cells. Most importantly, recent investigations have revealed the ability of SGK1 to skew immune-cell differentiation toward pro-tumorigenic phenotypes. Future studies are needed to fully evaluate the potential of SGK1 as a therapeutic target in combinatorial treatments of NSCLC. However, based on what is currently known, SGK1 inactivation can result in anti-oncogenic effects both on tumor cells and on the immune microenvironment. A first generation of small molecules to inactivate SGK1 has already been already produced.
Collapse
Affiliation(s)
- Ilaria Guerriero
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Gianni Monaco
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| | - Arturo Orlacchio
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| |
Collapse
|
9
|
Ros S, Wright AJ, D'Santos P, Hu DE, Hesketh RL, Lubling Y, Georgopoulou D, Lerda G, Couturier DL, Razavi P, Pelossof R, Batra AS, Mannion E, Lewis DY, Martin A, Baird RD, Oliveira M, de Boo LW, Linn SC, Scaltriti M, Rueda OM, Bruna A, Caldas C, Brindle KM. Metabolic Imaging Detects Resistance to PI3Kα Inhibition Mediated by Persistent FOXM1 Expression in ER + Breast Cancer. Cancer Cell 2020; 38:516-533.e9. [PMID: 32976773 PMCID: PMC7562820 DOI: 10.1016/j.ccell.2020.08.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 06/26/2020] [Accepted: 08/25/2020] [Indexed: 12/25/2022]
Abstract
PIK3CA, encoding the PI3Kα isoform, is the most frequently mutated oncogene in estrogen receptor (ER)-positive breast cancer. Isoform-selective PI3K inhibitors are used clinically but intrinsic and acquired resistance limits their utility. Improved selection of patients that will benefit from these drugs requires predictive biomarkers. We show here that persistent FOXM1 expression following drug treatment is a biomarker of resistance to PI3Kα inhibition in ER+ breast cancer. FOXM1 drives expression of lactate dehydrogenase (LDH) but not hexokinase 2 (HK-II). The downstream metabolic changes can therefore be detected using MRI of LDH-catalyzed hyperpolarized 13C label exchange between pyruvate and lactate but not by positron emission tomography measurements of HK-II-mediated trapping of the glucose analog 2-deoxy-2-[18F]fluorodeoxyglucose. Rapid assessment of treatment response in breast cancer using this imaging method could help identify patients that benefit from PI3Kα inhibition and design drug combinations to counteract the emergence of resistance.
Collapse
Affiliation(s)
- Susana Ros
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK.
| | - Alan J Wright
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Paula D'Santos
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - De-En Hu
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Richard L Hesketh
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Yaniv Lubling
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Dimitra Georgopoulou
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Giulia Lerda
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Dominique-Laurent Couturier
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Pedram Razavi
- Human Oncology and Pathogenesis Program, X and Department of Pathology, Y and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rapahel Pelossof
- Human Oncology and Pathogenesis Program, X and Department of Pathology, Y and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ankita S Batra
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Elizabeth Mannion
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - David Y Lewis
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Alistair Martin
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Richard D Baird
- Breast Cancer Research Programme, Cancer Research UK Cambridge Centre, Cambridge, UK
| | - Mafalda Oliveira
- Medical Oncology, Vall d'Hebron Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Leonora W de Boo
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sabine C Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program, X and Department of Pathology, Y and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Oscar M Rueda
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Alejandra Bruna
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK
| | - Kevin M Brindle
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Cancer Research UK Cambridge Cancer Centre, Cambridge, UK; Department of Biochemistry, University of Cambridge, Cambridge UK.
| |
Collapse
|
10
|
Zhou Y, An H, Wu G. MicroRNA-6071 Suppresses Glioblastoma Progression Through the Inhibition of PI3K/AKT/mTOR Pathway by Binding to ULBP2. Onco Targets Ther 2020; 13:9429-9441. [PMID: 33061429 PMCID: PMC7520159 DOI: 10.2147/ott.s265791] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Objective The purpose of this study was to explore the effect of microRNA-6071 (miR-6071) on glioblastoma (GBM) and its potential mechanisms. Methods In this study, the expressions of miR-6071 and UL16 binding protein 2 (ULBP2) were measured by qRT-RCR in GBM tissues and cells. The prognostic values of miR-6071 and ULBP2 were evaluated by Kaplan–Meier methods using the data obtained from The Cancer Genome Atlas (TCGA) database. The cell clones, proliferation, apoptosis, migration and invasion in GBM cells were detected by colony formation assay, EdU assay, flow cytometry, wound-healing assay and transwell assay. The targeting relationship between miR-6071 and ULBP2 was predicted by Targetscan 7.2 and further verified by dual-luciferase reporter gene assay. Moreover, the expressions of Bax, caspase-3, Bcl-2, matrix metalloproteinases 2 (MMP-2), MMP-9, phosphatidylinositol 3′-kinase (PI3K), p-PI3K, protein kinase B (AKT), p-AKT, mammalian target of rapamycin (mTOR) and p-mTOR were measured by Western blot. Results miR-6071 was lowly expressed and ULBP2 was highly expressed in GBM tissues and cells. miR-6071 significantly repressed the proliferation, migration and invasion, and promoted apoptosis in GBM cells. Moreover, miR-6071 also inhibited the activation of PI3K/AKT/mTOR pathway in GBM cells. Additionally, miR-6071 has been shown to negatively regulate ULBP2 expression. We also confirmed that ULBP2 could reverse the effects of miR-6071 on GBM cells through regulating PI3K/AKT/mTOR pathway. Conclusion Our study demonstrated that miR-6071 could suppress cell proliferation, migration and invasion, as well as promote apoptosis through the inhibition of PI3K/Akt/mTOR pathway by binding to ULBP2 in GBM.
Collapse
Affiliation(s)
- Yunyan Zhou
- Second Department of Neurology, Rongcheng People's Hospital, Shandong Province, Rongcheng, Shandong 264300, People's Republic of China
| | - Hongwei An
- Surgery of Lingcheng, Hospital of Traditional Chinese Medicine in Dezhou City, Dezhou, Shandong 253500, People's Republic of China
| | - Gang Wu
- Department of Neurology, Yan'an Hospital of Kunming, Kunming, Yunnan 650051, People's Republic of China
| |
Collapse
|
11
|
Yu Q, Luo J, Zhang J, Chen Y, Chen K, Lin J, Sun S, Lin X. Oxymatrine inhibits the development of non-small cell lung cancer through miR-367-3p upregulation and target gene SGK3 downregulation. Am J Transl Res 2020; 12:5538-5550. [PMID: 33042436 PMCID: PMC7540135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
Oxymatrine (OM), an important active ingredient extracted from sophora flavescens, has attracted more attention for its anti-tumor effect in recent years, with pronounced effects on the development of multiple tumors, acting as a potential effective low toxic drug in clinical tumor treatment. In this study, CCK-8 and transwell experiments were applied to detect cell proliferation and migration. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were used to test the expression of miR-367-3p and serum and glucocorticoid regulated kinase 3 (SGK3). The function of oxymatrine in non-small cell lung cancer (NSCLC) progression was also confirmed in vivo. Then, CCK-8 and transwell assays revealed that oxymatrine could repress NSCLC cell migration and proliferation. qRT-PCR showed the striking promotion roles of oxymatrine in cancer suppressor gene miR-367-3p expression. The results of further dual luciferase reporter gene experiment demonstrated that SGK3 was a target gene of miR-367-3p and under the regulation of oxymatrine. The rescue experiments indicated that OM functioned via miR-367-3p, while miR-367-3p exerted its function by action on SGK3. Finally, in vivo studies showed that OM could also inhibit tumor growth. As a result, this study found that OM inhibited the development of NSCLC through reducing the expression of a downstream target gene SGK3 by promoting miR-367-3p expression.
Collapse
Affiliation(s)
- Qinghua Yu
- Department of Radiology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| | - Jiewei Luo
- Department of Traditional Chinese Medicine, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| | - Jiguang Zhang
- Department of Thoracic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| | - Yangming Chen
- Department of Thoracic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| | - Kai Chen
- Department of Thoracic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| | - Jianbin Lin
- Department of Thoracic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| | - Shihui Sun
- Department of Thoracic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| | - Xing Lin
- Department of Thoracic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical UniversityFuzhou, Fujian, China
| |
Collapse
|
12
|
Serum- and glucocorticoid- inducible kinase 2, SGK2, is a novel autophagy regulator and modulates platinum drugs response in cancer cells. Oncogene 2020; 39:6370-6386. [PMID: 32848212 PMCID: PMC7529585 DOI: 10.1038/s41388-020-01433-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 08/07/2020] [Accepted: 08/13/2020] [Indexed: 12/16/2022]
Abstract
For many tumor types chemotherapy still represents the therapy of choice and many standard treatments are based on the use of platinum (PT) drugs. However, de novo or acquired resistance to platinum is frequent and leads to disease progression. In Epithelial Ovarian Cancer (EOC) patients, PT-resistant recurrences are very common and improving the response to treatment still represents an unmet clinical need. To identify new modulators of PT-sensitivity, we performed a loss-of-function screening targeting 680 genes potentially involved in the response of EOC cells to platinum. We found that SGK2 (Serum-and Glucocorticoid-inducible kinase 2) plays a key role in PT-response. We show here that EOC cells relay on the induction of autophagy to escape PT-induced death and that SGK2 inhibition increases PT sensitivity inducing a block in the autophagy cascade due to the impairment of lysosomal acidification. Mechanistically we demonstrate that SGK2 controls autophagy in a kinase-dependent manner by binding and inhibiting the V-ATPase proton pump. Accordingly, SGK2 phosphorylates the subunit V1H (ATP6V1H) of V-ATPase and silencing or chemical inhibition of SGK2, affects the normal autophagic flux and sensitizes EOC cells to platinum. Hence, we identified a new pathway that links autophagy to the survival of cancer cells under platinum treatment in which the druggable kinase SGK2 plays a central role. Our data suggest that blocking autophagy via SGK2 inhibition could represent a novel therapeutic strategy to improve patients' response to platinum.
Collapse
|
13
|
Zhu R, Yang G, Cao Z, Shen K, Zheng L, Xiao J, You L, Zhang T. The prospect of serum and glucocorticoid-inducible kinase 1 (SGK1) in cancer therapy: a rising star. Ther Adv Med Oncol 2020; 12:1758835920940946. [PMID: 32728395 PMCID: PMC7364809 DOI: 10.1177/1758835920940946] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022] Open
Abstract
Serum and glucocorticoid-inducible kinase 1 (SGK1) is an AGC kinase that has been reported to be involved in a variety of physiological and pathological processes. Recent evidence has accumulated that SGK1 acts as an essential Akt-independent mediator of phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling pathway in cancer. SGK1 is overexpressed in several tumors, including prostate cancer, colorectal carcinoma, glioblastoma, breast cancer, and endometrial cancer. The functions of SGK1 include regulating tumor growth, survival, metastasis, autophagy, immunoregulation, calcium (Ca2+) signaling, cancer stem cells, cell cycle, and therapeutic resistance. In this review, we introduce the pleiotropic role of SGK1 in the development and progression of tumors, summarize its downstream targets, and integrate the knowledge provided by preclinical studies that the prospect of SGK1 inhibition as a potential therapeutic approach.
Collapse
Affiliation(s)
- Ruizhe Zhu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Shen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing 100730, China
| |
Collapse
|
14
|
Abstract
Oocyte maturation is a process that occurs in the ovaries, where an immature oocyte resumes meiosis to attain competence for normal fertilization after ovulation/spawning. In starfish, the hormone 1-methyladenine binds to an unidentified receptor on the plasma membrane of oocytes, inducing a conformational change in the heterotrimeric GTP-binding protein α-subunit (Gα), so that the α-subunit binds GTP in exchange of GDP on the plasma membrane. The GTP-binding protein βγ-subunit (Gβγ) is released from Gα, and the released Gβγ activates phosphatidylinositol-3 kinase (PI3K), followed by the target of rapamycin kinase complex2 (TORC2) and 3-phosphoinositide-dependent protein kinase 1 (PDK1)-dependent phosphorylation of serum- and glucocorticoid-regulated kinase (SGK) of ovarian oocytes. Thereafter, SGK activates Na+/H+ exchanger (NHE) to increase the intracellular pH (pHi) from ~6.7 to ~6.9. Moreover, SGK phosphorylates Cdc25 and Myt1, thereby inducing the de-phosphorylation and activation of cyclin B–Cdk1, causing germinal vesicle breakdown (GVBD). Both pHi increase and GVBD are required for spindle assembly at metaphase I, followed by MI arrest at pHi 6.9 until spawning. Due to MI arrest or SGK-dependent pHi control, spawned oocytes can be fertilized normally
Collapse
Affiliation(s)
- Kazuyoshi Chiba
- Department of Biological Sciences, Ochanomizu University, Tokyo 112-8610, Japan
| |
Collapse
|
15
|
Hiraoka D, Hosoda E, Chiba K, Kishimoto T. SGK phosphorylates Cdc25 and Myt1 to trigger cyclin B-Cdk1 activation at the meiotic G2/M transition. J Cell Biol 2019; 218:3597-3611. [PMID: 31537708 PMCID: PMC6829662 DOI: 10.1083/jcb.201812122] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/03/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
The kinase cyclin B-Cdk1 complex is a master regulator of M-phase in both mitosis and meiosis. At the G2/M transition, cyclin B-Cdk1 activation is initiated by a trigger that reverses the balance of activities between Cdc25 and Wee1/Myt1 and is further accelerated by autoregulatory loops. In somatic cell mitosis, this trigger was recently proposed to be the cyclin A-Cdk1/Plk1 axis. However, in the oocyte meiotic G2/M transition, in which hormonal stimuli induce cyclin B-Cdk1 activation, cyclin A-Cdk1 is nonessential and hence the trigger remains elusive. Here, we show that SGK directly phosphorylates Cdc25 and Myt1 to trigger cyclin B-Cdk1 activation in starfish oocytes. Upon hormonal stimulation of the meiotic G2/M transition, SGK is activated by cooperation between the Gβγ-PI3K pathway and an unidentified pathway downstream of Gβγ, called the atypical Gβγ pathway. These findings identify the trigger in oocyte meiosis and provide insights into the role and activation of SGK.
Collapse
Affiliation(s)
- Daisaku Hiraoka
- Science and Education Center, Ochanomizu University, Tokyo, Japan
| | - Enako Hosoda
- Department of Biological Sciences, Ochanomizu University, Tokyo, Japan
| | - Kazuyoshi Chiba
- Department of Biological Sciences, Ochanomizu University, Tokyo, Japan
| | - Takeo Kishimoto
- Science and Education Center, Ochanomizu University, Tokyo, Japan
| |
Collapse
|
16
|
Oza M, Becker W, Gummadidala PM, Dias T, Omebeyinje MH, Chen L, Mitra C, Jesmin R, Chakraborty P, Sajish M, Hofseth LJ, Banerjee K, Wang Q, Moeller PDR, Nagarkatti M, Nagarkatti P, Chanda A. Acute and short-term administrations of delta-9-tetrahydrocannabinol modulate major gut metabolomic regulatory pathways in C57BL/6 mice. Sci Rep 2019; 9:10520. [PMID: 31324830 PMCID: PMC6642200 DOI: 10.1038/s41598-019-46478-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 06/19/2019] [Indexed: 01/07/2023] Open
Abstract
Delta-9-tetrahydrocannabinol (THC) is the primary psychoactive compound in Cannabis, which is studied extensively for its medicinal value. A central gap in the science is the underlying mechanisms surrounding THC's therapeutic effects and the role of gut metabolite profiles. Using a mass-spectrometry based metabolomics, we show here that intraperitoneal injection of THC in C57BL/6 mice modulates metabolic profiles that have previously been identified as integral to health. Specifically, we investigated the effects of acute (single THC injection denoted here as '1X') and short -term (five THC injections on alternate days denoted as '5X') THC administration on fecal and intestinal tissue metabolite profiles. Results are consistent with the hypothesis that THC administration alters host metabolism by targeting two prominent lipid metabolism pathways: glycerophospholipid metabolism and fatty acid biosynthesis.
Collapse
Affiliation(s)
- Megha Oza
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - William Becker
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Phani M Gummadidala
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Travis Dias
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Mayomi H Omebeyinje
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Li Chen
- Creative Proteomics Inc., Shirley, New York, USA
| | - Chandrani Mitra
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Rubaiya Jesmin
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | | | - Mathew Sajish
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Lorne J Hofseth
- Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | | | - Qian Wang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Peter D R Moeller
- National Ocean Service, Hollings Marine Laboratory, Charleston, SC, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Anindya Chanda
- Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
17
|
Wang L, Chen R, Zhang Y. miR-296-3p targets APEX1 to suppress cell migration and invasion of non-small-cell lung cancer. Oncol Lett 2019; 18:2612-2618. [PMID: 31402954 DOI: 10.3892/ol.2019.10572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 05/03/2019] [Indexed: 02/07/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the most common cause of cancer-associated mortality worldwide. MicroRNAs (miRs) are a class of small non-coding RNAs that are commonly dysregulated in human cancer. The aim of the current study was to evaluate the effect of miR-296-3p on the cell migration and invasion of NSCLC. Pairs of tumor tissues and para-cancerous tissues (n=50) were collected from patients with NSCLC, and the expression of miR-296-3p was analyzed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Additionally, tumor cell viability, migration and invasion were examined in vitro using Cell Counting Kit-8, wound healing and Matrigel assays, respectively. Furthermore, potential targets of miR-296-3p were screened for using TargetScan and validated using a dual-luciferase reporter assay. The expression levels of phosphoinositide-3-kinase (PI3K), AKT serine/threonine kinase (AKT), mammalian target of rapamycin (mTOR), matrix metallopeptidase 2 (MMP2) and SRY-box 4 (SOX4) were detected by RT-qPCR and western blot analysis. The data indicated that miR-296-3p was downregulated in tumor tissues compared with adjacent normal tissues. Overexpression of miR-296-3p inhibited NSCLC cell viability, migration and invasion in vitro. Furthermore, apurinic/apyrimidinic endodeoxyribonuclease 1 (APEX1) was identified as a direct target of miR-296-3p. APEX1 expression was upregulated in tumor tissues compared with para-cancerous tissues, and the mRNA and protein expression levels of APEX1 were decreased following transfection of NSCLC cells with miR-296-3p mimics compared with control cells. Additional investigations revealed that miR-296-3p was involved in regulating the PI3K/AKT/mTOR signaling pathway, and miR-296-3p mimics decreased the mRNA and protein expression levels of MMP2 and SOX4. In summary, the findings demonstrated that miR-296-3p may function as a tumor suppressor, and inhibits the migration and invasion of NSCLC cells by targeting APEX1. miR-296-3p is therefore a potential therapeutic molecular modulator of NSCLC.
Collapse
Affiliation(s)
- Lifeng Wang
- Department of Respiration, Xi'an High-tech Hospital, Xi'an, Shaanxi 710075, P.R. China
| | - Ruilin Chen
- Department of Respiration, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yongqing Zhang
- Department of Respiration, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
18
|
Emmanouilidi A, Fyffe CA, Ferro R, Edling CE, Capone E, Sestito S, Rapposelli S, Lattanzio R, Iacobelli S, Sala G, Maffucci T, Falasca M. Preclinical validation of 3-phosphoinositide-dependent protein kinase 1 inhibition in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:191. [PMID: 31088502 PMCID: PMC6518649 DOI: 10.1186/s13046-019-1191-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 04/25/2019] [Indexed: 11/02/2022]
Abstract
BACKGROUND The very aggressive nature and low survival rate of pancreatic ductal adenocarcinoma (PDAC) dictates the necessity to find novel efficacious therapies. Recent evidence suggests that phosphoinositide 3-kinase (PI3K) and 3-phosphoinositide-dependent protein kinase 1 (PDK1) are key effectors of oncogenic KRAS in PDAC. Herein, we report the role and mechanism of action of PDK1, a protein kinase of the AGC family, in PDAC. METHODS PDAC cell lines were treated with selective PDK1 inhibitors or transfected with specific PDK1-targeting siRNAs. In vitro and in vivo assays were performed to investigate the functional role of PDK1 in PDAC. Specifically, anchorage-dependent and anchorage-independent growth was assessed in PDAC cells upon inhibition or downregulation of PDK1. Detailed investigation of the effect of PDK1 inhibition/downregulation on specific signalling pathways was also performed by Western blotting analysis. A xenograft tumour mouse model was used to determine the effect of pharmacological inhibition of PDK1 on PDAC cells growth in vivo. RESULTS Treatment with specific inhibitors of PDK1 impaired anchorage-dependent and anchorage-independent growth of pancreatic cancer cell lines, as well as pancreatic tumour growth in a xenograft model. Mechanistically, inhibition or downregulation of PDK1 resulted in reduced activation of the serum/glucocorticoid regulated kinase family member 3 and subsequent reduced phosphorylation of its target N-Myc downstream regulated 1. Additionally, we found that combination of sub-optimal concentrations of inhibitors selective for PDK1 and the class IB PI3K isoform p110γ inhibits pancreatic cancer cell growth and colonies formation more potently than each single treatment. CONCLUSIONS Our data indicate that PDK1 is a suitable target for therapeutic intervention in PDAC and support the clinical development of PDK1 inhibitors for PDAC.
Collapse
Affiliation(s)
- Aikaterini Emmanouilidi
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, 6102, Australia
| | - Chanse A Fyffe
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Riccardo Ferro
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Charlotte E Edling
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Emily Capone
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy
| | - Simona Sestito
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126, Pisa, Italy
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126, Pisa, Italy
| | - Rossano Lattanzio
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy
| | - Stefano Iacobelli
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy.,MediaPharma Srl, Via della Colonnetta, 50/A, 66100, Chieti, Italy
| | - Gianluca Sala
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University G. d'Annunzio di Chieti-Pescara, Centro Studi sull Invecchiamento, CeSI-MeT, 66100, Chieti, Italy
| | - Tania Maffucci
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, 6102, Australia. .,Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Cell Biology and Cutaneous Research, E1 2AT, London, UK.
| |
Collapse
|
19
|
Cao H, Xu Z, Wang J, Cigliano A, Pilo MG, Ribback S, Zhang S, Qiao Y, Che L, Pascale RM, Calvisi DF, Chen X. Functional role of SGK3 in PI3K/Pten driven liver tumor development. BMC Cancer 2019; 19:343. [PMID: 30975125 PMCID: PMC6458829 DOI: 10.1186/s12885-019-5551-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a leading cause of cancer related deaths worldwide. The PI3K cascade is one of the major signaling pathways underlying HCC development and progression. Activating mutations of PI3K catalytic subunit alpha (PIK3CA) and/or loss of Pten often occur in human HCCs. Serum and glucocorticoid kinase 3 (SGK3) belongs to the SGK family of AGK kinases and functions in parallel to AKT downstream of PI3K. Previous studies have shown that SGK3 may be the major kinase responsible for the oncogenic potential of PIK3CA helical domain mutants, such as PIK3CA(E545K), but not kinase domain mutants, such as PIK3CA(H1047R). METHODS We investigated the functional contribution of SGK3 in mediating activated PIK3CA mutant or loss of Pten induced HCC development using Sgk3 knockout mice. RESULTS We found that ablation of Sgk3 does not affect PIK3CA(H1047R) or PIK3CA(E545K) induced lipogenesis in the liver. Using PIK3CA(H1047R)/c-Met, PIK3CA(E545K)/c-Met, and sgPten/c-Met murine HCC models, we also demonstrated that deletion of Sgk3 moderately delays PIK3CA(E545K)/c-Met driven HCC, while not affecting PIK3CA(H1047R)/c-Met or sgPten/c-Met HCC formation in mice. Similarly, in human HCC cell lines, silencing of SGK3 reduced PIK3CA(E545K) -but not PIK3CA(H1047R)- induced accelerated tumor cell proliferation. CONCLUSION Altogether, our data suggest that SGK3 plays a role in transducing helical domain mutant PIK3CA signaling during liver tumor development.
Collapse
Affiliation(s)
- Hui Cao
- Department of Oncology, Guizhou Provincial People’s Hospital, Medical College of Guizhou University, Guiyang, People’s Republic of China
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, UCSF, 513 Parnassus Ave, San Francisco, CA 94143 USA
| | - Zhong Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, UCSF, 513 Parnassus Ave, San Francisco, CA 94143 USA
- Department of Gastroenterology, Guizhou Provincial People’s Hospital, Medical College of Guizhou University, Guiyang, People’s Republic of China
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, UCSF, 513 Parnassus Ave, San Francisco, CA 94143 USA
- Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Antonio Cigliano
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, Italy
| | - Maria G. Pilo
- Department of Clinical and Experimental Medicine, University of Sassari, via P. Manzella 4, 07100 Sassari, Italy
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Shu Zhang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, UCSF, 513 Parnassus Ave, San Francisco, CA 94143 USA
| | - Yu Qiao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, UCSF, 513 Parnassus Ave, San Francisco, CA 94143 USA
- Department of Oncology, Beijing Hospital, Beijing, People’s Republic of China
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, UCSF, 513 Parnassus Ave, San Francisco, CA 94143 USA
| | - Rosa M. Pascale
- Department of Clinical and Experimental Medicine, University of Sassari, via P. Manzella 4, 07100 Sassari, Italy
| | - Diego F. Calvisi
- Department of Clinical and Experimental Medicine, University of Sassari, via P. Manzella 4, 07100 Sassari, Italy
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, UCSF, 513 Parnassus Ave, San Francisco, CA 94143 USA
| |
Collapse
|
20
|
Wang H, Huang F, Zhang Z, Wang P, Luo Y, Li H, Li N, Wang J, Zhou J, Wang Y, Li S. Feedback Activation of SGK3 and AKT Contributes to Rapamycin Resistance by Reactivating mTORC1/4EBP1 Axis via TSC2 in Breast Cancer. Int J Biol Sci 2019; 15:929-941. [PMID: 31182914 PMCID: PMC6535796 DOI: 10.7150/ijbs.32489] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/14/2019] [Indexed: 12/20/2022] Open
Abstract
The mTORC1 inhibitors, such as rapamycin and its analogs, show limited antitumor activity in clinic, reasons for which have not been clearly elucidated. Here, we undertook an effort to uncover the mechanisms underlying the limited efficacy of rapamycin, and found that the transit suppression of 4EBP1 phosphorylation led to cap-dependent translation and cell proliferation in breast cancer cells. AKT only partially contributed to 4EBP1 re-phosphorylation. By taking advantage of mass spectrometry-based phosphoproteomic analysis, we identified SGK3 as a potent kinase involved in 4EBP1 re-phosphorylation. SGK3 deletion inhibited 4EBP1 phosphorylation and cap-dependent translation. Importantly, 4EBP1 phosphorylation was positively correlated with SGK3 activity in 67 clinical breast cancer specimens. Moreover, SGK3 deletion in combination with AKT inhibition almost blocked the 4EBP1 re-phosphorylation that was induced by rapamycin and profoundly enhanced rapamycin-induced growth inhibition in vitro and in an MCF7 breast cancer mouse xenograft model in vivo. Mechanistically, the feedback activation of SGK3 by rapamycin was dependent on hVps34 and mTORC2, and reactivated mTORC1/4EBP1 axis by phosphorylating TSC2. Collectively, our study reveals a critical role of SGK3 in mediating rapamycin resistance, and provides a rationale for targeting SGK3 to improve mTOR-targeted therapies.
Collapse
Affiliation(s)
- Hongtao Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300200, China
| | - Fang Huang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Zhe Zhang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Peng Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hailiang Li
- Zhuhai People's Hospital, Zhuhai Hospital Affiliated Jinan University, Zhuhai 519000, China
| | - Na Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Jian Wang
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Jianguang Zhou
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing 100850, China
| |
Collapse
|
21
|
Chen JB, Zhang M, Zhang XL, Cui Y, Liu PH, Hu J, Li HH, Jin H, Liu LF, Chen MF, Chen HQ, Liang CZ, Zu XB. Glucocorticoid-Inducible Kinase 2 Promotes Bladder Cancer Cell Proliferation, Migration and Invasion by Enhancing β-catenin/c-Myc Signaling Pathway. J Cancer 2018; 9:4774-4782. [PMID: 30588263 PMCID: PMC6299383 DOI: 10.7150/jca.25811] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 10/21/2018] [Indexed: 12/23/2022] Open
Abstract
Background: Bladder cancer is one of the most common malignancies in urologic system. The glucocorticoid-inducible kinase 2 (SGK2) expression and function were largely unknown in cancers. Current study was aimed to investigate the role of SGK2 in bladder cancer and its potential mechanisms. Methods: SGK2 expression was quantified by western blot (WB) in multiple bladder cancer cell lines (T24, 5637, J82 and UMUC3) compared with normal urothelial cell line (SVHUC). SGK2 knocking down and overexpression model were established by lentivirus transfection. MTT, colony formation, wound healing and transwell assay were used to assess the tumor cell proliferation, migration and invasion abilities, respectively. In addition, molecular function analysis was performed using FunRich software V3. Immunoprecipitation (IP) assay was applied to investigate the interaction between SGK2 and β-catenin at protein level. TCGA database was retrieved to verify the association between these genes and clinical tumor stage as well as prognosis among bladder cancer patients. Results: SGK2 expression was significantly upregulated in multiple bladder cancer cell lines compared with SVHUC at protein level. Cell proliferation, migration and invasion abilities were significantly decreased after knocking down SGK2 in J82 and UMUC3 cell lines. Inversely, cell aggressive phenotypes were significantly increased after overexpressing SGK2 in T24 cell line. Furthermore, functional analyses of SGK2 based on TCGA database showed that SGK2 related genes were involved in receptor activity, ATP binding, DNA repair protein, trans-membrane receptor activity and lipid binding. In addition, protein interaction analysis identified c-Myc was significantly enriched in SGK2 positively associated genes. The prediction was validated by WB and IP assay that SGK2 could directly bind with β-catenin at protein level to regulate their downstream gene c-Myc expression in bladder cancer to influence tumor progression. And clinical data generated from TCGA database also identified these downstream genes were significantly associated with tumor stage and survival status of bladder cancer patients. Conclusion: Taken together, our findings suggest SGK2 promotes bladder cancer progression via mediating β-catenin/c-Myc signaling pathway, which may serve as a potential therapeutic target for bladder cancer patients.
Collapse
Affiliation(s)
- Jin-Bo Chen
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Meng Zhang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, NO. 218 Jixi Road, Hefei, Anhui 230022, P.R. China
| | - Xiao-Long Zhang
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, NO.651 East Dongfeng Road, Guangzhou, Guangdong 510060, P.R. China
| | - Yu Cui
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Pei-Hua Liu
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Hui-Huang Li
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Hang Jin
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Long-Fei Liu
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Min-Feng Chen
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - He-Qun Chen
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| | - Chao-Zhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, NO. 218 Jixi Road, Hefei, Anhui 230022, P.R. China
| | - Xiong-Bing Zu
- Department of Urology, Xiangya Hospital, Central South University, NO. 87 Xiangya Road, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
22
|
Identification, structure modification, and characterization of potential small-molecule SGK3 inhibitors with novel scaffolds. Acta Pharmacol Sin 2018; 39:1902-1912. [PMID: 30038340 DOI: 10.1038/s41401-018-0087-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/10/2018] [Indexed: 12/16/2022]
Abstract
The serum and glucocorticoid-regulated kinase (SGK) family has been implicated in the regulation of many cellular processes downstream of the PI3K pathway. It plays a crucial role in PI3K-mediated tumorigenesis, making it a potential therapeutic target for cancer. SGK family consists of three isoforms (SGK1, SGK2, and SGK3), which have high sequence homology in the kinase domain and similar substrate specificity with the AKT family. In order to identify novel compounds capable of inhibiting SGK3 activity, a high-throughput screening campaign against 50,400 small molecules was conducted using a fluorescence-based kinase assay that has a Z' factor above 0.5. It identified 15 hits (including nitrogen-containing aromatic, flavone, hydrazone, and naphthalene derivatives) with IC50 values in the low micromolar to sub-micromolar range. Four compounds with a similar scaffold (i.e., a hydrazone core) were selected for structural modification and 18 derivatives were synthesized. Molecular modeling was then used to investigate the structure-activity relationship (SAR) and potential protein-ligand interactions. As a result, a series of SGK inhibitors that are active against both SGK1 and SGK3 were developed and important functional groups that control their inhibitory activity identified.
Collapse
|
23
|
Ju C, Zhou R, Sun J, Zhang F, Tang X, Chen KK, Zhao J, Lan X, Lin S, Zhang Z, Lv XB. LncRNA SNHG5 promotes the progression of osteosarcoma by sponging the miR-212-3p/SGK3 axis. Cancer Cell Int 2018; 18:141. [PMID: 30250399 PMCID: PMC6145323 DOI: 10.1186/s12935-018-0641-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023] Open
Abstract
Background Long non-coding RNA (lncRNA) SNHG5 has been found to play an important role in tumors. Nevertheless, the function and mechanism of lncRNA SNHG5 in osteosarcoma (OS) remains unclear. The purpose of this study was to investigate whether lncRNA SNHG5 can regulate the occurrence and development of OS cells. Methods We performed quantitative real time PCR to detect the expression of lncRNA SNHG5 in OS cells. 143B, MG63 (knockdown) and U2OS, U2R (overexpression) cell lines were chosen for the function study of SNHG5. The effect of SNHG5, miR-212-3p, and SGK3 in OS cells was explored by MTT assays, clony formation, flow cytometry, transwell assays, wound healing assays, and cell spreading assays. Quantitative real-time PCR, Western blot analysis and luciferase assays were used to detect the interaction between lncRNA SNHG5 and miR-212-3p. Results In this study, knockdown of lncRNA SNHG5 suppressed the growth and metastasis of OS cells, whereas the overexpression of SNHG5 produced an opposite result. Mechanistically, lncRNA SNHG5 functions as a sponger against miR-212-3p and suppresses the miR-212-3p/SGK3 signaling pathway. Introduction of miR-212-3p mimics or inhibitors reverses SNHG5 overexpression or silences the exerted tumor promoting or suppressing effect. In addition, our results showed that the function of SNHG5 can be rescued by miR-212-3p and can regulate the growth and metastasis of OS cells via SGK3, the downstream target of miR-212-3p. Conclusions In summary, our study demonstrated that lncRNA SNHG5 can regulate the proliferation and metastasis of OS cells through the miR-212-3p/SGK3 axis. This axis may provide a new target for future clinical treatment. Electronic supplementary material The online version of this article (10.1186/s12935-018-0641-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cheng Ju
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China.,2Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China.,3Medical Department of Graduate School, Nanchang University, Nanchang, 330006 Jiangxi People's Republic of China
| | - Ruihao Zhou
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China.,4First Clinical Department, Medical School of Nanchang University, Nanchang, 330006 Jiangxi People's Republic of China
| | - Jun Sun
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| | - Feifei Zhang
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| | - Xiaofeng Tang
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| | - Kaddie Kwok Chen
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| | - Junliang Zhao
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China.,4First Clinical Department, Medical School of Nanchang University, Nanchang, 330006 Jiangxi People's Republic of China
| | - Xiaoyong Lan
- 2Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| | - Shifan Lin
- 2Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| | - Zhiping Zhang
- 2Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| | - Xiao-Bin Lv
- 1Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, 128 Xiangshan Northern Road, Nanchang, 330008 Jiangxi People's Republic of China
| |
Collapse
|
24
|
Chen X, Hu X, Li Y, Zhu C, Dong X, Zhang R, Ma J, Huang S, Chen L. Resveratrol inhibits Erk1/2-mediated adhesion of cancer cells via activating PP2A-PTEN signaling network. J Cell Physiol 2018; 234:2822-2836. [PMID: 30066962 DOI: 10.1002/jcp.27100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
Abstract
Resveratrol, a natural polyphenol compound, has been shown to possess anticancer activity. However, how resveratrol inhibits cancer cell adhesion has not been fully elucidated. Here, we show that resveratrol suppressed the basal or type I insulin-like growth factor (IGF)-1-stimulated adhesion of cancer cells (Rh1, Rh30, HT29, and HeLa cells) by inhibiting the extracellular signal-regulated kinase 1/2 (Erk1/2) pathway. Inhibition of Erk1/2 with U0126, knockdown of Erk1/2, or overexpression of dominant-negative mitogen-activated protein kinase kinase 1 (MKK1) strengthened resveratrol's inhibition of the basal or IGF-1-stimulated of Erk1/2 phosphorylation and cell adhesion, whereas ectopic expression of constitutively active MKK1 attenuated the inhibitory effects of resveratrol. Further research revealed that both protein phosphatase 2A (PP2A) and phosphatase and tensin homolog (PTEN)-Akt were implicated in resveratrol-inactivated Erk1/2-dependent cell adhesion. Inhibition of PP2A with okadaic acid or overexpression of dominant-negative PP2A rendered resistance to resveratrol's suppression of the basal or IGF-1-stimulated phospho-Erk1/2 and cell adhesion, whereas expression of wild-type PP2A enhanced resveratrol's inhibitory effects. Overexpression of wild-type PTEN or dominant-negative Akt or inhibition of Akt with Akt inhibitor X strengthened resveratrol's inhibition of the basal or IGF-1-stimulated Erk1/2 phosphorylation and cell adhesion. Furthermore, inhibition of mechanistic/mammalian target of rapamycin (mTOR) with rapamycin or silencing mTOR enhanced resveratrol's inhibitory effects on the basal and IGF-1-induced inhibition of PP2A-PTEN, activation of Akt-Erk1/2, and cell adhesion. The results indicate that resveratrol inhibits Erk1/2-mediated adhesion of cancer cells via activating PP2A-PTEN signaling network. Our data highlight that resveratrol has a great potential in the prevention of cancer cell adhesion.
Collapse
Affiliation(s)
- Xin Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiaoyu Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yue Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Cuilan Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiaoqing Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruijie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jing Ma
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
25
|
Liu F, Wu X, Jiang X, Qian Y, Gao J. Prolonged inhibition of class I PI3K promotes liver cancer stem cell expansion by augmenting SGK3/GSK-3β/β-catenin signalling. J Exp Clin Cancer Res 2018; 37:122. [PMID: 29940988 PMCID: PMC6020243 DOI: 10.1186/s13046-018-0801-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/07/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Serum and glucocorticoid-regulated kinase 3 (SGK3) has been reported to play an important role in tumour progression, but its role in cancer stem cells (CSCs) remains obscure. The phosphoinositide 3-kinase (PI3K) pathway is considered a hallmark of cancer. Although many PI3K pathway-targeted therapies have been tested in oncology trials, the results are not satisfactory. METHODS We used spheroids cultured in serum-free culture medium and MicroBead isolation to obtain liver CSCs. Spheroid formation assay and flow cytometric analysis were performed to investigate liver CSC expansion. Real-time polymerase chain reaction (PCR), western blot and immunofluorescence were used to assess gene expression in cell lines. RESULTS We found that SGK3 is preferentially activated in liver CSCs. Upregulated SGK3 significantly increases the expansion of liver CSCs. Conversely, suppression of SGK3 in human hepatocarcinoma (HCC) cells had an opposite effect. Mechanistically, SGK3 promoted β-catenin accumulation by suppressing GSK-3β-mediated β-catenin degradation in liver CSCs, and then promoting the expansion of liver CSCs. Prolonged treatment of HCC cells with class I PI3K inhibitors leads to activation of SGK3 and expansion of liver CSCs. Inhibition of hVps34 can block SGK3 activity and suppress liver CSC expansion induced by PI3K inhibitors. More importantly, we also found that prolonged treatment of HCC cells with PI3K inhibitors stimulates the β-catenin signalling pathway via activation of SGK3. CONCLUSIONS Prolonged inhibition of class I PI3K promotes liver CSC expansion by augmenting SGK3-dependent β-catenin stabilisation, and effective inhibition of SGK3 signalling may be useful in eliminating liver CSCs and in PI3K pathway-targeted cancer therapies.
Collapse
Affiliation(s)
- Fengchao Liu
- Department of Gastroenterology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaoling Wu
- Department of Gastroenterology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Jiang
- Department of Gastroenterology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yanzhi Qian
- Department of Gastroenterology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jian Gao
- Department of Gastroenterology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Jin H, Yang L, Wang L, Yang Z, Zhan Q, Tao Y, Zou Q, Tang Y, Xian J, Zhang S, Jing Y, Zhang L. INPP4B promotes cell survival via SGK3 activation in NPM1-mutated leukemia. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:8. [PMID: 29343273 PMCID: PMC5773044 DOI: 10.1186/s13046-018-0675-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 01/04/2018] [Indexed: 12/15/2022]
Abstract
Background Acute myeloid leukemia (AML) with mutated nucleophosmin (NPM1) has been recognized as a distinct leukemia entity in the 2016 World Health Organization (WHO) classification. The genetic events underlying oncogenesis in NPM1-mutated AML that is characterized by a normal karyotype remain unclear. Inositol polyphosphate 4-phosphatase type II (INPP4B), a new factor in the phosphoinositide-3 kinase (PI3K) pathway-associated cancers, has been recently found a clinically relevant role in AML. However, little is known about the specific mechanistic function of INPP4B in NPM1-mutated AML. Methods The INPP4B expression levels in NPM1-mutated AML primary blasts and AML OCI-AML3 cell lines were determined by qRT-PCR and western blotting. The effect of INPP4B knockdown on OCI-AML3 leukemia cell proliferation was evaluated, using the Cell Counting Kit-8 and colony formation assay. After INPP4B overexpression or knockdown, the activation of serum and glucocorticoid-regulated kinase 3 (SGK3) and AKT was assessed. The effects of PI3K signaling pathway inhibitors on the levels of p-SGK3 in OCI-AML3 cells were tested. The mass of PI (3,4) P2 and PI (3) P was analyzed by ELISA upon INPP4B overexpression. Knockdown of SGK3 by RNA interference and a rescue assay were performed to confirm the critical role of SGK3 in INPP4B-mediated cell survival. In addition, the molecular mechanism underlying INPP4B expression in NPM1-mutated leukemia cells was explored. Finally, Kaplan–Meier survival analysis was conducted on the NPM1-mutated AML cohort stratified into quartiles for INPP4B expression in The Cancer Genome Atlas (TCGA) dataset. Results High expression of INPP4B was observed in NPM1-mutated AML. Knockdown of INPP4B repressed cell proliferation in OCI-AML3 cells, whereas recovered INPP4B rescued this inhibitory effect in vitro. Mechanically, INPP4B enhanced phosphorylated SGK3 (p-SGK3) status, but did not affect AKT activation. SGK3 was required for INPP4B-induced cell proliferation in OCI-AML3 cells. High levels of INPP4B were at least partially caused by the NPM1 mutant via ERK/Ets-1 signaling. Finally, high expression of INPP4B showed a trend towards lower overall survival and event-free survival in NPM1-mutated AML patients. Conclusions Our results indicate that INPP4B promotes leukemia cell survival via SGK3 activation, and INPP4B might be a potential target in the treatment of NPM1-mutated AML.
Collapse
Affiliation(s)
- Hongjun Jin
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Liyuan Yang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Lu Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Zailin Yang
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qian Zhan
- The Center for Clinical Molecular Medical detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yao Tao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Qin Zou
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Yuting Tang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Jingrong Xian
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Shuaishuai Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Yipei Jing
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China
| | - Ling Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China. No.1, Yixueyuan Road, Chongqing, 400016, China.
| |
Collapse
|
27
|
Wu M, Huang C, Huang X, Liang R, Feng Y, Luo X. MicroRNA-144-3p suppresses tumor growth and angiogenesis by targeting SGK3 in hepatocellular carcinoma. Oncol Rep 2017; 38:2173-2181. [PMID: 28849156 PMCID: PMC5652965 DOI: 10.3892/or.2017.5900] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/03/2017] [Indexed: 12/27/2022] Open
Abstract
In our previous studies, the Illumine Soledad massively parallel signature sequencing of miRNomes in non-tumor and hepatocellular carcinoma (HCC) tissues revealed that microRNA (miR)-144-3p was significantly downregulated in HCC, but its role in HCC development, especially angiogenesis, remains unclear. In this investigation, we found recovering miR-144-3p expression can significantly suppress the growth, migration and induced angiogenic capacity of HCC cells through both in vivo and in vitro experiments. Moreover, clinical correlation analysis showed that low expression of miR-144-3p was positively correlated to poor disease-free survival (DFS) of HCC patients. Mechanistically, serum and glucocorticoid kinase 3 (SGK3), the putative targets of miR-144-3p, was predicted by Target Scan database and identified to be suppressed by miR-144-3p so that inhibiting the activation of mTOR-VEGF downstream signals was activated by the phosphoinositide 3-kinase (PI3K)-independent pathway. Hence, we concluded that miR-144-3p, which is frequently downregulated in HCC, can inhibit proliferation, migration and repress angiogenesis by regulating SGK3 activation with PI3K independent signal pathway, and acts as a prognostic factor for HCC patients.
Collapse
Affiliation(s)
- Manya Wu
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chaoyuan Huang
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xinping Huang
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rong Liang
- First Department of Chemotherapy, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yan Feng
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiaoling Luo
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
28
|
Brenner AK, Andersson Tvedt TH, Bruserud Ø. The Complexity of Targeting PI3K-Akt-mTOR Signalling in Human Acute Myeloid Leukaemia: The Importance of Leukemic Cell Heterogeneity, Neighbouring Mesenchymal Stem Cells and Immunocompetent Cells. Molecules 2016; 21:molecules21111512. [PMID: 27845732 PMCID: PMC6273124 DOI: 10.3390/molecules21111512] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/11/2022] Open
Abstract
Therapeutic targeting of PI3K-Akt-mTOR is considered a possible strategy in human acute myeloid leukaemia (AML); the most important rationale being the proapoptotic and antiproliferative effects of direct PI3K/mTOR inhibition observed in experimental studies of human AML cells. However, AML is a heterogeneous disease and these effects caused by direct pathway inhibition in the leukemic cells are observed only for a subset of patients. Furthermore, the final effect of PI3K-Akt-mTOR inhibition is modulated by indirect effects, i.e., treatment effects on AML-supporting non-leukemic bone marrow cells. In this article we focus on the effects of this treatment on mesenchymal stem cells (MSCs) and monocytes/macrophages; both these cell types are parts of the haematopoietic stem cell niches in the bone marrow. MSCs have unique membrane molecule and constitutive cytokine release profiles, and mediate their support through bidirectional crosstalk involving both cell-cell contact and the local cytokine network. It is not known how various forms of PI3K-Akt-mTOR targeting alter the molecular mechanisms of this crosstalk. The effect on monocytes/macrophages is also difficult to predict and depends on the targeted molecule. Thus, further development of PI3K-Akt-mTOR targeting into a clinical strategy requires detailed molecular studies in well-characterized experimental models combined with careful clinical studies, to identify patient subsets that are likely to respond to this treatment.
Collapse
Affiliation(s)
- Annette K Brenner
- Section for Haematology, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
| | - Tor Henrik Andersson Tvedt
- Section for Haematology, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway.
| | - Øystein Bruserud
- Section for Haematology, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway.
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway.
| |
Collapse
|
29
|
Chi MN, Guo ST, Wilmott JS, Guo XY, Yan XG, Wang CY, Liu XY, Jin L, Tseng HY, Liu T, Croft A, Hondermarck H, Scolyer RA, Jiang CC, Zhang XD. INPP4B is upregulated and functions as an oncogenic driver through SGK3 in a subset of melanomas. Oncotarget 2016; 6:39891-907. [PMID: 26573229 PMCID: PMC4741868 DOI: 10.18632/oncotarget.5359] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 10/27/2015] [Indexed: 01/15/2023] Open
Abstract
Inositol polyphosphate 4-phosphatase type II (INPP4B) negatively regulates PI3K/Akt signalling and has a tumour suppressive role in some types of cancers. However, we have found that it is upregulated in a subset of melanomas. Here we report that INPP4B can function as an oncogenic driver through activation of serum- and glucocorticoid-regulated kinase 3 (SGK3) in melanoma. While INPP4B knockdown inhibited melanoma cell proliferation and retarded melanoma xenograft growth, overexpression of INPP4B enhanced melanoma cell and melanocyte proliferation and triggered anchorage-independent growth of melanocytes. Noticeably, INPP4B-mediated melanoma cell proliferation was not related to activation of Akt, but was mediated by SGK3. Upregulation of INPP4B in melanoma cells was associated with loss of miRNA (miR)-494 and/or miR-599 due to gene copy number reduction. Indeed, overexpression of miR-494 or miR-599 downregulated INPP4B, reduced SGK3 activation, and inhibited melanoma cell proliferation, whereas introduction of anti-miR-494 or anti-miR-599 upregulated INPP4B, enhanced SGK3 activation, and promoted melanoma cell proliferation. Collectively, these results identify upregulation of INPP4B as an oncogenic mechanism through activation of SGK3 in a subset of melanomas, with implications for targeting INPP4B and restoring miR-494 and miR-599 as novel approaches in the treatment of melanomas with high INPP4B expression.
Collapse
Affiliation(s)
- Meng Na Chi
- School of Medicine and Public Health, The University of Newcastle, NSW 2308, Australia
| | - Su Tang Guo
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, NSW 2308, Australia.,Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030013, China
| | - James S Wilmott
- Discipline of Pathology, The University of Sydney, and Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW 2006, Australia
| | - Xiang Yun Guo
- Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030013, China
| | - Xu Guang Yan
- School of Medicine and Public Health, The University of Newcastle, NSW 2308, Australia
| | - Chun Yan Wang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, NSW 2308, Australia.,Department of Molecular Biology, Shanxi Cancer Hospital and Institute, Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030013, China
| | - Xiao Ying Liu
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, NSW 2308, Australia
| | - Lei Jin
- School of Medicine and Public Health, The University of Newcastle, NSW 2308, Australia
| | - Hsin-Yi Tseng
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, NSW 2308, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Amanda Croft
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, NSW 2308, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, NSW 2308, Australia
| | - Richard A Scolyer
- Discipline of Pathology, The University of Sydney, and Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW 2006, Australia
| | - Chen Chen Jiang
- School of Medicine and Public Health, The University of Newcastle, NSW 2308, Australia
| | - Xu Dong Zhang
- School of Medicine and Public Health, The University of Newcastle, NSW 2308, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, NSW 2308, Australia
| |
Collapse
|
30
|
Bago R, Sommer E, Castel P, Crafter C, Bailey FP, Shpiro N, Baselga J, Cross D, Eyers PA, Alessi DR. The hVps34-SGK3 pathway alleviates sustained PI3K/Akt inhibition by stimulating mTORC1 and tumour growth. EMBO J 2016; 35:1902-22. [PMID: 27481935 PMCID: PMC5007552 DOI: 10.15252/embj.201693929] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 07/04/2016] [Indexed: 12/16/2022] Open
Abstract
We explore mechanisms that enable cancer cells to tolerate PI3K or Akt inhibitors. Prolonged treatment of breast cancer cells with PI3K or Akt inhibitors leads to increased expression and activation of a kinase termed SGK3 that is related to Akt. Under these conditions, SGK3 is controlled by hVps34 that generates PtdIns(3)P, which binds to the PX domain of SGK3 promoting phosphorylation and activation by its upstream PDK1 activator. Furthermore, under conditions of prolonged PI3K/Akt pathway inhibition, SGK3 substitutes for Akt by phosphorylating TSC2 to activate mTORC1. We characterise 14h, a compound that inhibits both SGK3 activity and activation in vivo, and show that a combination of Akt and SGK inhibitors induced marked regression of BT‐474 breast cancer cell‐derived tumours in a xenograft model. Finally, we present the kinome‐wide analysis of mRNA expression dynamics induced by PI3K/Akt inhibition. Our findings highlight the importance of the hVps34‐SGK3 pathway and suggest it represents a mechanism to counteract inhibition of PI3K/Akt signalling. The data support the potential of targeting both Akt and SGK as a cancer therapeutic.
Collapse
Affiliation(s)
- Ruzica Bago
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| | - Eeva Sommer
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| | - Pau Castel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Claire Crafter
- Oncology iMED, AstraZeneca CRUK Cambridge Institute, Cambridge, UK
| | - Fiona P Bailey
- Department of Biochemistry, Institute of Integrative Biology University of Liverpool, Liverpool, UK
| | - Natalia Shpiro
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| | - José Baselga
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Darren Cross
- Oncology iMED, AstraZeneca CRUK Cambridge Institute, Cambridge, UK
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology University of Liverpool, Liverpool, UK
| | - Dario R Alessi
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| |
Collapse
|
31
|
Brasseur K, Fabi F, Adam P, Parent S, Lessard L, Asselin E. Post-translational regulation of the cleaved fragment of Par-4 in ovarian and endometrial cancer cells. Oncotarget 2016; 7:36971-36987. [PMID: 27175591 PMCID: PMC5095052 DOI: 10.18632/oncotarget.9235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/24/2016] [Indexed: 11/25/2022] Open
Abstract
We recently reported the caspase3-dependent cleavage of Par-4 resulting in the accumulation of a 25kDa cleaved-Par-4 (cl-Par-4) fragment and we investigated in the present study the mechanisms regulating this fragment using cl-Par-4-expressing stable clones derived from ovarian and endometrial cancer cell lines.Cl-Par-4 protein was weakly express in all stable clones despite constitutive expression. However, upon cisplatin treatment, cl-Par-4 levels increased up to 50-fold relative to baseline conditions. Treatment of stable clones with proteasome and translation inhibitors revealed that cisplatin exposure might in fact protect cl-Par-4 from proteasome-dependent degradation. PI3K and MAPK pathways were also implicated as evidenced by an increase of cl-Par-4 in the presence of PI3K inhibitors and a decrease using MAPK inhibitors. Finally using bioinformatics resources, we found diverse datasets showing similar results to those we observed with the proteasome and cl-Par-4 further supporting our data.These new findings add to the complex mechanisms regulating Par-4 expression and activity, and justify further studies addressing the biological significance of this phenomenon in gynaecological cancer cells.
Collapse
Affiliation(s)
- Kevin Brasseur
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada
| | - François Fabi
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada
| | - Pascal Adam
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada
| | - Sophie Parent
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada
| | - Laurent Lessard
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada
| | - Eric Asselin
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada
| |
Collapse
|
32
|
Tsc1 deficiency impairs mammary development in mice by suppression of AKT, nuclear ERα, and cell-cycle-driving proteins. Sci Rep 2016; 6:19587. [PMID: 26795955 PMCID: PMC4726182 DOI: 10.1038/srep19587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/15/2015] [Indexed: 01/10/2023] Open
Abstract
Loss of Tsc1/Tsc2 results in excess cell growth that eventually forms hamartoma in multiple organs. Our study using a mouse model with Tsc1 conditionally knockout in mammary epithelium showed that Tsc1 deficiency impaired mammary development. Phosphorylated S6 was up-regulated in Tsc1−/− mammary epithelium, which could be reversed by rapamycin, suggesting that mTORC1 was hyperactivated in Tsc1−/− mammary epithelium. The mTORC1 inhibitor rapamycin restored the development of Tsc1−/− mammary glands whereas suppressed the development of Tsc1wt/wt mammary glands, indicating that a modest activation of mTORC1 is critical for mammary development. Phosphorylated PDK1 and AKT, nuclear ERα, nuclear IRS-1, SGK3, and cell cycle regulators such as Cyclin D1, Cyclin E, CDK2, CDK4 and their target pRB were all apparently down-regulated in Tsc1−/− mammary glands, which could be reversed by rapamycin, suggesting that suppression of AKT by hyperactivation of mTORC1, inhibition on nuclear ERα signaling, and down-regulation of cell-cycle-driving proteins play important roles in the retarded mammary development induced by Tsc1 deletion. This study demonstrated for the first time the in vivo role of Tsc1 in pubertal mammary development of mice, and revealed that loss of Tsc1 does not necessarily lead to tissue hyperplasia.
Collapse
|
33
|
Wharton's Jelly-Derived Mesenchymal Stromal Cells and Fibroblast-Derived Extracellular Matrix Synergistically Activate Apoptosis in a p21-Dependent Mechanism in WHCO1 and MDA MB 231 Cancer Cells In Vitro. Stem Cells Int 2016; 2016:4842134. [PMID: 26880967 PMCID: PMC4737007 DOI: 10.1155/2016/4842134] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/13/2015] [Accepted: 12/06/2015] [Indexed: 12/14/2022] Open
Abstract
The tumour microenvironment plays a crucial role in tumour progression and comprises tumour stroma which is made up of different cell types and the extracellular matrix (ECM). Mesenchymal stromal cells (MSCs) are part of the tumour stroma and may have conflicting effects on tumour growth. In this study we investigated the effect of Wharton's Jelly-derived MSCs (WJ-MSCs) and a fibroblast-derived ECM (fd-ECM) on esophageal (WHCO1) and breast (MDA MB 231) cancer cells in vitro. Both WJ-MSCs and the fd-ECM, alone or in combination, downregulate PCNA, cyclin D1, Bcl-2, Bcl-xL, and MMPs and upregulate p53 and p21. p21 induction resulted in G2 phase cell cycle arrest and induced apoptosis in vitro. Our data suggest that p21 induction is via p53-dependent and p53-independent mechanisms in WHCO1 and MDA MB 231 cells, respectively. Vascular endothelial growth factor, Akt, and Nodal pathways were downregulated in cancer cells cocultured with WJ-MSCs. We also demonstrate that WJ-MSCs effects on cancer cells appear to be short-lived whilst the fd-ECM effect is long-lived. This study shows the influence of tumour microenvironment on cancer cell behaviour and provides alternative therapeutic targets for potential regulation of tumour cells.
Collapse
|
34
|
Chen Y, Tang Q, Wu J, Zheng F, Yang L, Hann SS. Inactivation of PI3-K/Akt and reduction of SP1 and p65 expression increase the effect of solamargine on suppressing EP4 expression in human lung cancer cells. J Exp Clin Cancer Res 2015; 34:154. [PMID: 26689593 PMCID: PMC4687355 DOI: 10.1186/s13046-015-0272-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 12/15/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Lung cancer is the most common cause of cancer-related deaths worldwide. Natural phytochemicals from traditional medicinal plants such as solamargine have been shown to have anticancer properties. The prostaglandin E2 receptor EP4 is highly expressed in human cancer, however, the functional role of EP4 in the occurrence and progression of non small cell lung cancer (NSCLC) remained to be elucidated. METHODS Cell viability was measured by MTT assays. Western blot was performed to measure the phosphorylation and protein expression of PI3-K downstream effector Akt, transcription factors SP1, p65, and EP4. Quantitative real-time PCR (qRT-PCR) was used to examine the mRNA levels of EP4 gene. Exogenous expression of SP1, p65, and EP4 genes was carried out by transient transfection assays. EP4 promoter activity was measured by Dual Luciferase Reporter Kit. RESULTS We showed that solamargine inhibited the growth of lung cancer cells. Mechanistically, we found that solamargine decreased the phosphorylation of Akt, the protein, mRNA expression, and promoter activity of EP4. Moreover, solamargine inhibited protein expression of SP1 and NF-κB subunit p65, all of which were abrogated in cells transfected with exogenous expressed Akt. Intriguingly, exogenous expressed SP1 overcame the effect of solamargine on inhibition of p65 protein expression, and EP4 protein expression and promoter activity. Finally, exogenous expressed EP4 feedback reversed the effect of solamargine on phosphorylation of Akt and cell growth inhibition. CONCLUSION Our results show that solamargine inhibits the growth of human lung cancer cells through inactivation of Akt signaling, followed by reduction of SP1 and p65 protein expression. This results in the inhibition of EP4 gene expression. The cross-talk between SP1 and p65, and the positive feedback regulatory loop of PI3-K/Akt signaling by EP4 contribute to the overall responses of solamargine in this process. This study unveils a novel mechanism by which solamargine inhibits growth of human lung cancer cells.
Collapse
MESH Headings
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Oncogene Protein v-akt/biosynthesis
- Oncogene Protein v-akt/genetics
- Phosphatidylinositol 3-Kinases/biosynthesis
- Phosphatidylinositol 3-Kinases/genetics
- Phosphorylation/drug effects
- Promoter Regions, Genetic
- RNA, Messenger/biosynthesis
- Receptors, Prostaglandin E, EP4 Subtype/biosynthesis
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Signal Transduction/drug effects
- Solanaceous Alkaloids/administration & dosage
- Sp1 Transcription Factor/biosynthesis
- Sp1 Transcription Factor/genetics
- Transcription Factor RelA/biosynthesis
- Transcription Factor RelA/genetics
Collapse
Affiliation(s)
- YuQing Chen
- Laboratory of Tumor Biology, Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Qing Tang
- Laboratory of Tumor Biology, Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - JingJing Wu
- Laboratory of Tumor Biology, Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Fang Zheng
- Laboratory of Tumor Biology, Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - LiJun Yang
- Laboratory of Tumor Biology, Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
| | - Swei Sunny Hann
- Laboratory of Tumor Biology, Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, China.
- Higher Education Mega Center, No. 55, Neihuan West Road, Panyu District, Guangzhou, Guangdong Province, 510006, PR China.
| |
Collapse
|
35
|
Lou H, Villagran G, Boland JF, Im KM, Polo S, Zhou W, Odey U, Juárez-Torres E, Medina-Martínez I, Roman-Basaure E, Mitchell J, Roberson D, Sawitzke J, Garland L, Rodríguez-Herrera M, Wells D, Troyer J, Pinto FC, Bass S, Zhang X, Castillo M, Gold B, Morales H, Yeager M, Berumen J, Alvirez E, Gharzouzi E, Dean M. Genome Analysis of Latin American Cervical Cancer: Frequent Activation of the PIK3CA Pathway. Clin Cancer Res 2015; 21:5360-70. [PMID: 26080840 PMCID: PMC4668220 DOI: 10.1158/1078-0432.ccr-14-1837] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 05/14/2015] [Indexed: 12/30/2022]
Abstract
PURPOSE Cervical cancer is one of the most common causes of cancer mortality for women living in poverty, causing more than 28,000 deaths annually in Latin America and 266,000 worldwide. To better understand the molecular basis of the disease, we ascertained blood and tumor samples from Guatemala and Venezuela and performed genomic characterization. EXPERIMENTAL DESIGN We performed human papillomavirus (HPV) typing and identified somatically mutated genes using exome and ultra-deep targeted sequencing with confirmation in samples from Mexico. Copy number changes were also assessed in the exome sequence. RESULTS Cervical cancer cases in Guatemala and Venezuela have an average age of diagnosis of 50 years and 5.6 children. Analysis of 675 tumors revealed activation of PIK3CA and other PI3K/AKT pathway genes in 31% of squamous carcinomas and 24% of adeno- and adenosquamous tumors, predominantly at two sites (E542K, E545K) in the helical domain of the PIK3CA gene. This distribution of PIK3CA mutations is distinct from most other cancer types and does not result in the in vitro phosphorylation of AKT. Somatic mutations were more frequent in squamous carcinomas diagnosed after the age of 50 years. Frequent gain of chromosome 3q was found, and low PIK3CA mutation fractions in many tumors suggest that PI3K mutation can be a late event in tumor progression. CONCLUSIONS PI3K pathway mutation is important to cervical carcinogenesis in Latin America. Therapeutic agents that directly target PI3K could play a role in the therapy of this common malignancy.
Collapse
Affiliation(s)
- Hong Lou
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick, Maryland
| | | | - Joseph F Boland
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Gaithersburg, Maryland
| | - Kate M Im
- Laboratory of Experimental Immunology, National Cancer Institute, Frederick, Maryland
| | - Sarita Polo
- Instituto de Cancerologia, Guatemala City, Guatemala
| | - Weiyin Zhou
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Gaithersburg, Maryland
| | - Ushie Odey
- Hospital Central Universitario "Dr. Antonio M Pineda," Barquisimeto, Lara State, Venezuela
| | - Eligia Juárez-Torres
- Unidad de Medicina Genómica, Hospital General de México/Facultad de Medicina, Universidad Nacional Autónoma de México, México, D.F. México
| | - Ingrid Medina-Martínez
- Unidad de Medicina Genómica, Hospital General de México/Facultad de Medicina, Universidad Nacional Autónoma de México, México, D.F. México
| | | | - Jason Mitchell
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Gaithersburg, Maryland
| | - David Roberson
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Gaithersburg, Maryland
| | - Julie Sawitzke
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Lisa Garland
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick, Maryland
| | | | - David Wells
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Jennifer Troyer
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick, Maryland
| | | | - Sara Bass
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Gaithersburg, Maryland
| | - Xijun Zhang
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Gaithersburg, Maryland
| | | | - Bert Gold
- Laboratory of Experimental Immunology, National Cancer Institute, Frederick, Maryland
| | | | - Meredith Yeager
- Cancer Genetics Research Laboratory, Division of Cancer Epidemiology and Genetics, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Gaithersburg, Maryland
| | - Jaime Berumen
- Unidad de Medicina Genómica, Hospital General de México/Facultad de Medicina, Universidad Nacional Autónoma de México, México, D.F. México
| | - Enrique Alvirez
- Hospital Central Universitario "Dr. Antonio M Pineda," Barquisimeto, Lara State, Venezuela
| | | | - Michael Dean
- Laboratory of Experimental Immunology, National Cancer Institute, Frederick, Maryland.
| |
Collapse
|
36
|
Faes S, Dormond O. PI3K and AKT: Unfaithful Partners in Cancer. Int J Mol Sci 2015; 16:21138-52. [PMID: 26404259 PMCID: PMC4613246 DOI: 10.3390/ijms160921138] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 08/28/2015] [Accepted: 09/01/2015] [Indexed: 02/07/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway regulates multiple cellular processes. An overactivation of the pathway is frequently present in human malignancies and plays a key role in cancer progression. Hence, its inhibition has become a promising approach in cancer therapy. However, the development of resistances, such as the abrogation of negative feedback mechanisms or the activation of other proliferative signaling pathways, has considerably limited the anticancer efficacy of PI3K/AKT inhibitors. In addition, emerging evidence points out that although AKT is acknowledged as the major downstream effector of PI3K, both PI3K and AKT can operate independently of each other in cancer, revealing another level of complexity in this pathway. Here, we highlight the complex relationship between PI3K and AKT in cancer and further discuss the consequences of this relationship for cancer therapy.
Collapse
Affiliation(s)
- Seraina Faes
- Department of Visceral Surgery, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Pavillon 4, Av. de Beaumont, Lausanne 1011, Switzerland.
| | - Olivier Dormond
- Department of Visceral Surgery, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Pavillon 4, Av. de Beaumont, Lausanne 1011, Switzerland.
| |
Collapse
|
37
|
Deregulation of the EGFR/PI3K/PTEN/Akt/mTORC1 pathway in breast cancer: possibilities for therapeutic intervention. Oncotarget 2015; 5:4603-50. [PMID: 25051360 PMCID: PMC4148087 DOI: 10.18632/oncotarget.2209] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The EGFR/PI3K/PTEN/Akt/mTORC1/GSK-3 pathway plays prominent roles in malignant transformation, prevention of apoptosis, drug resistance and metastasis. The expression of this pathway is frequently altered in breast cancer due to mutations at or aberrant expression of: HER2, ERalpha, BRCA1, BRCA2, EGFR1, PIK3CA, PTEN, TP53, RB as well as other oncogenes and tumor suppressor genes. In some breast cancer cases, mutations at certain components of this pathway (e.g., PIK3CA) are associated with a better prognosis than breast cancers lacking these mutations. The expression of this pathway and upstream HER2 has been associated with breast cancer initiating cells (CICs) and in some cases resistance to treatment. The anti-diabetes drug metformin can suppress the growth of breast CICs and herceptin-resistant HER2+ cells. This review will discuss the importance of the EGFR/PI3K/PTEN/Akt/mTORC1/GSK-3 pathway primarily in breast cancer but will also include relevant examples from other cancer types. The targeting of this pathway will be discussed as well as clinical trials with novel small molecule inhibitors. The targeting of the hormone receptor, HER2 and EGFR1 in breast cancer will be reviewed in association with suppression of the EGFR/PI3K/PTEN/Akt/mTORC1/GSK-3 pathway.
Collapse
|
38
|
Hou M, Lai Y, He S, He W, Shen H, Ke Z. SGK3 (CISK) may induce tumor angiogenesis (Hypothesis). Oncol Lett 2015; 10:23-26. [PMID: 26170971 DOI: 10.3892/ol.2015.3182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 03/27/2015] [Indexed: 12/18/2022] Open
Abstract
Serum- and glucocorticoid-inducible protein kinase 3 (SGK3), also known as cytokine-independent survival kinase (CISK), encoded by chromosome 8q12.2, is a downstream mediator of phosphatidylinositol 3-kinase (PI3K) oncogenic signaling. As a downstream target of PI3K, SGK3 has been reported to mediate pivotal roles in oncogenic progress in various cancers, including breast cancer, ovarian cancer and hepatocellular carcinoma. Functionally parallel to v-akt murine thymoma viral oncogene homolog (AKT)/protein kinase B, SGK3 serves as a hallmark mediating glycogen synthase kinase-β (GSK3-β), B-cell lymphoma (Bcl)-2-associated death promoter, forkead family of transcription factors, Bcl-extra large, Bcl-2, mammalian target of rapamycin, C-X-C chemokine receptor type 4 (CXCR4) and numerous other molecules in cell proliferation, growth, survival, migration and even tumor angiogenesis. Tumor angiogenesis is recognized as an essential step for tumor growth, invasion and metastasis, and it has become an intriguing target for anticancer drug development for tumor investigators worldwide. An abundance of experiments have been performed to investigate the role of the phosphoinositide 3-kinase (PI3K)/AKT pathway in regulating tumor angiogenesis. The mechanism of angiogenesis regulated by the PI3K/AKT pathway is, to a certain extent, clear. Although a number of SGK3 target molecules, including CXCR4 and GSK3β, have demonstrated potential roles in promoting angiogenesis, the exact association between angiogenesis and SGK3 remains unclear. Thus, we hypothesize that SGK3, parallel to AKT, may also be important in mediating angiogenesis. Identifying the role of SGK3 in tumor angiogenesis will certainly present a novel perspective on the malignant transformation of tumors, as well as a target for tumor therapy.
Collapse
Affiliation(s)
- Minzhi Hou
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yingrong Lai
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Shanyang He
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hongwei Shen
- Department of Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
39
|
Hausmann S, Brandt E, Köchel C, Einsele H, Bargou RC, Seggewiss-Bernhardt R, Stühmer T. Loss of serum and glucocorticoid-regulated kinase 3 (SGK3) does not affect proliferation and survival of multiple myeloma cell lines. PLoS One 2015; 10:e0122689. [PMID: 25837824 PMCID: PMC4383545 DOI: 10.1371/journal.pone.0122689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 02/15/2015] [Indexed: 11/28/2022] Open
Abstract
Multiple myeloma (MM) is a generally fatal plasma cell cancer that often shows activation of the phosphoinositide 3-kinase/Akt (PI3K/Akt) pathway. Targeted pharmacologic therapies, however, have not yet progressed beyond the clinical trial stage, and given the complexity of the PI3K/Akt signalling system (e.g. multiple protein isoforms, diverse feedback regulation mechanisms, strong variability between patients) it is mandatory to characterise its ramifications in order to better guide informed decisions about the best therapeutic approaches. Here we explore whether serum and glucocorticoid-regulated kinase 3 (SGK3), a potential downstream effector of PI3K, plays a role in oncogenic signalling in MM cells—either in concert with or independent of Akt. SGK3 was expressed in all MM cell lines and in all primary MM samples tested. Four MM cell lines representing a broad range of intrinsic Akt activation (very strong: MM.1s, moderate: L 363 and JJN-3, absent: AMO-1) were chosen to test the effects of transient SGK3 knockdown alone and in combination with pharmacological inhibition of Akt, PI3K-p110α, or in the context of serum starvation. Although the electroporation protocol led to strong SGK3 depletion for at least 5 days its absence had no substantial effect on the activation status of potential downstream substrates, or on the survival, viability or proliferation of MM cells in all experimental contexts tested. We conclude that it is unlikely that SGK3 plays a significant role for oncogenic signalling in multiple myeloma.
Collapse
Affiliation(s)
- Stefan Hausmann
- Department of Internal Medicine II, Division of Hematology and Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Evelyn Brandt
- Department of Internal Medicine II, Division of Hematology and Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Carolin Köchel
- Department of Internal Medicine II, Division of Hematology and Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, Division of Hematology and Oncology, University Hospital of Würzburg, Würzburg, Germany
| | - Ralf C. Bargou
- Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany
| | | | - Thorsten Stühmer
- Department of Internal Medicine II, Division of Hematology and Oncology, University Hospital of Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
40
|
Underwood TJ, Hayden AL, Derouet M, Garcia E, Noble F, White MJ, Thirdborough S, Mead A, Clemons N, Mellone M, Uzoho C, Primrose JN, Blaydes JP, Thomas GJ. Cancer-associated fibroblasts predict poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma. J Pathol 2015; 235:466-77. [PMID: 25345775 PMCID: PMC4312957 DOI: 10.1002/path.4467] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/06/2014] [Accepted: 10/16/2014] [Indexed: 12/15/2022]
Abstract
Interactions between cancer cells and cancer-associated fibroblasts (CAFs) play an important role in tumour development and progression. In this study we investigated the functional role of CAFs in oesophageal adenocarcinoma (EAC). We used immunochemistry to analyse a cohort of 183 EAC patients for CAF markers related to disease mortality. We characterized CAFs and normal oesophageal fibroblasts (NOFs) using western blotting, immunofluorescence and gel contraction. Transwell assays, 3D organotypic culture and xenograft models were used to examine the effects on EAC cell function and to dissect molecular mechanisms regulating invasion. Most EACs (93%) contained CAFs with a myofibroblastic (α-SMA-positive) phenotype, which correlated significantly with poor survival [p = 0.016; HR 7. 1 (1.7–29.4)]. Primary CAFs isolated from EACs have a contractile, myofibroblastic phenotype and promote EAC cell invasion in vitro (Transwell assays, p ≤ 0.05; organotypic culture, p < 0.001) and in vivo (p ≤ 0.05). In vitro, this pro-invasive effect is modulated through the matricellular protein periostin. Periostin is secreted by CAFs and acts as a ligand for EAC cell integrins αvβ3 and αvβ5, promoting activation of the PI3kinase–Akt pathway. In patient samples, periostin expression at the tumour cell–stromal interface correlates with poor overall and disease-free survival. Our study highlights the importance of the tumour stroma in EAC progression. Paracrine interaction between CAF-secreted periostin and EAC-expressed integrins results in PI3 kinase–Akt activation and increased tumour cell invasion. Most EACs contain a myofibroblastic CAF-rich stroma; this may explain the aggressive, highly infiltrative nature of the disease, and suggests that stromal targeting may produce therapeutic benefit in EAC patients.
Collapse
Affiliation(s)
- Timothy J Underwood
- Cancer Sciences Unit, Somers Cancer Research Building, University of Southampton, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Characterization of VPS34-IN1, a selective inhibitor of Vps34, reveals that the phosphatidylinositol 3-phosphate-binding SGK3 protein kinase is a downstream target of class III phosphoinositide 3-kinase. Biochem J 2014; 463:413-27. [PMID: 25177796 PMCID: PMC4209782 DOI: 10.1042/bj20140889] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The Vps34 (vacuolar protein sorting 34) class III PI3K (phosphoinositide 3-kinase) phosphorylates PtdIns (phosphatidylinositol) at endosomal membranes to generate PtdIns(3)P that regulates membrane trafficking processes via its ability to recruit a subset of proteins possessing PtdIns(3)P-binding PX (phox homology) and FYVE domains. In the present study, we describe a highly selective and potent inhibitor of Vps34, termed VPS34-IN1, that inhibits Vps34 with 25 nM IC50in vitro, but does not significantly inhibit the activity of 340 protein kinases or 25 lipid kinases tested that include all isoforms of class I as well as class II PI3Ks. Administration of VPS34-IN1 to cells induces a rapid dose-dependent dispersal of a specific PtdIns(3)P-binding probe from endosome membranes, within 1 min, without affecting the ability of class I PI3K to regulate Akt. Moreover, we explored whether SGK3 (serum- and glucocorticoid-regulated kinase-3), the only protein kinase known to interact specifically with PtdIns(3)P via its N-terminal PX domain, might be controlled by Vps34. Mutations disrupting PtdIns(3)P binding ablated SGK3 kinase activity by suppressing phosphorylation of the T-loop [PDK1 (phosphoinositide-dependent kinase 1) site] and hydrophobic motif (mammalian target of rapamycin site) residues. VPS34-IN1 induced a rapid ~50–60% loss of SGK3 phosphorylation within 1 min. VPS34-IN1 did not inhibit activity of the SGK2 isoform that does not possess a PtdIns(3)P-binding PX domain. Furthermore, class I PI3K inhibitors (GDC-0941 and BKM120) that do not inhibit Vps34 suppressed SGK3 activity by ~40%. Combining VPS34-IN1 and GDC-0941 reduced SGK3 activity ~80–90%. These data suggest SGK3 phosphorylation and hence activity is controlled by two pools of PtdIns(3)P. The first is produced through phosphorylation of PtdIns by Vps34 at the endosome. The second is due to the conversion of class I PI3K product, PtdIns(3,4,5)P3 into PtdIns(3)P, via the sequential actions of the PtdIns 5-phosphatases [SHIP1/2 (Src homology 2-domain-containing inositol phosphatase 1/2)] and PtdIns 4-phosphatase [INPP4B (inositol polyphosphate 4-phosphatase type II)]. VPS34-IN1 will be a useful probe to delineate physiological roles of the Vps34. Monitoring SGK3 phosphorylation and activity could be employed as a biomarker of Vps34 activity, in an analogous manner by which Akt is used to probe cellular class I PI3K activity. Combining class I (GDC-0941) and class III (VPS34-IN1) PI3K inhibitors could be used as a strategy to better analyse the roles and regulation of the elusive class II PI3K. We characterize VPS34-IN, a potent and selective inhibitor of class III Vps34 PI3K. Using VPS34-IN1, we demonstrate that PtdIns(3)P, produced by Vps34 controls phosphorylation and activity of the SGK3 protein kinase.
Collapse
|
42
|
Mizunuma M, Neumann‐Haefelin E, Moroz N, Li Y, Blackwell TK. mTORC2-SGK-1 acts in two environmentally responsive pathways with opposing effects on longevity. Aging Cell 2014; 13:869-78. [PMID: 25040785 PMCID: PMC4172656 DOI: 10.1111/acel.12248] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2014] [Indexed: 01/22/2023] Open
Abstract
The nematode worm Caenorhabditis elegans provides a powerful system for elucidating how genetic, metabolic, nutritional, and environmental factors influence aging. The mechanistic target of rapamycin (mTOR) kinase is important in growth, disease, and aging and is present in the mTORC1 and mTORC2 complexes. In diverse eukaryotes, lifespan can be increased by inhibition of mTORC1, which transduces anabolic signals to stimulate protein synthesis and inhibit autophagy. Less is understood about mTORC2, which affects C. elegans lifespan in a complex manner that is influenced by the bacterial food source. mTORC2 regulates C. elegans growth, reproduction, and lipid metabolism by activating the SGK-1 kinase, but current data on SGK-1 and lifespan seem to be conflicting. Here, by analyzing the mTORC2 component Rictor (RICT-1), we show that mTORC2 modulates longevity by activating SGK-1 in two pathways that affect lifespan oppositely. RICT-1/mTORC2 limits longevity by directing SGK-1 to inhibit the stress-response transcription factor SKN-1/Nrf in the intestine. Signals produced by the bacterial food source determine how this pathway affects SKN-1 and lifespan. In addition, RICT-1/mTORC2 functions in neurons in an SGK-1-mediated pathway that increases lifespan at lower temperatures. RICT-1/mTORC2 and SGK-1 therefore oppose or accelerate aging depending upon the context in which they are active. Our findings reconcile data on SGK-1 and aging, show that the bacterial microenvironment influences SKN-1/Nrf, mTORC2 functions, and aging, and identify two longevity-related mTORC2 functions that involve SGK-regulated responses to environmental cues.
Collapse
Affiliation(s)
- Masaki Mizunuma
- Joslin Diabetes Center Harvard Stem Cell Institute Harvard Medical School Department of Genetics Boston MA 02215USA
- Department of Molecular Biotechnology Graduate School of Advanced Sciences of Matter Hiroshima University Higashi‐Hiroshima 739‐8530 Japan
| | - Elke Neumann‐Haefelin
- Joslin Diabetes Center Harvard Stem Cell Institute Harvard Medical School Department of Genetics Boston MA 02215USA
- Renal Division University Hospital Freiburg Freiburg 79106Germany
| | - Natalie Moroz
- Joslin Diabetes Center Harvard Stem Cell Institute Harvard Medical School Department of Genetics Boston MA 02215USA
- Division of Biological Sciences Department of Genetics and Complex Diseases Harvard School of Public Health Boston MA 02115USA
| | - Yujie Li
- Renal Division University Hospital Freiburg Freiburg 79106Germany
| | - T. Keith Blackwell
- Joslin Diabetes Center Harvard Stem Cell Institute Harvard Medical School Department of Genetics Boston MA 02215USA
| |
Collapse
|
43
|
Paplomata E, O'Regan R. The PI3K/AKT/mTOR pathway in breast cancer: targets, trials and biomarkers. Ther Adv Med Oncol 2014; 6:154-66. [PMID: 25057302 DOI: 10.1177/1758834014530023] [Citation(s) in RCA: 397] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The phosphoinositide 3 kinase (PI3K)/Akt/mammalian (or mechanistic) target of rapamycin (mTOR) pathway is a complicated intracellular pathway, which leads to cell growth and tumor proliferation and plays a significant role in endocrine resistance in breast cancer. Multiple compounds targeting this pathway are being evaluated in clinical trials. These agents are generally well tolerated and can be used in combination with targeted therapies, endocrine therapy or cytotoxic agents. The identification of subtypes of tumors more likely to respond to these therapeutics cannot be overemphasized, since breast cancer is a very heterogeneous malignancy. Activation of pathways such as KRAS and MEK can act as escape mechanisms that lead to resistance, thus a combination of agents targeting multiple steps of the intracellular machinery is promising. There is evidence that tumors with PIK3CA mutations are more sensitive to inhibitors of the PI3K pathway but this has yet to be validated. Large clinical trials with correlative studies are necessary to identify reliable biomarkers of efficacy.
Collapse
Affiliation(s)
| | - Ruth O'Regan
- Winship Cancer Institute of Emory University, 1365C Clifton Road, Atlanta, GA 30329, USA
| |
Collapse
|
44
|
Wang Y, Zhou D, Chen S. SGK3 is an androgen-inducible kinase promoting prostate cancer cell proliferation through activation of p70 S6 kinase and up-regulation of cyclin D1. Mol Endocrinol 2014; 28:935-48. [PMID: 24739041 DOI: 10.1210/me.2013-1339] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Both androgen and phosphatidylinositol 3-kinase (PI3K) signaling are critical for cell proliferation of androgen receptor (AR)-positive prostate cancer cells, but the underlying mechanisms are still not fully understood. Here we report that serum- and glucocorticoid-inducible kinase 3 (SGK3), a Ser/Thr kinase functioning downstream of PI3K, is an AR transcriptional target and promotes prostate cancer cell proliferation. SGK3 expression is up-regulated by androgen DHT via AR. We identified an AR-binding region at the sgk3 locus, which confers androgen responsiveness of sgk3 promoters. Interestingly, we found that androgen/AR-dependent SGK3 expression requires estrogen receptor (ER) (including both isoforms, ERα and ERβ). Depletion of ER blocked DHT-induced SGK3 expression. Functionally, knockdown of SGK3 expression significantly decreased LNCaP prostate cancer cell proliferation by inhibiting G1 to S phase cell cycle progression. We further provided evidence that SGK3 promotes p70 S6 kinase (p70S6K) activation and increases cyclin D1 levels. In summary, our study identifies SGK3 as an AR target and provides a novel androgen-induced cell proliferation mechanism mediated by the AR-SGK3-p70S6K-cyclin D1 pathway in prostate cancer cells.
Collapse
Affiliation(s)
- Yuanzhong Wang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | | | | |
Collapse
|
45
|
Mori S, Nada S, Kimura H, Tajima S, Takahashi Y, Kitamura A, Oneyama C, Okada M. The mTOR pathway controls cell proliferation by regulating the FoxO3a transcription factor via SGK1 kinase. PLoS One 2014; 9:e88891. [PMID: 24558442 PMCID: PMC3928304 DOI: 10.1371/journal.pone.0088891] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 01/12/2014] [Indexed: 01/09/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) functions as a component of two large complexes, mTORC1 and mTORC2, which play crucial roles in regulating cell growth and homeostasis. However, the molecular mechanisms by which mTOR controls cell proliferation remain elusive. Here we show that the FoxO3a transcription factor is coordinately regulated by mTORC1 and mTORC2, and plays a crucial role in controlling cell proliferation. To dissect mTOR signaling, mTORC1 was specifically inactivated by depleting p18, an essential anchor of mTORC1 on lysosomes. mTORC1 inactivation caused a marked retardation of cell proliferation, which was associated with upregulation of cyclin-dependent kinase inhibitors (CDKIs). Although Akt was activated by mTORC1 inactivation, FoxO3a was upregulated via an epigenetic mechanism and hypophosphorylated at Ser314, which resulted in its nuclear accumulation. Consistently, mTORC1 inactivation induced downregulation of serum- and glucocorticoid-inducible kinase 1 (SGK1), the kinase responsible for Ser314 phosphorylation. Expression of FoxO3a mutated at Ser314 suppressed cell proliferation by inducing CDKI expression. SGK1 overexpression suppressed CDKI expression in p18-deficient cells, whereas SGK1 knockdown induced CDKI expression in wild-type cells, resulting in the suppression of cell proliferation. These results suggest that mTORC1, in coordination with mTORC2, controls cell proliferation by regulating FoxO3a gene expression and SGK1-mediated phosphorylation of FoxO3a at Ser314.
Collapse
Affiliation(s)
- Shunsuke Mori
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | - Shigeyuki Nada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | - Hironobu Kimura
- Laboratory of Epigenetics, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, Japan
| | - Shoji Tajima
- Laboratory of Epigenetics, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, Japan
| | - Yusuke Takahashi
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | - Ayaka Kitamura
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | - Chitose Oneyama
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
| | - Masato Okada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
- * E-mail:
| |
Collapse
|
46
|
Wang Y, Xu W, Zhou D, Neckers L, Chen S. Coordinated regulation of serum- and glucocorticoid-inducible kinase 3 by a C-terminal hydrophobic motif and Hsp90-Cdc37 chaperone complex. J Biol Chem 2013; 289:4815-26. [PMID: 24379398 DOI: 10.1074/jbc.m113.518480] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Serum- and glucocorticoid-inducible kinase 3 (SGK3) mediates a variety of cellular processes including membrane transport, cell proliferation, and survival, and it has been implicated in Akt-independent signaling downstream of oncogenic PIK3CA mutations (activating mutations in the α catalytic subunit of PI3K) in human cancers. However, the regulation of SGK3 is poorly understood. Here we report that SGK3 stability and kinase activation are regulated by the Hsp90-Cdc37 chaperone complex. Hsp90-Cdc37 associates with the kinase domain of SGK3 and acts in concert with a C-terminal hydrophobic motif of SGK3 to prevent Hsp70 association and ubiquitin ligase CHIP (C terminus of Hsc70-interacting protein)-mediated degradation. Phosphorylation of hydrophobic motif triggers release of Cdc37 and concomitant association of 3-phosphoinositide dependent kinase 1 (PDK1) to activate SGK3. Our study provides new insights into regulation of SGK3 stability and activation and the rationale for application of Hsp90 inhibitors in treating SGK3-dependent cancers.
Collapse
Affiliation(s)
- Yuanzhong Wang
- From the Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California 91010 and
| | | | | | | | | |
Collapse
|