1
|
Sharma G, Paganin M, Lauria F, Perenthaler E, Viero G. The SMN-ribosome interplay: a new opportunity for Spinal Muscular Atrophy therapies. Biochem Soc Trans 2024; 52:465-479. [PMID: 38391004 PMCID: PMC10903476 DOI: 10.1042/bst20231116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
The underlying cause of Spinal Muscular Atrophy (SMA) is in the reduction of survival motor neuron (SMN) protein levels due to mutations in the SMN1 gene. The specific effects of SMN protein loss and the resulting pathological alterations are not fully understood. Given the crucial roles of the SMN protein in snRNP biogenesis and its interactions with ribosomes and translation-related proteins and mRNAs, a decrease in SMN levels below a specific threshold in SMA is expected to affect translational control of gene expression. This review covers both direct and indirect SMN interactions across various translation-related cellular compartments and processes, spanning from ribosome biogenesis to local translation and beyond. Additionally, it aims to outline deficiencies and alterations in translation observed in SMA models and patients, while also discussing the implications of the relationship between SMN protein and the translation machinery within the context of current and future therapies.
Collapse
|
2
|
Giordano F, Comità S, Venneri G, Rago V, Naimo GD, De Amicis F, De Bartolo A, Tundis R, Mauro L, Panno ML. Poncirus trifoliata (L.) Raf. Seed Extract Induces Cell Cycle Arrest and Apoptosis in the Androgen Receptor Positive LNCaP Prostate Cancer Cells. Int J Mol Sci 2023; 24:16351. [PMID: 38003541 PMCID: PMC10671002 DOI: 10.3390/ijms242216351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Prostate cancer (PCa) is the second most common male cancer. Its incidence derives from the interaction between modifiable and non-modifiable factors. The progression of prostate cancer into a more aggressive phenotype is associated with chronic inflammation and increased ROS production. For their biological properties, some phytochemicals from fruits and vegetable emerge as a promise strategy for cancer progression delay. These bioactive compounds are found in the highest amounts in peels and seeds. Poncirus trifoliata (L.) Raf. (PT) has been widely used in traditional medicine and retains anti-inflammatory, anti-bacterial, and anticancer effects. The seeds of P. trifoliata were exhaustively extracted by maceration with methanol as the solvent. The cell proliferation rate was performed by MTT and flow cytometry, while the apoptosis signals were analyzed by Western blotting and TUNEL assay. P. trifoliata seed extract reduced LNCaP and PC3 cell viability and induced cell cycle arrest at the G0/G1phase and apoptosis. In addition, a reduction in the AKT/mTOR pathway has been observed together with the up-regulation of stress-activated MAPK (p38 and c-Jun N-terminal kinase). Based on the study, the anti-growth effects of PT seed extract on prostate tumor cells give indications on the potential of the phytochemical drug for the treatment of this type of cancer. However, future in-depth studies are necessary to identify which components are mainly responsible for the anti-neoplastic response.
Collapse
Affiliation(s)
- Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Stefano Comità
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Giulia Venneri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Anna De Bartolo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy;
| | - Rosa Tundis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Loredana Mauro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (S.C.); (G.V.); (V.R.); (G.D.N.); (F.D.A.); (R.T.); (M.L.P.)
| |
Collapse
|
3
|
Islam M, Jones S, Ellis I. Role of Akt/Protein Kinase B in Cancer Metastasis. Biomedicines 2023; 11:3001. [PMID: 38002001 PMCID: PMC10669635 DOI: 10.3390/biomedicines11113001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Metastasis is a critical step in the process of carcinogenesis and a vast majority of cancer-related mortalities result from metastatic disease that is resistant to current therapies. Cell migration and invasion are the first steps of the metastasis process, which mainly occurs by two important biological mechanisms, i.e., cytoskeletal remodelling and epithelial to mesenchymal transition (EMT). Akt (also known as protein kinase B) is a central signalling molecule of the PI3K-Akt signalling pathway. Aberrant activation of this pathway has been identified in a wide range of cancers. Several studies have revealed that Akt actively engages with the migratory process in motile cells, including metastatic cancer cells. The downstream signalling mechanism of Akt in cell migration depends upon the tumour type, sites, and intracellular localisation of activated Akt. In this review, we focus on the role of Akt in the regulation of two events that control cell migration and invasion in various cancers including head and neck squamous cell carcinoma (HNSCC) and the status of PI3K-Akt pathway inhibitors in clinical trials in metastatic cancers.
Collapse
Affiliation(s)
- Mohammad Islam
- Unit of Cell and Molecular Biology, School of Dentistry, University of Dundee, Park Place, Dundee DD1 4HR, UK; (S.J.); (I.E.)
| | | | | |
Collapse
|
4
|
Riquelme I, Pérez-Moreno P, Mora-Lagos B, Ili C, Brebi P, Roa JC. Long Non-Coding RNAs (lncRNAs) as Regulators of the PI3K/AKT/mTOR Pathway in Gastric Carcinoma. Int J Mol Sci 2023; 24:ijms24076294. [PMID: 37047267 PMCID: PMC10094576 DOI: 10.3390/ijms24076294] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Gastric cancer (GC) represents ~10% of the global cancer-related deaths, increasingly affecting the younger population in active stages of life. The high mortality of GC is due to late diagnosis, the presence of metastasis and drug resistance development. Additionally, current clinical markers do not guide the patient management adequately, thereby new and more reliable biomarkers and therapeutic targets are still needed for this disease. RNA-seq technology has allowed the discovery of new types of RNA transcripts including long non-coding RNAs (lncRNAs), which are able to regulate the gene/protein expression of many signaling pathways (e.g., the PI3K/AKT/mTOR pathway) in cancer cells by diverse molecular mechanisms. In addition, these lncRNAs might also be proposed as promising diagnostic or prognostic biomarkers or as potential therapeutic targets in GC. This review describes important topics about some lncRNAs that have been described as regulators of the PI3K/AKT/mTOR signaling pathway, and hence, their potential oncogenic role in the development of this malignancy.
Collapse
Affiliation(s)
- Ismael Riquelme
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Temuco 4810101, Chile
- Correspondence: (I.R.); (J.C.R.); Tel.: +56-95923-6933 (I.R.); +56-22354-1061 (J.C.R.)
| | - Pablo Pérez-Moreno
- Millennium Institute on Immunology and Immunotherapy (MIII), Center for Cancer Prevention and Control (CECAN), Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380000, Chile
| | - Bárbara Mora-Lagos
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Temuco 4810101, Chile
| | - Carmen Ili
- Millennium Institute on Immunology and Immunotherapy (MIII), Laboratory of Integrative Biology (LIBi), Center for Excellence in Translational Medicine—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile
| | - Priscilla Brebi
- Millennium Institute on Immunology and Immunotherapy (MIII), Laboratory of Integrative Biology (LIBi), Center for Excellence in Translational Medicine—Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco 4810296, Chile
| | - Juan Carlos Roa
- Millennium Institute on Immunology and Immunotherapy (MIII), Center for Cancer Prevention and Control (CECAN), Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380000, Chile
- Correspondence: (I.R.); (J.C.R.); Tel.: +56-95923-6933 (I.R.); +56-22354-1061 (J.C.R.)
| |
Collapse
|
5
|
Manai F, Zanoletti L, Morra G, Mansoor S, Carriero F, Bozzola E, Muscianisi S, Comincini S. Gluten Exorphins Promote Cell Proliferation through the Activation of Mitogenic and Pro-Survival Pathways. Int J Mol Sci 2023; 24:3912. [PMID: 36835317 PMCID: PMC9966116 DOI: 10.3390/ijms24043912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Celiac disease (CD) is a chronic and systemic autoimmune disorder that affects preferentially the small intestine of individuals with a genetic predisposition. CD is promoted by the ingestion of gluten, a storage protein contained in the endosperm of the seeds of wheat, barley, rye, and related cereals. Once in the gastrointestinal (GI) tract, gluten is enzymatically digested with the consequent release of immunomodulatory and cytotoxic peptides, i.e., 33mer and p31-43. In the late 1970s a new group of biologically active peptides, called gluten exorphins (GEs), was discovered and characterized. In particular, these short peptides showed a morphine-like activity and high affinity for the δ-opioid receptor (DOR). The relevance of GEs in the pathogenesis of CD is still unknown. Recently, it has been proposed that GEs could contribute to asymptomatic CD, which is characterized by the absence of symptoms that are typical of this disorder. In the present work, GEs cellular and molecular effects were in vitro investigated in SUP-T1 and Caco-2 cells, also comparing viability effects with human normal primary lymphocytes. As a result, GEs treatments increased tumor cell proliferation by cell cycle and Cyclins activation as well as by induction of mitogenic and pro-survival pathways. Finally, a computational model of GEs interaction with DOR is provided. Altogether, the results might suggest a possible role of GEs in CD pathogenesis and on its associated cancer comorbidities.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
- Laboratory for Mucosal Immunology, TARGID, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Giulia Morra
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Samman Mansoor
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
| | - Francesca Carriero
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Elena Bozzola
- Pediatric Unit, I.R.C.C.S. Bambino Gesù Children Hospital, 00165 Roma, Italy
| | - Stella Muscianisi
- Cell Factory and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
6
|
Sahu R, Jha S, Pattanayak SP. Therapeutic silencing of mTOR by systemically administered siRNA-loaded neutral liposomal nanoparticles inhibits DMBA-induced mammary carcinogenesis. Br J Cancer 2022; 127:2207-2219. [PMID: 36261586 PMCID: PMC9726943 DOI: 10.1038/s41416-022-02011-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Mammary carcinogenesis possesses great challenges due to the lack of effectiveness of the multiple therapeutic options available. Gene therapy-based cancer treatment strategy provides more targeting accuracy, fewer side effects, and higher therapeutic efficiency. Downregulation of the oncogene mTOR by mTOR-siRNA is an encouraging approach to reduce cancer progression. However, its employment as means of therapeutic strategy has been restricted due to the unavailability of a suitable delivery system. METHODS A suitable nanocarrier system made up of 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) has been developed to prevent degradation and for proficient delivery of siRNA. This was followed by in vitro and in vivo anti-breast cancer efficiency analysis of the mTOR siRNA-loaded neutral liposomal formulation (NL-mTOR-siRNA). RESULTS In our experiment, a profound reduction in MCF-7 cell growth, proliferation and invasion was ascertained following extensive downregulation of mTOR expression. NL-mTOR-siRNA suppressed tumour growth and restored morphological alterations of DMBA-induced breast cancer. In addition, neutral liposome enhanced accumulation of siRNA in mammary cancer tissues facilitating its deep cytosolic distribution within the tumour, which allows apoptosis thereby facilitating its anti-tumour potential. CONCLUSION Hence, the current study highlighted the augmented ground for therapies aiming toward cancerous cells to diminish mTOR expression by RNAi in managing mammary carcinoma.
Collapse
Affiliation(s)
- Roja Sahu
- Division of Advanced Pharmacology, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology (BIT), Mesra, Ranchi, Jharkhand, 835 215, India
| | - Shivesh Jha
- Division of Pharmacognosy and Phytochemistry, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology (BIT), Mesra, Ranchi, Jharkhand, 835 215, India
| | - Shakti Prasad Pattanayak
- Department of Pharmacy, School of Health Science, Central University of South Bihar (Gaya), Gaya, Bihar, 824 236, India.
| |
Collapse
|
7
|
Autophagy Mediates MMP-2 Expression in Glaucomatous Trabecular Meshwork Cells. J Ophthalmol 2022; 2022:6026464. [PMID: 36211598 PMCID: PMC9536984 DOI: 10.1155/2022/6026464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the effect of 3-methyladenine (3-MA) and starvation on the expression of matrix metalloproteinase (MMP-2) in patients with primary open-angle glaucoma. Methods Primary TM cells were cultured and divided into three groups. The control group was treated with a normal medium, the 3-MA group was stimulated with 3-MA, and the starvation group received nutrient depletion by replacing the normal media with Earle's balanced salt solution. Cellular mRNA and protein were measured at different 3-MA concentrations and starvation time periods. The level of autophagy was accessed by monodansylcadaverine fluorescent staining and expression of specific autophagy-related genes, light chain 3 (LC3), and Beclin1. The effects of 3-MA and starvation on cell proliferation were determined with a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay kit. The mRNA and protein expression of LC3-II, Beclin1, and MMP-2 were measured by reverse transcription-polymerase chain reaction and western blot, respectively. Results Compared to the control group, starvation significantly upregulated LC3-II and Beclin1 in TM cells after 3 h of stimulation, which peaked at 6 h and 9 h, respectively. Increased MDC-labeled cells were also observed. Starvation downregulated the expression of MMP-2. On the contrary, 3-MA suppressed the activation of autophagy, as shown by the marked downregulation of LC3-II and Beclin1. The expressions of MMP-2 were higher in the 3-MA group compared to the control group, reaching a peak at a concentration of 5 mM. Conclusion Autophagy may be involved in the pathogenesis of POAG via regulating the expression of MMP-2 and, subsequently, the deposition of the extracellular matrix.
Collapse
|
8
|
Joechle K, Jumaa H, Thriene K, Hellerbrand C, Kulemann B, Fichtner-Feigl S, Lang SA, Guenzle J. Dual Inhibition of mTORC1/2 Reduces Migration of Cholangiocarcinoma Cells by Regulation of Matrixmetalloproteinases. Front Cell Dev Biol 2022; 9:785979. [PMID: 35096817 PMCID: PMC8793831 DOI: 10.3389/fcell.2021.785979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/17/2021] [Indexed: 12/30/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a rare but highly aggressive tumor entity for which systemic therapies only showed limited efficacy so far. As OSI-027—a dual kinase inhibitor targeting both mTOR complexes, mTORC1 and mTORC2 - showed improved anti-cancer effects, we sought to evaluate its impact on the migratory and metastatic capacity of CCA cells in vitro. We found that treatment with OSI-027 leads to reduced cell mobility and migration as well as a reduced surviving fraction in colony-forming ability. While neither cell viability nor proliferation rate was affected, OSI-027 decreased the expression of MMP2 and MMP9. Moreover, survival as well as anti-apoptotic signaling was impaired upon the use of OSI-027 as determined by AKT and MAPK blotting. Dual targeting of mTORC1/2 might therefore be a viable option for anti-neoplastic therapy in CCA.
Collapse
Affiliation(s)
- Katharina Joechle
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany.,Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Huda Jumaa
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Kerstin Thriene
- Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Birte Kulemann
- Department of Surgery, University Medical Center Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany.,Comprehensive Cancer Center Freiburg-CCCF, Medical Center-University, Freiburg, Germany
| | - Sven A Lang
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany.,Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Jessica Guenzle
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
9
|
EBV LMP1-activated mTORC1 and mTORC2 Coordinately Promote Nasopharyngeal Cancer Stem Cell Properties. J Virol 2022; 96:e0194121. [PMID: 35019715 DOI: 10.1128/jvi.01941-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epstein-Barr Virus (EBV) is associated with several malignant diseases, including Burkitt's lymphoma, nasopharyngeal carcinoma (NPC), certain types of lymphomas, and a portion of gastric cancers. Virus-encoded oncoprotein LMP1 induces the epithelial-to-mesenchymal transition (EMT), leading to cancer stem cell formation. In the current study, we investigated how LMP1 contributes to cancer stem cell development in NPC. We found that LMP1 plays an essential role in acquiring CSC characteristics, including tumor initiation, metastasis, and therapeutic resistance by activating the PI3K/mTOR/Akt signaling pathway. We dissected the functions of distinct signaling (mTORC1 and mTORC2) in the acquisition of different CSC characteristics. Side population (SP) formation, which represents the chemotherapy resistance feature of CSC, requires mTORC1 signaling. Tumor initiation capability is mainly attributed to mTORC2, which confers on NPC the capabilities of proliferation and survival by activating mTORC2 downstream genes c-Myc. Both mTORC1 and mTORC2 enhance cell migration and invasion of NPC cells, suggesting that mTORC1/2 co-regulate metastasis of NPC. The revelation of the roles of the mTOR signaling pathways in distinct tumorigenic features provides a guideline for designing efficient therapies by choosing specific mTOR inhibitors targeting mTORC1, mTORC2, or both to achieve durable remission of NPC in patients. Importance LMP1 endows NPC to gain cancer stem cell characteristics through activating mTORC1 and mTORC2 pathways. The different mTOR pathways are responsible for distinct tumorigenic features. Rapamycin-insensitive mTORC1 is essential for CSC drug resistance. NPC tumor initiation capacity is mainly attributed to mTORC2 signaling. mTORC1 and mTORC2 co-regulate NPC cell migration and invasion. The revelation of the roles of mTOR signaling in NPC CSC establishment has implications for novel therapeutic strategies to treat relapsed and metastatic NPC and achieve durable remission.
Collapse
|
10
|
Ramchandani D, Berisa M, Tavarez DA, Li Z, Miele M, Bai Y, Lee SB, Ban Y, Dephoure N, Hendrickson RC, Cloonan SM, Gao D, Cross JR, Vahdat LT, Mittal V. Copper depletion modulates mitochondrial oxidative phosphorylation to impair triple negative breast cancer metastasis. Nat Commun 2021; 12:7311. [PMID: 34911956 PMCID: PMC8674260 DOI: 10.1038/s41467-021-27559-z] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/05/2021] [Indexed: 12/26/2022] Open
Abstract
Copper serves as a co-factor for a host of metalloenzymes that contribute to malignant progression. The orally bioavailable copper chelating agent tetrathiomolybdate (TM) has been associated with a significant survival benefit in high-risk triple negative breast cancer (TNBC) patients. Despite these promising data, the mechanisms by which copper depletion impacts metastasis are poorly understood and this remains a major barrier to advancing TM to a randomized phase II trial. Here, using two independent TNBC models, we report a discrete subpopulation of highly metastatic SOX2/OCT4+ cells within primary tumors that exhibit elevated intracellular copper levels and a marked sensitivity to TM. Global proteomic and metabolomic profiling identifies TM-mediated inactivation of Complex IV as the primary metabolic defect in the SOX2/OCT4+ cell population. We also identify AMPK/mTORC1 energy sensor as an important downstream pathway and show that AMPK inhibition rescues TM-mediated loss of invasion. Furthermore, loss of the mitochondria-specific copper chaperone, COX17, restricts copper deficiency to mitochondria and phenocopies TM-mediated alterations. These findings identify a copper-metabolism-metastasis axis with potential to enrich patient populations in next-generation therapeutic trials.
Collapse
Affiliation(s)
- Divya Ramchandani
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mirela Berisa
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Diamile A Tavarez
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhuoning Li
- Department of Microchemistry and Proteomics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Matthew Miele
- Department of Microchemistry and Proteomics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yang Bai
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sharrell B Lee
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Yi Ban
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Noah Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ronald C Hendrickson
- Department of Microchemistry and Proteomics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Suzanne M Cloonan
- Department of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- The School of Medicine and Tallaght University Hospital, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Dingcheng Gao
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Cell and Developmental biology, Weill Cornell Medicine, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Linda T Vahdat
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA.
- Department of Cell and Developmental biology, Weill Cornell Medicine, New York, NY, 10065, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
11
|
Ma H, Liu C, Zhang S, Yuan W, Hu J, Huang D, Zhang X, Liu Y, Qiu Y. miR-328-3p promotes migration and invasion by targeting H2AFX in head and neck squamous cell carcinoma. J Cancer 2021; 12:6519-6530. [PMID: 34659543 PMCID: PMC8489127 DOI: 10.7150/jca.60743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Migration and invasion are the initial step in the metastatic process, while metastasis is responsible for the poor prognosis of head and neck squamous cell carcinoma (HNSCC). Since miRNA has been found as an important regulator of gene expression at the post-transcriptional level in various diseases including carcinoma, exploring the role of miRNA in cancer metastasis will facilitate the target therapy of advanced HNSCC. MiR-328-3p has been reported to be an onco-miRNA or a tumor suppressor in several cancers. However, the role of miR-328-3p in HNSCC migration and invasion remains undefined. In this study, we first demonstrated that miR-328-3p enhanced migration and invasion of HNSCC in vitro, accompanying with a promotion of epithelial-mesenchymal transition (EMT) and mTOR activity. Meanwhile, we confirmed that miR-328-3p directly targeted the 3'UTR of H2A histone family, member X (H2AFX), which served as a tumor suppressor in migration and invasion of HNSCC. Moreover, H2AFX could partially reverse the migration and invasion of HNSCC caused by miR-328-3p. Overall, our results indicated that miR-328-3p enhanced migration and invasion of HNSCC through targeting H2AFX and activated the mTOR pathway.
Collapse
Affiliation(s)
- Huiling Ma
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China
| | - Chao Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Shuiting Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Wenhui Yuan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Junli Hu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Donghai Huang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, China.,Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Rispoli MG, Valentinuzzi S, De Luca G, Del Boccio P, Federici L, Di Ioia M, Digiovanni A, Grasso EA, Pozzilli V, Villani A, Chiarelli AM, Onofrj M, Wise RG, Pieragostino D, Tomassini V. Contribution of Metabolomics to Multiple Sclerosis Diagnosis, Prognosis and Treatment. Int J Mol Sci 2021; 22:11112. [PMID: 34681773 PMCID: PMC8541167 DOI: 10.3390/ijms222011112] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Metabolomics-based technologies map in vivo biochemical changes that may be used as early indicators of pathological abnormalities prior to the development of clinical symptoms in neurological conditions. Metabolomics may also reveal biochemical pathways implicated in tissue dysfunction and damage and thus assist in the development of novel targeted therapeutics for neuroinflammation and neurodegeneration. Metabolomics holds promise as a non-invasive, high-throughput and cost-effective tool for early diagnosis, follow-up and monitoring of treatment response in multiple sclerosis (MS), in combination with clinical and imaging measures. In this review, we offer evidence in support of the potential of metabolomics as a biomarker and drug discovery tool in MS. We also use pathway analysis of metabolites that are described as potential biomarkers in the literature of MS biofluids to identify the most promising molecules and upstream regulators, and show novel, still unexplored metabolic pathways, whose investigation may open novel avenues of research.
Collapse
Affiliation(s)
- Marianna Gabriella Rispoli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Silvia Valentinuzzi
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giovanna De Luca
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Piero Del Boccio
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Luca Federici
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Maria Di Ioia
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Anna Digiovanni
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Eleonora Agata Grasso
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Valeria Pozzilli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Alessandro Villani
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Antonio Maria Chiarelli
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Marco Onofrj
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| | - Richard G. Wise
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
| | - Damiana Pieragostino
- Analytical Biochemistry and Proteomics Research Unit, Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (S.V.); (P.D.B.); (L.F.)
- Department of Paediatrics, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Valentina Tomassini
- Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.G.R.); (A.D.); (V.P.); (A.V.); (A.M.C.); (M.O.); (R.G.W.)
- Department of Neurology, “SS. Annunziata” University Hospital, 66100 Chieti, Italy; (G.D.L.); (M.D.I.)
| |
Collapse
|
13
|
Liu X, Wang Y, He Y, Wang X, Zhang R, Bachhuka A, Madathiparambil Visalakshan R, Feng Q, Vasilev K. Synergistic Effect of Surface Chemistry and Surface Topography Gradient on Osteogenic/Adipogenic Differentiation of hMSCs. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30306-30316. [PMID: 34156811 DOI: 10.1021/acsami.1c03915] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Much attention has been paid to understanding the individual effects of surface chemistry or topography on cell behavior. However, the synergistic influence of both surface chemistry and surface topography on differentiation of human mesenchymal stem cells (hMSCs) should also be addressed. Here, gold nanoparticles were immobilized in an increasing number density manner to achieve a surface topography gradient; a thin film rich in amine (-NH2) or methyl (-CH3) chemical groups was plasma-polymerized to adjust the surface chemistry of the outermost layer (ppAA and ppOD, respectively). hMSCs were cultured on these model substrates with defined surface chemistry and surface topography gradient. The morphology and focal adhesion (FA) formation of hMSCs were first examined. hMSC differentiation was then co-induced in osteogenic and adipogenic medium, as well as in the presence of extracellular-signal-regulated kinase1/2 (ERK1/2) and RhoA/Rho-associated protein kinase (ROCK) inhibitors. The results show that the introduction of nanotopography could enhance FA formation and osteogenesis but inhibited adipogenesis on both ppAA and ppOD surfaces, indicating that the surface chemistry could regulate hMSC differentiation, in a surface topography-dependent manner. RhoA/ROCK and ERK1/2 signaling pathways may participate in this process. This study demonstrated that surface chemistry and surface topography can jointly affect cell morphology, FA formation, and thus osteogenic/adipogenic differentiation of hMSCs. These findings highlight the importance of the synergistic effect of different material properties on regulation of cell response, which has important implications in designing functional biomaterials.
Collapse
Affiliation(s)
- Xujie Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yakun Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaofeng Wang
- Department of Hand Surgery, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315040, China
| | - Ranran Zhang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Akash Bachhuka
- Unit of STEM, University of South Australia, Mawson Lakes 5095, Australia
| | | | - Qingling Feng
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Krasimir Vasilev
- Unit of STEM, University of South Australia, Mawson Lakes 5095, Australia
| |
Collapse
|
14
|
Xie S, Wang X, Gan S, Tang X, Kang X, Zhu S. The Mitochondrial Chaperone TRAP1 as a Candidate Target of Oncotherapy. Front Oncol 2021; 10:585047. [PMID: 33575209 PMCID: PMC7870996 DOI: 10.3389/fonc.2020.585047] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor necrosis factor receptor-associated protein 1 (TRAP1), a member of the heat shock protein 90 (Hsp90) chaperone family, protects cells against oxidative stress and maintains mitochondrial integrity. To date, numerous studies have focused on understanding the relationship between aberrant TRAP1 expression and tumorigenesis. Mitochondrial TRAP1 is a key regulatory factor involved in metabolic reprogramming in tumor cells that favors the metabolic switch of tumor cells toward the Warburg phenotype. In addition, TRAP1 is involved in dual regulation of the mitochondrial apoptotic pathway and exerts an antiapoptotic effect on tumor cells. Furthermore, TRAP1 is involved in many cellular pathways by disrupting the cell cycle, increasing cell motility, and promoting tumor cell invasion and metastasis. Thus, TRAP1 is a very important therapeutic target, and treatment with TRAP1 inhibitors combined with chemotherapeutic agents may become a new therapeutic strategy for cancer. This review discusses the molecular mechanisms by which TRAP1 regulates tumor progression, considers its role in apoptosis, and summarizes recent advances in the development of selective, targeted TRAP1 and Hsp90 inhibitors.
Collapse
Affiliation(s)
- Shulan Xie
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuanwei Wang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuyuan Gan
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaodong Tang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
New insights into molecular chaperone TRAP1 as a feasible target for future cancer treatments. Life Sci 2020; 254:117737. [PMID: 32376268 DOI: 10.1016/j.lfs.2020.117737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
Tumor necrosis factor receptor-associated protein 1 (TRAP1), a molecular chaperone, is a major member of the mitochondrial heat shock protein 90 (Hsp90) family. Studies have shown that TRAP1 can prevent hypoxia-induced damage to cardiomyocytes, maintain cardiomyocytes viability and mitochondrial membrane potential, and protect cardiomyocytes. In addition, it can also protect astrocytes from ischemic damage in vitro. In recent years, there have been many new discoveries in tumors. The abnormal expression of TRAP1 is closely related to the occurrence and development of various tumors. TRAP1 protein seems to be a central regulatory protein, involved in the activation of various oncogenic proteins and signaling pathways, and has a balanced function at tumor transformation and the intersection of different metabolic processes. Targeting its chaperone activity and molecular interactions can destroy the metabolism and survival adaptability of tumor cells, paving the way for the development of highly selective mitochondrial anti-tumor drugs. Moreover, the combination of TRAP1 inhibition and current traditional cancer therapies has shown promising applications. These findings have important implications for the diagnosis and treatment of tumors. Therefore, we reviewed the recently identified functions of the molecular chaperone TRAP1 in cancer development and progression, as well as the discovery and recent advances in selective TRAP1 inhibitors as anticancer drug therapies, opening up new attractive prospects for exploring strategies for targeting TRAP1 as a tumor cell target.
Collapse
|
16
|
Zhang N, Ma S. Research Progress of 70 kDa Ribosomal Protein S6 Kinase (P70S6K) Inhibitors as Effective Therapeutic Tools for Obesity, Type II Diabetes and Cancer. Curr Med Chem 2020; 27:4699-4719. [PMID: 31942845 DOI: 10.2174/0929867327666200114113139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/06/2019] [Accepted: 01/05/2020] [Indexed: 01/07/2023]
Abstract
At present, diseases such as obesity, type Ⅱ diabetes and cancer have brought serious health problems, which are closely related to mTOR pathway. 70 kDa ribosomal protein S6 kinase (p70S6K), as a significant downstream effector of mTOR, mediates protein synthesis, RNA processing, glucose homeostasis, cell growth and apoptosis. Inhibiting the function of p70S6K can reduce the risk of obesity which helps to treat dyslipidemia, enhance insulin sensitivity, and extend the life span of mammals. Therefore, p70S6K has become a potential target for the treatment of these diseases. So far, except for the first p70S6K specific inhibitor PF-4708671 developed by Pfizer and LY2584702 developed by Lilai, all of them are in preclinical research. This paper briefly introduces the general situation of p70S6K and reviews their inhibitors in recent years, which are mainly classified into two categories: natural compounds and synthetic compounds. In particular, their inhibitory activities, structure-activity relationships (SARs) and mechanisms are highlighted.
Collapse
Affiliation(s)
- Na Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education) School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Shutao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education) School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| |
Collapse
|
17
|
Noble AR, Hogg K, Suman R, Berney DM, Bourgoin S, Maitland NJ, Rumsby MG. Phospholipase D2 in prostate cancer: protein expression changes with Gleason score. Br J Cancer 2019; 121:1016-1026. [PMID: 31673104 PMCID: PMC6964697 DOI: 10.1038/s41416-019-0610-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Phospholipases D1 and D2 (PLD1/2) are implicated in tumorigenesis through their generation of the signalling lipid phosphatidic acid and its downstream effects. Inhibition of PLD1 blocks prostate cell growth and colony formation. Here a role for PLD2 in prostate cancer (PCa), the major cancer of men in the western world, is examined. METHODS PLD2 expression was analysed by immunohistochemistry and western blotting. The effects of PLD2 inhibition on PCa cell viability and cell motility were measured using MTS, colony forming and wound-healing assays. RESULTS PLD2 protein is expressed about equally in luminal and basal prostate epithelial cells. In cells from different Gleason-scored PCa tissue PLD2 protein expression is generally higher than in non-tumorigenic cells and increases in PCa tissue scored Gleason 6-8. PLD2 protein is detected in the cytosol and nucleus and had a punctate appearance. In BPH tissue stromal cells as well as basal and luminal cells express PLD2. PLD2 protein co-expresses with chromogranin A in castrate-resistant PCa tissue. PLD2 inhibition reduces PCa cell viability, colony forming ability and directional cell movement. CONCLUSIONS PLD2 expression correlates with increasing Gleason score to GS8. PLD2 inhibition has the potential to reduce PCa progression.
Collapse
Affiliation(s)
- Amanda R Noble
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK
| | - Karen Hogg
- Technology Facility, Department of Biology, University of York, York, YO10 5DD, UK
| | - Rakesh Suman
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK
| | - Daniel M Berney
- Department of Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Sylvain Bourgoin
- Centre de Recherche du CHU de Québec, Axe des Maladies Infectieuses et Immunitaires, local T1-58, 2705 boulevard Laurier, Québec, G1V 4G2, QC, Canada
| | - Norman J Maitland
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK
| | - Martin G Rumsby
- Cancer Research Unit, Department of Biology, University of York, York, YO10 5DD, UK.
| |
Collapse
|
18
|
Down-regulation of STIP1 regulate apoptosis and invasion of glioma cells via TRAP1/AKT signaling pathway. Cancer Genet 2019; 237:1-9. [PMID: 31447061 DOI: 10.1016/j.cancergen.2019.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 04/15/2019] [Accepted: 05/29/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND In recent years, many studies have confirmed that STIP1 (phosphorylation-induced protein 1) is involved in the development and progression of various tumors. However, its potential role in glioma progression and the underlying mechanisms of glioma development remain unclear. METHODS We analyzed the expression of STIP1 in 35 human glioma tissue specimens of different grades, using 6 normal brain tissues for comparison. We transfected U87 and U251 cell lines with small interfering RNA (siRNA) to downregulate STIP1, and set up a negative control group and a blank group for comparison. The MTT assay was used to detect cell proliferation, and cell cycle progression and apoptosis were analyzed through flow cytometry. Transwell experiments were employed to detect the invasion and migration of STIP1-depleted and control U87 and U251 cells and western blotting was used to detect the expression of TRAP1/Akt pathway proteins. In addition, immunohistochemical analysis was used to reveal differences in expression and localization between transplanted tumor specimens of each group. RESULTS We observed a high expression of STIP1 in glioblastoma, MTT assay revealed a decreased cell proliferation rate in the STIP1-downregulated cells. Cell cycle analysis revealed an increased proportion of cells in G1 phase, as well as an increase in apoptosis, upon STIP1 downregulation. Western blotting showed that TRAP1, pAkt, and MMP2 expression was decreased upon STIP1 downregulation. In addition, TRAP1, ki-67, and MMP2 displayed a decreased expression in vivo. CONCLUSIONS STIP1 is highly expressed in glioblastoma compared to normal brain tissues. Downregulation of STIP1 in glioma cells reduces cell proliferation rate and invasion and increases cell apoptosis.
Collapse
|
19
|
Chen Z, Wei H, Zhao X, Xin X, Peng L, Ning Y, Wang Y, Lan Y, Zhang Q. Metformin treatment alleviates polycystic ovary syndrome by decreasing the expression of MMP‐2 and MMP‐9 via H19/miR‐29b‐3p and AKT/mTOR/autophagy signaling pathways. J Cell Physiol 2019; 234:19964-19976. [PMID: 30989649 DOI: 10.1002/jcp.28594] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Zhilan Chen
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Huafang Wei
- Department of Obstetrics and Gynecology Central Theater General Hospital of PLA Wuhan China
| | - Xiaoling Zhao
- Department of Obstetrics and Gynecology Central Theater General Hospital of PLA Wuhan China
| | - Xin Xin
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Ling Peng
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yang Ning
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yapei Wang
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yanli Lan
- Department of Obstetrics and Gynecology Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| | - Qinghua Zhang
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
20
|
Zhai Y, Bai J, Wang S, Gao H, Li M, Li C, Gui S, Zhang Y. Analysis of clinical factors and PDGFR-β in predicting prognosis of patients with clival chordoma. J Neurosurg 2018; 129:1429-1437. [PMID: 29303447 DOI: 10.3171/2017.6.jns17562] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 06/05/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVEIn this study, the authors' aim was to research clinical features and prognostic factors in patients harboring clival chordomas and explore the relationship between platelet-derived growth factor receptor-β (PDGFR-β) expression and tumor invasion and prognosis of clival chordoma.METHODSA total of 242 patients were retrospectively analyzed. Clinical information, including extent of resection, Al-Mefty classification, postoperative complications, and postoperative radiotherapy, was reviewed. Kaplan-Meier analysis was used to estimate survival time. Immunohistochemical analysis, quantitative reverse transcription polymerase chain reaction, and Western blotting were used to measure the expression level of proteins or mRNA. Transwell assaying was performed to measure the invasive ability of the tumor cells.RESULTSAccording to the Al-Mefty classification, there were 37, 112, and 93 type I, II, and III tumors, respectively. Gross-total resection (GTR) was achieved in 86 cases (35.5%), subtotal resection (STR) in 63 cases (26.0%), and partial resection (PR) in 93 cases (38.4%). The 5-year progression-free survival (PFS) and overall survival (OS) rates in the GTR group were significantly higher than those in the non-total resection (NTR; i.e., STR and PR) group (p < 0.001). The 5-year PFS and OS rates for patients with type I tumors were significantly higher than those for patients harboring types II and III tumors (p < 0.001). In the NTR group, the median PFS and OS of patients with lower PDGFR-β expression were significantly longer than those of patients with higher PDGFR-β expression. Reduction of PDGFR-β suppressed the invasion ability of cells in vitro. In addition, reduction of PDGFR-β can obviously downregulate the expression levels of mammalian target of rapamycin (mTOR) or phospho-mTOR.CONCLUSIONSExtent of resection, Al-Mefty classification, primary tumor, postoperative radiotherapy, and PDGFR-β expression level are valuable prognostic factors in patients with clival chordomas. PDGFR-β could regulate invasion through the mTOR pathway in clival chordoma cells.
Collapse
Affiliation(s)
- Yixuan Zhai
- 1Beijing Neurosurgical Institute, Capital Medical University; and
| | - Jiwei Bai
- 2Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Wang
- 1Beijing Neurosurgical Institute, Capital Medical University; and
| | - Hua Gao
- 1Beijing Neurosurgical Institute, Capital Medical University; and
| | - Mingxuan Li
- 1Beijing Neurosurgical Institute, Capital Medical University; and
| | - Chuzhong Li
- 1Beijing Neurosurgical Institute, Capital Medical University; and
| | - Songbai Gui
- 2Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- 1Beijing Neurosurgical Institute, Capital Medical University; and
| |
Collapse
|
21
|
NVP-BEZ235 Attenuated Cell Proliferation and Migration in the Squamous Cell Carcinoma of Oral Cavities and p70S6K Inhibition Mimics its Effect. Int J Mol Sci 2018; 19:ijms19113546. [PMID: 30423811 PMCID: PMC6274880 DOI: 10.3390/ijms19113546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 11/19/2022] Open
Abstract
NVP-BEZ235 or BEZ235 is a dual inhibitor of adenosine triphosphate (ATP)-competitive phosphoinositide 3-kinase (PI3K)/mammalian-target-of-rapamycin (mTOR) and is promising for cancer treatment. Because it targets more than one downstream effector, a dual approach is promising for cancer treatment. The aim of this study was to evaluate the efficacy of NVP-BEZ235 in treating oral cavity squamous cell carcinoma (OSCC). Two human OSCC cell lines, SCC-4 and SCC-25, were used in this study. PI3K-AKT signaling, proliferation, and cell migratory and invasion capabilities of OSCC cells were examined. In NVP-BEZ235-treated SCC-4 and SCC-25 cells, the phosphorylation of 70-kDa ribosomal S6 kinase (p70S6K), but not mTOR, decreased within 24 h. NVP-BEZ235 inhibited OSCC-cell proliferation, migration, and invasion possibly by directly deregulating the phosphorylation of p70S6K. The phospho-p70S6K inhibitor mimicked the effects of NVP-BEZ235 for preventing proliferation and weakening the migratory and invasion abilities of SCC-4 and SCC-25 cells. This study further confirmed the effect of NVP-BEZ235 on OSCC cells and provided a new strategy for controlling the proliferation, migration, and invasion of OSCC cells using the phopho-p70S6K inhibitor.
Collapse
|
22
|
PLGA-PEG nanoparticles for targeted delivery of the mTOR/PI3kinase inhibitor dactolisib to inflamed endothelium. Int J Pharm 2018; 548:747-758. [DOI: 10.1016/j.ijpharm.2017.10.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/27/2017] [Accepted: 10/13/2017] [Indexed: 12/17/2022]
|
23
|
Kosach V, Shkarina K, Kravchenko A, Tereshchenko Y, Kovalchuk E, Skoroda L, Krotevych M, Khoruzhenko A. Nucleocytoplasmic distribution of S6K1 depends on the density and motility of MCF-7 cells in vitro. F1000Res 2018; 7:1332. [PMID: 30705751 PMCID: PMC6343231 DOI: 10.12688/f1000research.15447.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Background: The ribosomal protein S6 kinase 1 (S6K1) is one of the main components of the mTOR/S6K signal transduction pathway, which controls cellular metabolism, autophagy, growth, and proliferation. Overexpression of S6K1 was detected in tumors of different origin including breast cancer, and correlated with the worse disease outcome. In addition, significant accumulation of S6K1 was found in the nuclei of breast carcinoma cells suggesting the implication of kinase nuclear substrates in tumor progression. However, this aspect of S6K1 functioning is still poorly understood. The main aim of the present work was to study the subcellular localization of S6K1 in breast cancer cells with the focus on cell migration. Methods: Multicellular spheroids of MCF-7 cells were generated using agarose-coated Petri dishes. Cell migration was induced by spheroids seeding onto adhesive growth surface and subsequent cultivation for 24 to 72 hours. The subcellular localization of S6K1 was studied in human normal breast and cancer tissue samples, 2D and 3D MCF-7 cell cultures using immunofluorescence analysis and confocal microscopy. Results: Analysis of histological sections of human breast tissue samples revealed predominantly nuclear localization of S6K1 in breast malignant cells and its mainly cytoplasmic localization in conditionally normal cells. In vitro studies of MCF-7 cells demonstrated that the subcellular localization of S6K1 depends on the cell density in the monolayer culture. S6K1 relocalization from the cytoplasm into the nucleus was detected in MCF-7 cells migrating from multicellular spheroids onto growth surface. Immunofluorescence analysis of S6K1 and immunocoprecipitation assay revealed the colocalization and interaction between S6K1 and transcription factor TBR2 (T-box brain protein 2) in MCF-7 cells. Conclusions: Subcellular localization of S6K1 depends on the density and locomotor activity of the MCF-7 cells.
Collapse
Affiliation(s)
- Viktoriia Kosach
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, 03143, Ukraine
| | - Kateryna Shkarina
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, 03143, Ukraine
- Educational and Scientific Center , Taras Shevchenko National University of Kyiv, Kyiv, 03022, Ukraine
| | - Anastasiia Kravchenko
- Educational and Scientific Center , Taras Shevchenko National University of Kyiv, Kyiv, 03022, Ukraine
| | - Yuliia Tereshchenko
- Educational and Scientific Center , Taras Shevchenko National University of Kyiv, Kyiv, 03022, Ukraine
| | | | | | | | - Antonina Khoruzhenko
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, 03143, Ukraine
| |
Collapse
|
24
|
Kosach V, Shkarina K, Kravchenko A, Tereshchenko Y, Kovalchuk E, Skoroda L, Krotevych M, Khoruzhenko A. Nucleocytoplasmic distribution of S6K1 depends on the density and motility of MCF-7 cells in vitro. F1000Res 2018; 7:1332. [PMID: 30705751 PMCID: PMC6343231 DOI: 10.12688/f1000research.15447.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/13/2018] [Indexed: 10/06/2023] Open
Abstract
Background: The ribosomal protein S6 kinase 1 (S6K1) is one of the main components of the mTOR/S6K signal transduction pathway, which controls cellular metabolism, autophagy, growth, and proliferation. Overexpression of S6K1 was detected in tumors of different origin including breast cancer, which was associated with a worse disease outcome. In addition, significant accumulation of S6K1 was found in the nuclei of breast carcinoma cells suggesting the implication of kinase nuclear substrates in tumor progression. However, this aspect of S6K1 functioning is poorly understood. The main aim of the present work was to study the subcellular localization of S6K1 in breast cancer cells with focus on cell migration. Methods: Multicellular spheroids of MCF-7 cells were generated using agarose-coated Petri dishes. Cell migration was initiated by spheroids seeding onto growth surface and subsequent cultivation for 24 and 72 hours. S6K1 subcellular localization was studied in human breast cancer and normal tissue, 2D and 3D MCF-7 cell culture using immunofluorescence analysis and confocal microscopy. Results: Analysis of histological sections of human breast cancer and normal tissue revealed predominantly nuclear localization of S6K1 in breast malignant cells and mainly cytoplasmic one in conditionally normal cells. In vitro studies of MCF-7 cells showed that the subcellular localization of S6K1 depends on the cell density in the monolayer culture. S6K1 relocalization from the cytoplasm into the nucleus was detected in MCF-7 cells migrating from multicellular spheroids onto growth surface. Immunofluorescence analysis of S6K1 and immunocoprecipitation assay revealed the colocalization and interaction between S6K1 and transcription factor TBR2 (T-box brain protein 2) in MCF-7 cells. Bioinformatical analysis revealed existence of several phosphorylation sites in TBR2 for S6K1 suggesting that TBR2 can be a target for phosphorylation and regulation by S6K1. Conclusions: Subcellular localization of S6K1 depends on the density and locomotor activity of the MCF-7 cells.
Collapse
Affiliation(s)
- Viktoriia Kosach
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, 03143, Ukraine
| | - Kateryna Shkarina
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, 03143, Ukraine
- Educational and Scientific Center , Taras Shevchenko National University of Kyiv, Kyiv, 03022, Ukraine
| | - Anastasiia Kravchenko
- Educational and Scientific Center , Taras Shevchenko National University of Kyiv, Kyiv, 03022, Ukraine
| | - Yuliia Tereshchenko
- Educational and Scientific Center , Taras Shevchenko National University of Kyiv, Kyiv, 03022, Ukraine
| | | | | | | | - Antonina Khoruzhenko
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, 03143, Ukraine
| |
Collapse
|
25
|
Mitochondrial Stress Tests Using Seahorse Respirometry on Intact Dictyostelium discoideum Cells. Methods Mol Biol 2017; 1407:41-61. [PMID: 27271893 DOI: 10.1007/978-1-4939-3480-5_4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mitochondria not only play a critical and central role in providing metabolic energy to the cell but are also integral to the other cellular processes such as modulation of various signaling pathways. These pathways affect many aspects of cell physiology, including cell movement, growth, division, differentiation, and death. Mitochondrial dysfunction which affects mitochondrial bioenergetics and causes oxidative phosphorylation defects can thus lead to altered cellular physiology and manifest in disease. The assessment of the mitochondrial bioenergetics can thus provide valuable insights into the physiological state, and the alterations to the state of the cells. Here, we describe a method to successfully use the Seahorse XF(e)24 Extracellular Flux Analyzer to assess the mitochondrial respirometry of the cellular slime mold Dictyostelium discoideum.
Collapse
|
26
|
LOC285629 regulates cell proliferation and motility in colorectal cancer cells. Clin Transl Oncol 2017; 20:775-784. [DOI: 10.1007/s12094-017-1788-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/23/2017] [Indexed: 10/18/2022]
|
27
|
Gholizadeh S, Visweswaran GRR, Storm G, Hennink WE, Kamps JAAM, Kok RJ. E-selectin targeted immunoliposomes for rapamycin delivery to activated endothelial cells. Int J Pharm 2017; 548:759-770. [PMID: 29038064 DOI: 10.1016/j.ijpharm.2017.10.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/25/2017] [Accepted: 10/11/2017] [Indexed: 02/07/2023]
Abstract
Activated endothelial cells play a pivotal role in the pathology of inflammatory disorders and thus present a target for therapeutic intervention by drugs that intervene in inflammatory signaling cascades, such as rapamycin (mammalian target of rapamycin (mTOR) inhibitor). In this study we developed anti-E-selectin immunoliposomes for targeted delivery to E-selectin over-expressing tumor necrosis factor-α (TNF-α) activated endothelial cells. Liposomes composed of 1,2-dipalmitoyl-sn-glycero-3.;hosphocholine (DPPC), Cholesterol, and 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethyleneglycol)-2000]-maleimide (DSPE-PEG-Mal) were loaded with rapamycin via lipid film hydration, after which they were further functionalized by coupling N-succinimidyl-S-acetylthioacetate (SATA)-modified mouse anti human E-selectin antibodies to the distal ends of the maleimidyl (Mal)-PEG groups. In cell binding assays, these immunoliposomes bound specifically to TNF-α activated endothelial cells. Upon internalization, rapamycin loaded immunoliposomes inhibited proliferation and migration of endothelial cells, as well as expression of inflammatory mediators. Our findings demonstrate that rapamycin-loaded immunoliposomes can specifically inhibit inflammatory responses in inflamed endothelial cells.
Collapse
Affiliation(s)
- Shima Gholizadeh
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ganesh Ram R Visweswaran
- Department of Pathology & Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jan A A M Kamps
- Department of Pathology & Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robbert J Kok
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
28
|
Hanai S, Sukigara S, Dai H, Owa T, Horike SI, Otsuki T, Saito T, Nakagawa E, Ikegaya N, Kaido T, Sato N, Takahashi A, Sugai K, Saito Y, Sasaki M, Hoshino M, Goto YI, Koizumi S, Itoh M. Pathologic Active mTOR Mutation in Brain Malformation with Intractable Epilepsy Leads to Cell-Autonomous Migration Delay. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1177-1185. [PMID: 28427592 DOI: 10.1016/j.ajpath.2017.01.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/19/2017] [Indexed: 01/08/2023]
Abstract
The activation of phosphatidylinositol 3-kinase-AKTs-mammalian target of rapamycin cell signaling pathway leads to cell overgrowth and abnormal migration and results in various types of cortical malformations, such as hemimegalencephaly (HME), focal cortical dysplasia, and tuberous sclerosis complex. However, the pathomechanism underlying abnormal cell migration remains unknown. With the use of fetal mouse brain, we performed causative gene analysis of the resected brain tissues from a patient with HME and investigated the pathogenesis. We obtained a novel somatic mutation of the MTOR gene, having approximately 11% and 7% mutation frequency in the resected brain tissues. Moreover, we revealed that the MTOR mutation resulted in hyperphosphorylation of its downstream molecules, S6 and 4E-binding protein 1, and delayed cell migration on the radial glial fiber and did not affect other cells. We suspect cell-autonomous migration arrest on the radial glial foot by the active MTOR mutation and offer potential explanations for why this may lead to cortical malformations such as HME.
Collapse
Affiliation(s)
- Sae Hanai
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, Kodaira, Japan
| | - Sayuri Sukigara
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, Kodaira, Japan
| | - Hongmei Dai
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, Kodaira, Japan
| | - Tomoo Owa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, Kodaira, Japan
| | - Shin-Ichi Horike
- Division of Functional Genomics, Advanced Science Research Center Kanazawa University, Kanazawa, Japan
| | - Taisuke Otsuki
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Neurosurgery, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Takashi Saito
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Child Neurology, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Eiji Nakagawa
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Child Neurology, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Naoki Ikegaya
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Neurosurgery, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Takanobu Kaido
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Neurosurgery, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Noriko Sato
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Radiology, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Akio Takahashi
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Neurosurgery, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Kenji Sugai
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Child Neurology, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Yuko Saito
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Laboratory Medicine, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Masayuki Sasaki
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Child Neurology, Hospital of National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Mikio Hoshino
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, Kodaira, Japan
| | - Yu-Ichi Goto
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, Kodaira, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Masayuki Itoh
- Epilepsy Center, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Kodaira, Japan; Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, Kodaira, Japan.
| |
Collapse
|
29
|
Kim LW. Dual TORCs driven and B56 orchestrated signaling network guides eukaryotic cell migration. BMB Rep 2017; 50:437-444. [PMID: 28571594 PMCID: PMC5625690 DOI: 10.5483/bmbrep.2017.50.9.091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Indexed: 11/20/2022] Open
Abstract
Different types of eukaryotic cells may adopt seemingly distinct modes of directional cell migration. However, several core aspects are regarded common whether the movement is either ameoboidal or mesenchymal. The region of cells facing the attractive signal is often termed leading edge where lamellipodial structures dominates and the other end of the cell called rear end is often mediating cytoskeletal F-actin contraction involving Myosin-II. Dynamic remodeling of cell-to-matrix adhesion involving integrin is also evident in many types of migrating cells. All these three aspects of cell migration are significantly affected by signaling networks of TorC2, TorC1, and PP2A/B56. Here we review the current views of the mechanistic understanding of these regulatory signaling networks and how these networks affect eukaryotic cell migration.
Collapse
Affiliation(s)
- Lou W Kim
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
30
|
Kim CJ, Tambe Y, Mukaisho KI, Sugihara H, Kageyama S, Kawauchi A, Inoue H. Periostin suppresses in vivo invasiveness via PDK1/Akt/mTOR signaling pathway in a mouse orthotopic model of bladder cancer. Oncol Lett 2017; 13:4276-4284. [PMID: 28599427 PMCID: PMC5452989 DOI: 10.3892/ol.2017.6004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/14/2017] [Indexed: 01/14/2023] Open
Abstract
Periostin is an extracellular matrix protein involved in the regulation of intercellular adhesion. The present study investigated the in vivo tumor suppressor function of periostin in a mouse orthotopic model of bladder cancer. Retroviral vectors were used to transfect human bladder cancer UMUC-3 cell line with periostin. Bladders of nude mice that were transurethrally instilled with periostin-expressing UMUC-3 cells were revealed to weigh less compared with bladders instilled with vector control cells. In total, five (83.3%) of six vector control UMUC-3 bladder tumors exhibited histological evidence of muscle invasion. However, none of the five periostin-expressing UMUC-3 bladder tumors revealed muscle invasion. Thick edematous lesions were present in the submucosa of periostin-expressing UMUC-3 bladder tumors. The expression of periostin also suppressed in vitro cell invasiveness of UMUC-3 cells without affecting cellular proliferation. The level of phosphorylation of phosphoinositide-dependent kinase-1 (PDK1), protein kinase B (Akt) and S6 ribosomal protein, a downstream protein of mammalian target of rapamycin (mTOR) was decreased in periostin-expressing UMUC-3 cells compared with vector control cells. Treatment with 100 ng/ml recombinant human periostin protein also suppressed cell invasiveness and phosphorylation of PDK1, Akt and S6 in UMUC-3 cells, consistent with results using periostin-expressing UMUC-3 cells. Treatment with PDK1, Akt and mTOR inhibitors significantly suppressed UMUC-3 cell invasiveness. These results demonstrate that periostin suppresses in vivo and in vitro invasiveness of bladder cancer via the PDK1/Akt/mTOR signaling pathway. Periostin may be useful as a potent chemotherapeutic agent by suppressing bladder cancer invasiveness.
Collapse
Affiliation(s)
- Chul Jang Kim
- Department of Urology, Kohka Public Hospital, Kohka, Shiga 528-6024, Japan
| | - Yukihiro Tambe
- Division of Microbiology and Infectious Diseases, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Ken-Ichi Mukaisho
- Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Hiroyuki Sugihara
- Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Hirokazu Inoue
- Division of Microbiology and Infectious Diseases, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
31
|
Amaral CL, Freitas LB, Tamura RE, Tavares MR, Pavan ICB, Bajgelman MC, Simabuco FM. S6Ks isoforms contribute to viability, migration, docetaxel resistance and tumor formation of prostate cancer cells. BMC Cancer 2016; 16:602. [PMID: 27491285 PMCID: PMC4974797 DOI: 10.1186/s12885-016-2629-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background The S6 Kinase (S6K) proteins are some of the main downstream effectors of the mammalian Target Of Rapamycin (mTOR) and act as key regulators of protein synthesis and cell growth. S6K is overexpressed in a variety of human tumors and is correlated to poor prognosis in prostate cancer. Due to the current urgency to identify factors involved in prostate cancer progression, we aimed to reveal the cellular functions of three S6K isoforms–p70-S6K1, p85-S6K1 and p54-S6K2–in prostate cancer, as well as their potential as therapeutic targets. Methods In this study we performed S6K knockdown and overexpression and investigated its role in prostate cancer cell proliferation, colony formation, viability, migration and resistance to docetaxel treatment. In addition, we measured tumor growth in Nude mice injected with PC3 cells overexpressing S6K isoforms and tested the efficacy of a new available S6K1 inhibitor in vitro. Results S6Ks overexpression enhanced PC3-luc cell line viability, migration, resistance to docetaxel and tumor formation in Nude mice. Only S6K2 knockdown rendered prostate cancer cells more sensitive to docetaxel. S6K1 inhibitor PF-4708671 was particularly effective for reducing migration and proliferation of PC3 cell line. Conclusions These findings demonstrate that S6Ks play an important role in prostate cancer progression, enhancing cell viability, migration and chemotherapy resistance, and place both S6K1 and S6K2 as a potential targets in advanced prostate cancer. We also provide evidence that S6K1 inhibitor PF-4708671 may be considered as a potential drug for prostate cancer treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2629-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camila L Amaral
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Lidia B Freitas
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Rodrigo E Tamura
- Viral Vector Laboratory, Center for Translational Investigation in Oncology/LIM24, Cancer Institute of São Paulo, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mariana R Tavares
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Isadora C B Pavan
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil
| | - Marcio C Bajgelman
- Brazilian Biosciences National Laboratory, Brazilian National Center for Research in Energy and Materials, Campinas, São Paulo, Brazil
| | - Fernando M Simabuco
- Laboratory of Disorders of Metabolism, School of Applied Sciences, University of Campinas, R. Pedro Zaccaria, 1300, sala LA 421, 13484-350, Limeira, São Paulo, Brazil.
| |
Collapse
|
32
|
Park JH, Cho YY, Yoon SW, Park B. Suppression of MMP-9 and FAK expression by pomolic acid via blocking of NF-κB/ERK/mTOR signaling pathways in growth factor-stimulated human breast cancer cells. Int J Oncol 2016; 49:1230-40. [PMID: 27573547 DOI: 10.3892/ijo.2016.3585] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/14/2016] [Indexed: 11/06/2022] Open
Abstract
The expression of matrix metalloproteinase-9 (MMP-9) and the phosphorylation of focal adhesion kinase (FAK) have been implicated in the invasion, metastasis and cell motility of cancer cells. It is considered that epidermal growth factor (EGF) may increase cell motility, an event involved in cancer cell invasion and metastasis. Pomolic acid (PA), an active triterpenoid from Euscaphis japonica, is known to inhibit the proliferation of a variety of cancer cells, but the effect of PA on the invasiveness of cancer cells is largely unknown. In this study, we first determined the molecular mechanism by which PA inhibits the migratory and invasive abilities of highly metastatic MDA-MB‑231 cells. Transwell invasion, wound-healing assay and F-actin reorganization showed that PA significantly inhibits the EGF-induced invasion, migration and cell motility by reducing expression of MMP-9 and FAK phosphorylation. In particular, PA potently suppressed the phosphorylation of nuclear factor (NF)-κB, extraceullar signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway. Furthermore, PA treatment inhibited the DNA binding activity of NF-κB and activator protein (AP)-1, which is known to mediate the expression of EGFR and MMP-9. These results suggest that PA may be a potential therapeutic candidate for treatment of breast cancer metastasis.
Collapse
Affiliation(s)
- Ji-Hyun Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Yoon Young Cho
- Department of Oncology/Hematology, Daegu Catholic University Medical Center, Daegu 42472, Republic of Korea
| | - Seong Woo Yoon
- Department of Korean Internal Medicine, Korean Medicine Cancer Center, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
33
|
Riquelme I, Tapia O, Leal P, Sandoval A, Varga MG, Letelier P, Buchegger K, Bizama C, Espinoza JA, Peek RM, Araya JC, Roa JC. miR-101-2, miR-125b-2 and miR-451a act as potential tumor suppressors in gastric cancer through regulation of the PI3K/AKT/mTOR pathway. Cell Oncol (Dordr) 2016; 39:23-33. [PMID: 26458815 PMCID: PMC4751587 DOI: 10.1007/s13402-015-0247-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is a deadly malignancy worldwide. In the past, it has been shown that cellular signaling pathway alterations play a crucial role in the development of GC. In particular, deregulation of the PI3K/AKT/mTOR pathway seems to affect multiple GC functions including growth, proliferation, metabolism, motility and angiogenesis. Targeting alterations in this pathway by microRNAs (miRNAs) represents a potential therapeutic strategy, especially in inhibitor-resistant tumors. The objective of this study was to evaluate the expression of 3 pre-selected miRNAs, miR-101-2, miR-125b-2 and miR-451a, in a series of primary GC tissues and matched non-GC tissues and in several GC-derived cell lines, and to subsequently evaluate the functional role of these miRNAs. METHODS Twenty-five primary GC samples, 25 matched non-GC samples and 3 GC-derived cell lines, i.e., AGS, MKN28 and MKN45, were included in this study. miRNA and target gene expression levels were assessed by quantitative RT-PCR and western blotting, respectively. Subsequently, cell viability, clone formation, cell death, migration and invasion assays were performed on AGS cells. RESULTS miR-101-2, miR-125b-2 and miR-451a were found to be down-regulated in the primary GC tissues and the GC-derived cell lines tested. MiRNA mimic transfections significantly reduced cell viability and colony formation, increased cell death and reduced cell migration and invasion in AGS cells. We also found that exogenous expression of miR-101-2, miR-125b-2 and miR-451a decreased the expression of their putative targets MTOR, PIK3CB and TSC1, respectively. CONCLUSIONS Our expression analyses and in vitro functional assays suggest that miR-101-2, miR-125b-2 and miR-451a act as potential tumor suppressors in primary GCs as well as in GC-derived AGS cells.
Collapse
Affiliation(s)
- Ismael Riquelme
- Laboratory of Molecular Pathology, Pathology Department, School of Medicine, BIOREN-CEGIN, Universidad de La Frontera, Avenida Alemania 0458, 4810296 Temuco, Chile
| | - Oscar Tapia
- Laboratory of Molecular Pathology, Pathology Department, School of Medicine, BIOREN-CEGIN, Universidad de La Frontera, Avenida Alemania 0458, 4810296 Temuco, Chile
| | - Pamela Leal
- Molecular Biology and Biomedicine Lab, CEGIN-BIOREN, Universidad de La Frontera, Avenida Alemania 0458, 4810296 Temuco, Chile
| | - Alejandra Sandoval
- Laboratory of Molecular Pathology, Pathology Department, School of Medicine, BIOREN-CEGIN, Universidad de La Frontera, Avenida Alemania 0458, 4810296 Temuco, Chile
| | - Matthew G. Varga
- Division of Gastroenterology, Departments of Medicine and Cancer Biology, Vanderbilt University School of Medicine, 2215 Garland Avenue Nashville, 37232 Nashville, TN, USA
| | - Pablo Letelier
- School of Health Sciences, Universidad Catolica de Temuco, Manuel Montt 56, 4813302 Temuco, Chile
| | - Kurt Buchegger
- Laboratory of Molecular Pathology, Pathology Department, School of Medicine, BIOREN-CEGIN, Universidad de La Frontera, Avenida Alemania 0458, 4810296 Temuco, Chile
| | - Carolina Bizama
- Department of Pathology, UC Centre for Investigational Oncology (CITO), Advanced Centre for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Catolica de Chile, Marcoleta 377, 7th Floor, 8330024 Santiago, Chile
| | - Jaime A. Espinoza
- Department of Pathology, UC Centre for Investigational Oncology (CITO), Advanced Centre for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Catolica de Chile, Marcoleta 377, 7th Floor, 8330024 Santiago, Chile
| | - Richard M. Peek
- Division of Gastroenterology, Departments of Medicine and Cancer Biology, Vanderbilt University School of Medicine, 2215 Garland Avenue Nashville, 37232 Nashville, TN, USA
| | - Juan Carlos Araya
- Department of Pathology, School of Medicine, Universidad de La Frontera, Avenida Alemania 0458, 4810296 Temuco, Chile
| | - Juan Carlos Roa
- Department of Pathology, UC Centre for Investigational Oncology (CITO), Advanced Centre for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Catolica de Chile, Marcoleta 377, 7th Floor, 8330024 Santiago, Chile
| |
Collapse
|
34
|
Global profiling of the signaling network of papillary thyroid carcinoma. Life Sci 2016; 147:9-14. [DOI: 10.1016/j.lfs.2016.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/26/2015] [Accepted: 01/04/2016] [Indexed: 01/01/2023]
|
35
|
Gabanella F, Pisani C, Borreca A, Farioli-Vecchioli S, Ciotti MT, Ingegnere T, Onori A, Ammassari-Teule M, Corbi N, Canu N, Monaco L, Passananti C, Di Certo MG. SMN affects membrane remodelling and anchoring of the protein synthesis machinery. J Cell Sci 2016; 129:804-16. [PMID: 26743087 DOI: 10.1242/jcs.176750] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/29/2015] [Indexed: 12/31/2022] Open
Abstract
Disconnection between membrane signalling and actin networks can have catastrophic effects depending on cell size and polarity. The survival motor neuron (SMN) protein is ubiquitously involved in assembly of spliceosomal small nuclear ribonucleoprotein particles. Other SMN functions could, however, affect cellular activities driving asymmetrical cell surface expansions. Genes able to mitigate SMN deficiency operate within pathways in which SMN can act, such as mRNA translation, actin network and endocytosis. Here, we found that SMN accumulates at membrane protrusions during the dynamic rearrangement of the actin filaments. In addition to localization data, we show that SMN interacts with caveolin-1, which mediates anchoring of translation machinery components. Importantly, SMN deficiency depletes the plasma membrane of ribosomes, and this correlates with the failure of fibroblasts to extend membrane protrusions. These findings strongly support a relationship between SMN and membrane dynamics. We propose that SMN could assembly translational platforms associated with and governed by the plasma membrane. This activity could be crucial in cells that have an exacerbated interdependence of membrane remodelling and local protein synthesis.
Collapse
Affiliation(s)
- Francesca Gabanella
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Cinzia Pisani
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Antonella Borreca
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Stefano Farioli-Vecchioli
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Maria Teresa Ciotti
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy European Brain Research Institute (EBRI) Rita Levi-Montalcini, Rome 00143, Italy
| | - Tiziano Ingegnere
- Department of Ecological and Biological Sciences, Tuscia University, Viterbo 01100, Italy
| | - Annalisa Onori
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Martine Ammassari-Teule
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| | - Nicoletta Corbi
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Nadia Canu
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy Department of System Medicine, University of 'Tor Vergata', Rome 00137, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome 00185, Italy
| | - Claudio Passananti
- CNR-IBPM, Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy
| | - Maria Grazia Di Certo
- CNR-Institute of Cell Biology and Neurobiology, Rome 00143, Italy IRCCS Fondazione Santa Lucia, Rome 00143, Italy
| |
Collapse
|
36
|
Cao W, Li F, Yao J, Yu J. Prostate specific G protein coupled receptor is associated with prostate cancer prognosis and affects cancer cell proliferation and invasion. BMC Cancer 2015; 15:915. [PMID: 26582057 PMCID: PMC4652354 DOI: 10.1186/s12885-015-1921-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 11/10/2015] [Indexed: 02/05/2023] Open
Abstract
Background There is limited information about the clinical and biological significance of prostate specific G protein coupled receptor (PSGR) in prostate cancer (PCa) initiation and progression. Here, we evaluated the expression of PSGR protein, studied its diagnostic and prognostic value in PCa, and also explored its role in cancer cell growth and invasion. Methods The expression of PSGR in paired adjacent normal prostate, high grade prostatic intraepithelial neoplasia (PIN), and PCa were determined by immunohistochemistry on tissue microarrays constructed from 150 radical prostatectomy specimens. The effects of PSGR on PCa cell growth and invasion were investigated using human PCa cell lines. Results Membranous and cytoplasmic PSGR staining was observed at luminal epithelial cells of prostate. PSGR protein expression was significantly higher in PIN compared to normal prostate. Interestingly, the expression of PSGR decreased as PIN progressed to PCa. Low PSGR expression in PCa was associated with high Gleason score, and poor overall survival. Activated PSGR increased cancer cell invasive ability, but retarded cell growth. PSGR did not affect mTOR activity, but suppressed P70 S6 kinase activity. Conclusions PSGR may participate in PCa progression through affecting cell proliferation and invasion. High expression of PSGR in PIN may implicate its role in early neoplastic transformation of PCa. Low expression of PSGR in PCa may serve as a potential indicator for poor prognosis.
Collapse
Affiliation(s)
- Wenqing Cao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Pathology, New York University Langone Medical Center, New York City, NY, USA.
| | - Faqian Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA.
| | - Jorge Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA. .,Pathline Laboratories, Ramsey, NJ, USA.
| | - Jiangzhou Yu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
37
|
Sonoda Y, Tooyama I, Mukai H, Maeda K, Akiyama H, Kawamata T. S6 kinase phosphorylated at T229 is involved in tau and actin pathologies in Alzheimer's disease. Neuropathology 2015; 36:325-32. [PMID: 26582459 DOI: 10.1111/neup.12275] [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: 07/17/2015] [Revised: 10/18/2015] [Accepted: 10/19/2015] [Indexed: 01/15/2023]
Abstract
The 70-kDa ribosomal protein S6 kinase (S6K), a serine/threonine kinase that modulates the phosphorylation of the 40S ribosomal protein S6, regulates cell cycle progression and is known as a tau kinase in Alzheimer's disease (AD). In AD brains, neurofibrillary tangles (NFTs) have been shown to be positively stained with antibodies against S6K proteins phosphorylated at T389 (pT389-S6K) or T421/S424 (pT421/S424-S6K) by the mammalian target of rapamycin and mitogen-activated protein kinase pathways, respectively. However, there is little information available about S6K proteins directly phosphorylated at T229 (pT229-S6K) by the PI3K-PDK1 pathway. In the present study, we investigated the distribution of pT229-S6K in post mortem human brain tissues from elderly (control) and patients with AD using immnunoblotting and immunohistochemistry. pT229-S6K immunoreactivity was localized to small granular structures in neurons and endothelial cells in control and AD brains. In AD brains, intense pT229-S6K immunoreactivity was detected in 16.3% of AT8-positive NFTs, neuropil threads, and dystrophic neurites in the hippocampus and other vulnerable brain areas. In addition, Hirano bodies were also positive for pT229-S6K but were negative for pT389-S6K or pT421/S424-S6K. The present results indicate that S6K phosphorylation via the PI3K-PD1 pathway is involved in tau pathology in NFTs and abnormal neurites as well as actin pathology in Hirano bodies.
Collapse
Affiliation(s)
- Yuma Sonoda
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Rehabilitation Units, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | | | - Kiyoshi Maeda
- Department of Medical Rehabilitation, Kobe Gakuin University, Kobe, Japan
| | - Haruhiko Akiyama
- Department of Neuropathology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | - Toshio Kawamata
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
38
|
Zhao G, Han X, Zheng S, Li Z, Sha Y, Ni J, Sun Z, Qiao S, Song Z. Curcumin induces autophagy, inhibits proliferation and invasion by downregulating AKT/mTOR signaling pathway in human melanoma cells. Oncol Rep 2015; 35:1065-74. [PMID: 26573768 DOI: 10.3892/or.2015.4413] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/26/2015] [Indexed: 01/12/2023] Open
Abstract
Melanoma is the foremost malignant cutaneous cancer and it is extremely resistant to chemotherapy and radiotherapy. Curcumin is an active component of turmeric, the yellow spice derived from the rhizome of Curcuma longa, and is widely known for its anti-inflammatory and anti-cancerogenic properties. Several recent studies suggest that curcumin induces apoptosis by modulating multiple signaling pathways to exert its anticancer effect. In the present study, we investigated the effect of curcumin on the viability, invasion potential, cell cycle, autophagy and the AKT, mTOR, P70S6K proteins of AKT/mTOR signaling pathway in human melanoma A375 and C8161 cell lines in vitro and in an in vivo tumorigenesis model. Curcumin effectively inhibited the proliferation of melanoma cells in vitro and in vivo. It suppressed cell invasion, arrested the cancer cells at G2/M phase of the cell cycle, and induced autophagy. Furthermore, curcumin suppressed the activation of AKT, mTOR and P70S6K proteins. Curcumin, therefore, is a potent suppressor of cell viability and invasion, and simultaneously an inducer of autophagy in A375 and C8161 cells. Accordingly, curcumin could be a novel therapeutic candidate for the management of melanoma.
Collapse
Affiliation(s)
- Guangming Zhao
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiaodong Han
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Siwen Zheng
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Zhen Li
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yang Sha
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Jing Ni
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Zhe Sun
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Song Qiao
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Zhiqi Song
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
39
|
Li C, Cui JF, Chen MB, Liu CY, Liu F, Zhang QD, Zou J, Lu PH. The preclinical evaluation of the dual mTORC1/2 inhibitor INK-128 as a potential anti-colorectal cancer agent. Cancer Biol Ther 2015; 16:34-42. [PMID: 25692620 PMCID: PMC4623257 DOI: 10.4161/15384047.2014.972274] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The colorectal cancer is the leading contributor of cancer-related mortality. Mammalian target of rapamycin (mTOR), existing in 2 complexes (mTORC1/2), is frequently dysregulated and constitutively activated in colorectal cancers. It represents an important drug target. Here we found that INK-128, the novel ATP-competitive kinase inhibitor of mTOR, blocked both mTORC1 and mTORC2 activation in colorectal cancer cells (both primary and transformed cells). The immunoprecipitation results showed that the assembly of mTORC1 (mTOR-Raptor association) and mTORC2 (mTOR-Rictor-Sin1 association) was also disrupted by INK-128. INK-128 inhibited colorectal cancer cell growth and survival, and induced both apoptotic and non-apoptotic cancer cell death. Further, INK-128 showed no effect on Erk/MAPK activation, while MEK/Erk inhibition by MEK-162 enhanced INK-128-induced cytotoxicity in colorectal cancer cells. Meanwhile, INK-128 downregulated Fascin1 (FSCN1)/E-Cadherin expressions and inhibited HT-29 cell in vitro migration. In vivo, daily INK-128 oral administration inhibited HT-29 xenograft growth in mice, which was further enhanced by MEK-162 administration. Finally, we found that INK-128 sensitized 5-fluorouracil-(5-FU)-mediated anti-HT-29 activity in vivo and in vitro. Thus, our preclinical studies strongly suggest that INK-128 might be investigated for colorectal cancer treatment in clinical trials.
Collapse
Key Words
- (S6K), p70S6K1
- (mTOR), mammalian target of rapamycin
- (mTORC1), mTOR complex 1
- (mTORC2), mTOR complex 2
- Co-IP, co-immunoprecipitation
- ECL, enhanced chemiluminescence
- FSCN10, Fascin1
- HRP, horseradish peroxidase
- INK-128
- PI, propidium iodide
- SD, standard deviation (SD)
- cell growth and migration
- colorectal cancer
- mTOR
Collapse
Affiliation(s)
- Chen Li
- a Department of Gastroenterology; Xuzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine ; Xuzhou , China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Franco-Villanueva A, Wandosell F, Antón IM. Neuritic complexity of hippocampal neurons depends on WIP-mediated mTORC1 and Abl family kinases activities. Brain Behav 2015; 5:e00359. [PMID: 26664784 PMCID: PMC4667760 DOI: 10.1002/brb3.359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/17/2015] [Accepted: 05/19/2015] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Neuronal morphogenesis is governed mainly by two interconnected processes, cytoskeletal reorganization, and signal transduction. The actin-binding molecule WIP (Wiskott-Aldrich syndrome protein [WASP]-interacting protein) was identified as a negative regulator of neuritogenesis. Although WIP controls activity of the actin-nucleation-promoting factor neural WASP (N-WASP) during neuritic differentiation, its implication in signal transduction remains unknown. METHODS Using primary neurons from WIP-deficient and wild-type mice we did an immunofluorescence, morphometric, and biochemical analysis of the signaling modified by WIP deficiency. RESULTS Here, we describe the WIP contribution to the regulation of neuritic elaboration and ramification through modification in phosphorylation levels of several kinases that participate in the mammalian target of rapamycin complex 1 (mTORC1)-p70S6K (phosphoprotein 70 ribosomal protein S6 kinase, S6K) intracellular signaling pathway. WIP deficiency induces an increase in the number of neuritic bifurcations and filopodial protrusions in primary embryonic neurons. This phenotype is not due to modifications in the activity of the phosphoinositide 3 kinase (PI3K)-Akt pathway, but to reduced phosphorylation of the S6K residues Ser(411) and Thr(389). The resulting decrease in kinase activity leads to reduced S6 phosphorylation in the absence of WIP. Incubation of control neurons with pharmacological inhibitors of mTORC1 or Abl, two S6K regulators, conferred a morphology resembling that of WIP-deficient neurons. Moreover, the preferential co-distribution of phospho-S6K with polymerized actin is altered in WIP-deficient neurons. CONCLUSION These experiments identify WIP as a member of a signaling cascade comprised of Abl family kinases, mTORC1 and S6K, which regulates neuron development and specifically, neuritic branching and complexity. Thus, we postulated a new role for WIP protein.
Collapse
Affiliation(s)
- Ana Franco-Villanueva
- Centro Nacional de Biotecnología (CNB-CSIC) Darwin 3 Campus Cantoblanco 28049 Madrid Spain ; CIBERNED, Centro Investigación Biomédica en Red de Enfermedades Neurodegenerativas Madrid Spain
| | - Francisco Wandosell
- CIBERNED, Centro Investigación Biomédica en Red de Enfermedades Neurodegenerativas Madrid Spain ; Centro de Biología Molecular Severo Ochoa (CBMSO) (CSIC-UAM) Nicolás Cabrera 1 Campus Cantoblanco 28049 Madrid Spain
| | - Inés M Antón
- Centro Nacional de Biotecnología (CNB-CSIC) Darwin 3 Campus Cantoblanco 28049 Madrid Spain ; CIBERNED, Centro Investigación Biomédica en Red de Enfermedades Neurodegenerativas Madrid Spain
| |
Collapse
|
41
|
Li WD, Hu N, Lei FR, Wei S, Rong JJ, Zhuang H, Li XQ. Autophagy inhibits endothelial progenitor cells migration via the regulation of MMP2, MMP9 and uPA under normoxia condition. Biochem Biophys Res Commun 2015; 466:376-80. [PMID: 26363453 DOI: 10.1016/j.bbrc.2015.09.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/05/2015] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The aim of this study was to explore the role of autophagy on the regulation of endothelial progenitor cells (EPCs) migration under normoxic condition. METHODS After EPCs were isolated and characterized in vitro, we employed Atg5 knocking down and rapamycin to monitor the autophagy, and performed wound healing and transwell assay to assess the cell migration. On the mechanism, the expression of matrix metalloproteinases (MMPs) and urokinase type plasminogen activator (uPA) was evaluated. RESULTS Atg5 knocking down and rapamycin could respectively inhibit and enhance autophagy, which could result in significantly increased and decreased cell migration in wound healing and transwell assay under normoxic condition. Moreover, Atg5 knocking down could significantly increase the expression of MMP2, MMP9 and uPA in EPCs while rapamycin could decrease the expression of uPA and MMP9. In addition, the mTOR-P70 S6K pathway was also involved in EPCs migration regulation. CONCLUSIONS These results demonstrated that autophagy could regulate the EPCs migration through mTOR-P70 S6K pathway, and MMP2, MMP9 and uPA may also involve in the regulation mechanism.
Collapse
Affiliation(s)
- Wen-Dong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Nan Hu
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng-Rui Lei
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Sen Wei
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Jie Rong
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Zhuang
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
42
|
Sabbatini M, Ruggiero G, Palatucci AT, Rubino V, Federico S, Giovazzino A, Apicella L, Santopaolo M, Matarese G, Galgani M, Terrazzano G. Oscillatory mTOR inhibition and Treg increase in kidney transplantation. Clin Exp Immunol 2015; 182:230-40. [PMID: 26077103 DOI: 10.1111/cei.12669] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2015] [Indexed: 01/13/2023] Open
Abstract
Intracellular metabolic pathways dependent upon the mammalian target of rapamycin (mTOR) play a key role in immune-tolerance control. In this study, we focused on long-term mTOR-dependent immune-modulating effects in kidney transplant recipients undergoing conversion from calcineurin inhibitors (CNI) to mTOR inhibitors (everolimus) in a 1-year follow-up. The conversion to everolimus is associated with a decrease of neutrophils and of CD8(+) T cells. In addition, we observed a reduced production of interferon (IFN)-γ by CD8(+) T cells and of interleukin (IL)-17 by CD4(+) T lymphocytes. An increase in CD4(+) CD25(+) forkhead box protein 3 (FoxP3)(+) [regulatory T cell [(Treg)] numbers was also seen. Treg increase correlated with a higher proliferation rate of this regulatory subpopulation when compared with the CD4(+) FoxP3(-) effector counterpart. Basal phosphorylation level of S6 kinase, a major mTOR-dependent molecular target, was substantially maintained in patients treated with everolimus. Moreover, oscillations in serum concentration of everolimus were associated with changes in basal and activation-dependent S6 kinase phosphorylation of CD4(+) and CD8(+) T cells. Indeed, T cell receptor (TCR) triggering was observed to induce significantly higher S6 kinase phosphorylation in the presence of lower everolimus serum concentrations. These results unveil the complex mTOR-dependent immune-metabolic network leading to long-term immune-modulation and might have relevance for novel therapeutic settings in kidney transplants.
Collapse
Affiliation(s)
- M Sabbatini
- Dipartimento di Sanità Pubblica, DH di Nefrologia e Trapianto di Rene, Università di Napoli 'Federico II', Napoli, Italy
| | - G Ruggiero
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy
| | - A T Palatucci
- Dottorato di Scienze.,Dipartimento di Scienze, Università della Basilicata, Potenza, Italy
| | - V Rubino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy
| | - S Federico
- Dipartimento di Sanità Pubblica, DH di Nefrologia e Trapianto di Rene, Università di Napoli 'Federico II', Napoli, Italy
| | - A Giovazzino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy.,Dipartimento di Scienze, Università della Basilicata, Potenza, Italy
| | - L Apicella
- Dipartimento di Sanità Pubblica, DH di Nefrologia e Trapianto di Rene, Università di Napoli 'Federico II', Napoli, Italy
| | - M Santopaolo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli 'Federico II', Napoli Italy
| | - G Matarese
- Dipartimento di Medicina e Chirurgia, Università di Salerno, Salerno, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milano, Italy
| | - M Galgani
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Napoli, Italy
| | - G Terrazzano
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli 'Federico II', Napoli, Italy.,Dipartimento di Scienze, Università della Basilicata, Potenza, Italy
| |
Collapse
|
43
|
Agliarulo I, Matassa DS, Amoroso MR, Maddalena F, Sisinni L, Sepe L, Ferrari MC, Arzeni D, Avolio R, Paolella G, Landriscina M, Esposito F. TRAP1 controls cell migration of cancer cells in metabolic stress conditions: Correlations with AKT/p70S6K pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2570-9. [PMID: 26071104 DOI: 10.1016/j.bbamcr.2015.05.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 05/14/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022]
Abstract
Cell motility is a highly dynamic phenomenon that is essential to physiological processes such as morphogenesis, wound healing and immune response, but also involved in pathological conditions such as metastatic dissemination of cancers. The involvement of the molecular chaperone TRAP1 in the regulation of cell motility, although still controversial, has been recently investigated along with some well-characterized roles in cancer cell survival and drug resistance in several tumour types. Among different functions, TRAP1-dependent regulation of protein synthesis seems to be involved in the migratory behaviour of cancer cells and, interestingly, the expression of p70S6K, a kinase responsible for translation initiation, playing a role in cell motility, is regulated by TRAP1. In this study, we demonstrate that TRAP1 silencing enhances cell motility in vitro but compromises the ability of cells to overcome stress conditions, and that this effect is mediated by the AKT/p70S6K pathway. In fact: i) inhibition of p70S6K activity specifically reduces migration in TRAP1 knock-down cells; ii) nutrient deprivation affects p70S6K activity thereby impairing cell migration only in TRAP1-deficient cells; iii) TRAP1 regulates the expression of both AKT and p70S6K at post-transcriptional level; and iii) TRAP1 silencing modulates the expression of genes involved in cell motility and epithelial-mesenchymal transition. Notably, a correlation between TRAP1 and AKT expression is found in vivo in human colorectal tumours. These results provide new insights into TRAP1 role in the regulation of cell migration in cancer cells, tumour progression and metastatic mechanisms.
Collapse
Affiliation(s)
- Ilenia Agliarulo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Danilo Swann Matassa
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Lorenza Sisinni
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Leandra Sepe
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; Ceinge Biotecnologie Avanzate, Via G. Salvatore 486, 80145 Naples, Italy
| | - Maria Carla Ferrari
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; Ceinge Biotecnologie Avanzate, Via G. Salvatore 486, 80145 Naples, Italy
| | - Diana Arzeni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Rosario Avolio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Giovanni Paolella
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; Ceinge Biotecnologie Avanzate, Via G. Salvatore 486, 80145 Naples, Italy
| | - Matteo Landriscina
- Clinical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
44
|
Annexin A1 Preferentially Predicts Poor Prognosis of Basal-Like Breast Cancer Patients by Activating mTOR-S6 Signaling. PLoS One 2015; 10:e0127678. [PMID: 26000884 PMCID: PMC4441370 DOI: 10.1371/journal.pone.0127678] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 04/17/2015] [Indexed: 11/24/2022] Open
Abstract
Introduction Annexin A1 (ANXA1) is an anti-inflammatory protein reported to play a role in cell proliferation and apoptosis, and to be deregulated in breast cancer. The exact role of annexin A1 in the biology of breast cancer remains unclear. We hypothesized that the annexin A1 plays an oncogenic role in basal subtype of breast cancer by modulating key growth pathway(s). Methods By mining the Cancer Genome Atlas (TCGA)-Breast Cancer dataset and manipulating annexin A1 levels in breast cancer cell lines, we studied the role of annexin A1 in breast cancer and underlying signaling pathways. Results Our in-silico analysis of TCGA-breast cancer dataset demonstrated that annexin A1 mRNA expression is higher in basal subtype compared to luminal and HER2 subtypes. Within the basal subtype, patients show significantly poorer overall survival associated with higher expression of annexin A1. In both TCGA patient samples and cell lines, annexin A1 levels were significantly higher in basal-like breast cancer than luminal and Her2/neu-positive breast cancer. Stable annexin A1 knockdown in TNBC cell lines suppressed the mTOR-S6 pathway likely through activation of AMPK but had no impact on the MAPK, c-Met, and EGFR pathways. In a cell migration assay, annexin A1-depleted TNBC cells showed delayed migration as compared to wild-type cells, which could be responsible for poor patient prognosis in basal like breast cancers that are known to express higher annexin A1. Conclusions Our data suggest that annexin A1 is prognostic only in patients with basal like breast cancer. This appears to be in part due to the role of annexin A1 in activating mTOR-pS6 pathway.
Collapse
|
45
|
Lu Q, Wang J, Yu G, Guo T, Hu C, Ren P. Expression and clinical significance of mammalian target of rapamycin/P70 ribosomal protein S6 kinase signaling pathway in human colorectal carcinoma tissue. Oncol Lett 2015; 10:277-282. [PMID: 26171014 DOI: 10.3892/ol.2015.3228] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Accepted: 02/11/2015] [Indexed: 11/06/2022] Open
Abstract
The activation of mammalian target of rapamycin (mTOR) has been reported in tumor development, but the role of mTOR in colorectal carcinomas remains unclear. The aim of the present study was to investigate the significance of mTOR and its downstream effector 70 kDa ribosomal protein S6 kinase (P70S6K) in human colorectal carcinomas. The phosphorylated (p-)mTOR and p-P70S6K proteins were examined by immunohistochemistry performed on tissue microarray containing tissue samples obtained from colorectal carcinoma (n=111), adenomatous polyps (n=40) and normal colonic mucosa (n=40), with a comparison between the expression of these proteins and the clinicopathological parameters of patients with carcinomas. The positive expression rates of p-mTOR and p-P70S6k were 60.4 and 65.8%, respectively, in colorectal carcinoma tissue, which was significantly increased compared with the tissue from adenomatous polyps (27.5 and 20%, respectively) and normal colonic mucosa (10.0 and 5.0%, respectively) (P<0.05). Overexpression of the p-mTOR and p-P70S6K proteins was significantly associated with the tumor-node-metastasis stage, the occurrence of distal and lymph node metastasis and the degree of differentiation. Aberrant expression of p-mTOR and p-P70S6K may contribute to the pathogenesis, growth, invasion and metastasis of colorectal carcinoma. The phosphorylation of these proteins was considered to be a promising marker to indicate the aggressive behaviors and prognosis of colorectal carcinomas. The overexpression of the mTOR/P70S6K signaling pathway may play an important role in colorectal carcinoma carcinogenesis. The expression of p-mTOR and p-P70S6K was considered as a promising marker to indicate the aggressive behaviors and prognosis of human colorectal carcinomas.
Collapse
Affiliation(s)
- Qingjun Lu
- Department of General Surgery, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong, P.R. China
| | - Jieshu Wang
- Department of General Surgery, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong, P.R. China
| | - Gang Yu
- Department of General Surgery, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong, P.R. China
| | - Tianhua Guo
- Department of General Surgery, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong, P.R. China
| | - Chun Hu
- Department of Oncology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Peng Ren
- Department of General Surgery, Binzhou Central Hospital, Binzhou Medical College, Binzhou, Shandong, P.R. China
| |
Collapse
|
46
|
Li CY, Li X, Liu SF, Qu WS, Wang W, Tian DS. Inhibition of mTOR pathway restrains astrocyte proliferation, migration and production of inflammatory mediators after oxygen–glucose deprivation and reoxygenation. Neurochem Int 2015; 83-84:9-18. [DOI: 10.1016/j.neuint.2015.03.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/02/2015] [Accepted: 03/05/2015] [Indexed: 12/01/2022]
|
47
|
Henkels KM, Mallets ER, Dennis PB, Gomez-Cambronero J. S6K is a morphogenic protein with a mechanism involving Filamin-A phosphorylation and phosphatidic acid binding. FASEB J 2014; 29:1299-313. [PMID: 25512366 DOI: 10.1096/fj.14-260992] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/18/2014] [Indexed: 01/13/2023]
Abstract
Change of cell shape in vivo plays many roles that are central to life itself, such as embryonic development, inflammation, wound healing, and pathologic processes such as cancer metastasis. Nonetheless, the spatiotemporal mechanisms that control the concerted regulation of cell shape remain understudied. Here, we show that ribosomal S6K, which is normally considered a protein involved in protein translation, is a morphogenic protein. Its presence in cells alters the overall organization of the cell surface and cell circularity [(4π × area)/(perimeter)(2)] from 0.47 ± 0.06 units in mock-treated cells to 0.09 ± 0.03 units in S6K-overexpressing macrophages causing stellation and arborization of cell shape. This effect was partially reversed in cells expressing a kinase-inactive S6K mutant and was fully reversed in cells silenced with small interference RNA. Equally important is that S6K is itself regulated by phospholipids, specifically phosphatidic acid, whereby 300 nM 1,2-dioleoyl-sn-glycero-3-phosphate (DOPA), but not the control 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), binds directly to S6K and causes an ∼ 2.9-fold increase in S6K catalytic activity. This was followed by an increase in Filamin A (FLNA) functionality as measured by phospho-FLNA (S(2152)) expression and by a subsequent elevation of actin nucleation. This reliance of S6K on phosphatidic acid (PA), a curvature-inducing phospholipid, explained the extra-large perimeter of cells that overexpressed S6K. Furthermore, the diversity of the response to S6K in several unrelated cell types (fibroblasts, leukocytes, and invasive cancer cells) that we report here indicates the existence of an underlying common mechanism in mammalian cells. This new signaling set, PA-S6K-FLNA-actin, sheds light for the first time into the morphogenic pathway of cytoskeletal structures that are crucial for adhesion and cell locomotion during inflammation and metastasis.
Collapse
Affiliation(s)
- Karen M Henkels
- *Wright State University School of Medicine, Department of Biochemistry and Molecular Biology, Dayton, Ohio, USA; and Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Ohio, USA
| | - Elizabeth R Mallets
- *Wright State University School of Medicine, Department of Biochemistry and Molecular Biology, Dayton, Ohio, USA; and Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Ohio, USA
| | - Patrick B Dennis
- *Wright State University School of Medicine, Department of Biochemistry and Molecular Biology, Dayton, Ohio, USA; and Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Ohio, USA
| | - Julian Gomez-Cambronero
- *Wright State University School of Medicine, Department of Biochemistry and Molecular Biology, Dayton, Ohio, USA; and Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Ohio, USA
| |
Collapse
|
48
|
Tang Y, Vater C, Jacobi A, Liebers C, Zou X, Stiehler M. Salidroside exerts angiogenic and cytoprotective effects on human bone marrow-derived endothelial progenitor cells via Akt/mTOR/p70S6K and MAPK signalling pathways. Br J Pharmacol 2014; 171:2440-56. [PMID: 24471788 DOI: 10.1111/bph.12611] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 01/12/2014] [Accepted: 01/23/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE With the increase of age, increased susceptibility to apoptosis and senescence may contribute to proliferative and functional impairment of endothelial progenitor cells (EPCs). The aim of this study was to investigate whether salidroside (SAL) can induce angiogenic differentiation and inhibit oxidative stress-induced apoptosis in bone marrow-derived EPCs (BM-EPCs), and if so, through what mechanism. EXPERIMENTAL APPROACH BM-EPCs were isolated and treated with different concentrations of SAL for up to 4 days. Cell proliferation, migration and tube formation ability were detected by DNA content quantification, transwell assay and Matrigel-based angiogenesis assay. Gene and protein expression were assessed by qRT-PCR and Western blot respectively. KEY RESULTS Treatment with SAL promoted cellular proliferation and angiogenic differentiation of BM-EPCs, and increased VEGF and NO secretion, which in turn mediated the enhanced angiogenic differentiation of BM-EPCs. Furthermore, SAL significantly attenuated hydrogen peroxide (H₂O₂)-induced cell apoptosis, reduced the intracellular level of reactive oxygen species and restored the mitochondrial membrane potential of BM-EPCs. Moreover, SAL stimulated the phosphorylation of Akt, mammalian target of rapamycin and p70 S6 kinase, as well as ERK1/2, which is associated with cell migration and capillary tube formation. Additionally, SAL reversed the phosphorylation of JNK and p38 MAPK induced by H₂O₂ and suppressed the changes in the Bax/Bcl-xL ratio observed after stimulation with H₂O₂. CONCLUSIONS AND IMPLICATIONS These findings identify novel mechanisms that regulate EPC function and suggest that SAL has therapeutic potential as a new agent to enhance vasculogenesis as well as protect against oxidative endothelial injury.
Collapse
Affiliation(s)
- Yubo Tang
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany; Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
p70 S6 kinase (p70S6K), a member of the AGC serine/threonine kinase family, was initially identified as a key player, together with its downstream effector S6, in the regulation of cellular growth and survival. The p70S6K protein has emerged in recent years as a multifunctional protein which also regulates the actin cytoskeleton and thus plays a role in cell migration. This new function is through two important activities of p70S6K, namely actin cross-linking and Rac1 and Cdc42 activation. The testis is critically dependent on an intricate balance of fundamental cellular processes such as adhesion, migration, and differentiation. It is increasingly evident that Rho GTPases and actin binding proteins play fundamental roles in regulating spermatogenesis within the testis. In this review, we will discuss current findings of p70S6K in the control of actin cytoskeleton dynamics. In addition, the potential role of p70S6K in spermatogenesis and testicular function will be highlighted.
Collapse
Affiliation(s)
- Carman K M Ip
- School of Biological Sciences; University of Hong Kong; Hong Kong, China
| | | |
Collapse
|
50
|
Activation of Akt/mTOR pathway is associated with poor prognosis of nasopharyngeal carcinoma. PLoS One 2014; 9:e106098. [PMID: 25165983 PMCID: PMC4148345 DOI: 10.1371/journal.pone.0106098] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 07/23/2014] [Indexed: 12/21/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor of the head and neck region, which frequently occurs in Southeast Asia, especially in the south of China. It is known that the mammalian target of rapamycin (mTOR) pathway plays a central role in regulating cellular functions, including proliferation, growth, survival, mobility, and angiogenesis. Aberrant expression of the mTOR signaling pathway molecules has been found in many types of cancer. However, whether the alterations of p-Akt, p-p70S6K and p-4EBP1 protein expression are associated with clinicopathological features and prognostic implications in NPC have not been reported. The purposes of the present study are to investigate the association between the expression of p-Akt, p-p70S6K and p-4EBP1 proteins and clinicopathological features in NPC by immunohistochemistry. The results showed that the positive percentage of p-Akt, p-p70S6K and p-4EBP1 proteins expression in NPC (47.2%, 73.0% and 61.7%, respectively) was significantly higher than that in the non-cancerous nasopharyngeal control tissue (33.3%, 59.1% and 47.0%, respectively). There was a significantly higher positive expression of p-Akt in undifferentiated non-keratinizing nasopharyngeal carcinoma than that in differentiated non-keratinizing nasopharyngeal carcinoma (P = 0.014). Additionally, positive expression of p-p70S6K and p-4EBP1 proteins, and positive expression of either of p-Akt, p-p70S6K and p-4EBP1 were significantly correlated inversely with overall survival rates of NPC patients (P = 0.023, P = 0.033, P = 0.008, respectively). Spearman’s rank correlation test showed that expression of p-Akt in NPC was significantly associated with expression of p-p70S6K (r = 0.263, P<0.001) and p-4EBP1(r = 0.284, P<0.001). Also there was an obviously positive association between expression of p-p70S6K and p-4EBP1 proteins in NPC (r = 0.286, P<0.001). Multivariate Cox regression analysis further identified positive expression of p-4EBP1 and p-p70S6K proteins were the independent poor prognostic factors for NPC (P = 0.043, P = 0.027, respectively). Taken together, high expression of p-p70S6K and p-4EBP1 proteins may act as valuable independent biomarkers to predict a poor prognosis of NPC.
Collapse
|