151
|
Sticz T, Molnár A, Márk Á, Hajdu M, Nagy N, Végső G, Micsik T, Kopper L, Sebestyén A. mTOR activity and its prognostic significance in human colorectal carcinoma depending on C1 and C2 complex-related protein expression. J Clin Pathol 2017; 70:410-416. [PMID: 27729429 DOI: 10.1136/jclinpath-2016-203913] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/13/2016] [Accepted: 09/23/2016] [Indexed: 12/19/2022]
Abstract
AIMS Tumour heterogeneity and altered activation of signalling pathways play important roles in therapy resistance. The PI3K/Akt/mTOR signalling network is a well-known regulator of several functions that contribute to tumour growth. mTOR exists in two functionally different multiprotein complexes. We aimed to determine mTOR activity-related proteins in clinically followed, conventionally treated colon carcinomas and to analyse the correlation between clinical data and mTORC1 and mTORC2 activity. METHODS Immunohistochemistry was performed with different antibodies on tissue microarray blocks from 103 patients with human colorectal adenocarcinoma. mTORC1- and mTORC2-related activity were scored on different stainings including analysis of the expression of Raptor and Rictor-specific elements of mTORC1 and C2 complexes. The staining scores and clinical/survival data were compared and analysed. RESULTS Detailed characterisation showed stage and grade independent high mTOR activity in 74% of cases. High mTOR activity was present in mTORC1 and/or mTORC2 complexes; >60% of cases had mTORC2-related high mTOR activity. Based on our analysis, high mTOR activity and Rictor overexpression could be markers of a bad prognosis. Combined phosphoprotein and Rictor/Raptor expression evaluation revealed even stronger statistical correlation with prognosis. CONCLUSIONS The presented staining panel could be appropriate and highly recommended for the accurate specification of mTORC1 and C2 activity of tumour tissues. This could help in the selection of mTOR inhibitors and can provide information about prognosis, which may guide decisions about the intensity of therapy.
Collapse
Affiliation(s)
- Tamás Sticz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Anna Molnár
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ágnes Márk
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Melinda Hajdu
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Noémi Nagy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gyula Végső
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Tamás Micsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - László Kopper
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
- Tumor Progression Research Group of Joint Research Organization of Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| |
Collapse
|
152
|
Ebrahim HY, Akl MR, Elsayed HE, Hill RA, El Sayed KA. Usnic Acid Benzylidene Analogues as Potent Mechanistic Target of Rapamycin Inhibitors for the Control of Breast Malignancies. JOURNAL OF NATURAL PRODUCTS 2017; 80:932-952. [PMID: 28245124 DOI: 10.1021/acs.jnatprod.6b00917] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
(+)-Usnic acid (1) is a common bioactive lichen-derived secondary metabolite with a characteristic dibenzofuran scaffold. It displayed low micromolar antiproliferative activity levels and, notably, induced autophagy in a panel of diverse breast cancer cell lines, suggesting the mechanistic (formerly "mammalian") target of rapamycin (mTOR) as a potential macromolecular target. The cellular autophagic markers were significantly upregulated due to the inhibition of mTOR downstream effectors. Additionally, 1 showed an optimal binding pose at the mTOR kinase pocket aided by multiple interactions to critical amino acids. Rationally designed benzylidene analogues of 1 displayed excellent fitting into a targeted deep hydrophobic pocket at the core of the kinase cleft, through stacking with the phenolic side chain of the Tyr2225 residue. Several potent analogues were generated, including 52, that exhibited potent (nM concentrations) antiproliferative, antimigratory, and anti-invasive activities against cells from multiple breast cancer clonal lines, without affecting the nontumorigenic MCF-10A mammary epithelial cells. Analogue 52 also exhibited potent mTOR inhibition and autophagy induction. Furthermore, 52 showed potent in vivo antitumor activity in two athymic nude mice breast cancer xenograft models. Collectively, usnic acid and analogues are potential lead mTOR inhibitors appropriate for future use to control breast malignancies.
Collapse
Affiliation(s)
- Hassan Y Ebrahim
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe , Monroe, Louisiana 71209, United States
| | - Mohamed R Akl
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe , Monroe, Louisiana 71209, United States
| | - Heba E Elsayed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe , Monroe, Louisiana 71209, United States
| | - Ronald A Hill
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe , Monroe, Louisiana 71209, United States
| | - Khalid A El Sayed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe , Monroe, Louisiana 71209, United States
| |
Collapse
|
153
|
Ruf S, Heberle AM, Langelaar-Makkinje M, Gelino S, Wilkinson D, Gerbeth C, Schwarz JJ, Holzwarth B, Warscheid B, Meisinger C, van Vugt MATM, Baumeister R, Hansen M, Thedieck K. PLK1 (polo like kinase 1) inhibits MTOR complex 1 and promotes autophagy. Autophagy 2017; 13:486-505. [PMID: 28102733 PMCID: PMC5361591 DOI: 10.1080/15548627.2016.1263781] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/09/2016] [Accepted: 11/16/2016] [Indexed: 02/08/2023] Open
Abstract
Mechanistic target of rapamycin complex 1 (MTORC1) and polo like kinase 1 (PLK1) are major drivers of cancer cell growth and proliferation, and inhibitors of both protein kinases are currently being investigated in clinical studies. To date, MTORC1's and PLK1's functions are mostly studied separately, and reports on their mutual crosstalk are scarce. Here, we identify PLK1 as a physical MTORC1 interactor in human cancer cells. PLK1 inhibition enhances MTORC1 activity under nutrient sufficiency and in starved cells, and PLK1 directly phosphorylates the MTORC1 component RPTOR/RAPTOR in vitro. PLK1 and MTORC1 reside together at lysosomes, the subcellular site where MTORC1 is active. Consistent with an inhibitory role of PLK1 toward MTORC1, PLK1 overexpression inhibits lysosomal association of the PLK1-MTORC1 complex, whereas PLK1 inhibition promotes lysosomal localization of MTOR. PLK1-MTORC1 binding is enhanced by amino acid starvation, a condition known to increase autophagy. MTORC1 inhibition is an important step in autophagy activation. Consistently, PLK1 inhibition mitigates autophagy in cancer cells both under nutrient starvation and sufficiency, and a role of PLK1 in autophagy is also observed in the invertebrate model organism Caenorhabditis elegans. In summary, PLK1 inhibits MTORC1 and thereby positively contributes to autophagy. Since autophagy is increasingly recognized to contribute to tumor cell survival and growth, we propose that cautious monitoring of MTORC1 and autophagy readouts in clinical trials with PLK1 inhibitors is needed to develop strategies for optimized (combinatorial) cancer therapies targeting MTORC1, PLK1, and autophagy.
Collapse
Affiliation(s)
- Stefanie Ruf
- Department of Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, AV Groningen, The Netherlands
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Research Training Group (RTG) 1104, University of Freiburg, Freiburg, Germany
| | - Alexander Martin Heberle
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, AV Groningen, The Netherlands
| | - Miriam Langelaar-Makkinje
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, AV Groningen, The Netherlands
| | - Sara Gelino
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Deepti Wilkinson
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Carolin Gerbeth
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- ZBMZ Centre for Biochemistry and Molecular Cell Research (Faculty of Medicine), University of Freiburg, Freiburg, Germany
- Institute of Biochemistry and Molecular Biology (Faculty of Medicine), University of Freiburg, Freiburg, Germany
| | - Jennifer Jasmin Schwarz
- Department of Biochemistry and Functional Proteomics, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Birgit Holzwarth
- Department of Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Bettina Warscheid
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Department of Biochemistry and Functional Proteomics, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Chris Meisinger
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- ZBMZ Centre for Biochemistry and Molecular Cell Research (Faculty of Medicine), University of Freiburg, Freiburg, Germany
- Institute of Biochemistry and Molecular Biology (Faculty of Medicine), University of Freiburg, Freiburg, Germany
| | - Marcel A. T. M. van Vugt
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, GZ Groningen, The Netherlands
| | - Ralf Baumeister
- Department of Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Research Training Group (RTG) 1104, University of Freiburg, Freiburg, Germany
- ZBMZ Centre for Biochemistry and Molecular Cell Research (Faculty of Medicine), University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Malene Hansen
- Program of Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Kathrin Thedieck
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, AV Groningen, The Netherlands
- Department for Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
154
|
Resistance to mTORC1 Inhibitors in Cancer Therapy: From Kinase Mutations to Intratumoral Heterogeneity of Kinase Activity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1726078. [PMID: 28280521 PMCID: PMC5322438 DOI: 10.1155/2017/1726078] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/22/2017] [Indexed: 01/19/2023]
Abstract
Targeting mTORC1 has been thoroughly explored in cancer therapy. Following encouraging preclinical studies, mTORC1 inhibitors however failed to provide substantial benefits in cancer patients. Several resistance mechanisms have been identified including mutations of mTOR and activation of alternate proliferation pathways. Moreover, emerging evidence discloses intratumoral heterogeneity of mTORC1 activity that further contributes to a reduced anticancer efficacy of mTORC1 inhibitors. Genetic heterogeneity as well as heterogeneous conditions of the tumor environment such as hypoxia profoundly modifies mTORC1 activity in tumors and hence influences the response of tumors to mTORC1 inhibitors. Intriguingly, the heterogeneity of mTORC1 activity also occurs towards its substrates at the single cell level, as mutually exclusive pattern of activation of mTORC1 downstream effectors has been reported in tumors. After briefly describing mTORC1 biology and the use of mTORC1 inhibitors in patients, this review will give an overview on concepts of resistance to mTORC1 inhibition in cancer with a particular focus on intratumoral heterogeneity of mTORC1 activity.
Collapse
|
155
|
Mao B, Gao S, Weng Y, Zhang L, Zhang L. Design, synthesis, and biological evaluation of imidazo[1,2-b]pyridazine derivatives as mTOR inhibitors. Eur J Med Chem 2017; 129:135-150. [PMID: 28235701 DOI: 10.1016/j.ejmech.2017.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/18/2022]
Abstract
ATP-competitive mTOR inhibitors have been studied as potential antitumor agents. Based on the structure-activity relationship of known mTOR inhibitors, a series of novel imidazo[1,2-b]pyridazine derivatives were synthesized and characterized. The anti-proliferative activities of these compounds were evaluated by SRB assay against six human cancer cell lines. Imidazo[1,2-b]pyridazine diaryl urea derivatives A15-A24 exhibited significant anti-proliferative activity especially against non-small cell lung cancer A549 and H460 with IC50 values ranging from 0.02 μM to 20.7 μM. Among them, compounds A17 and A18 showed mTOR inhibitory activity with IC50 of 0.067 μM and 0.062 μM, respectively. A more detailed analysis of compounds A17 and A18 showed that they induced G1-phase cell cycle arrest and suppressed the phosphorylation of AKT and S6 at cellular level. Moreover, obvious anticancer effect of A17 in vivo was observed in established nude mice A549 xenograft model.
Collapse
Affiliation(s)
- Beibei Mao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Shanyun Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Yiran Weng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China.
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| |
Collapse
|
156
|
Slator C, Molphy Z, McKee V, Kellett A. Triggering autophagic cell death with a di-manganese(II) developmental therapeutic. Redox Biol 2017; 12:150-161. [PMID: 28236767 PMCID: PMC5328722 DOI: 10.1016/j.redox.2017.01.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/07/2017] [Accepted: 01/11/2017] [Indexed: 12/16/2022] Open
Abstract
There is an unmet need for novel metal-based chemotherapeutics with alternative modes of action compared to clinical agents such as cisplatin and metallo-bleomycin. Recent attention in this field has focused on designing intracellular ROS-mediators as powerful cytotoxins of human cancers and identifying potentially unique toxic mechanisms underpinning their utility. Herein, we report the developmental di-manganese(II) therapeutic [Mn2(μ-oda)(phen)4(H2O)2][Mn2(μ-oda)(phen)4(oda)2]·4H2O (Mn-Oda) induces autophagy-promoted apoptosis in human ovarian cancer cells (SKOV3). The complex was initially identified to intercalate DNA by topoisomerase I unwinding and circular dichroism spectroscopy. Intracellular DNA damage, detected by γH2AX and the COMET assay, however, is not linked to direct Mn-Oda free radical generation, but is instead mediated through the promotion of intracellular reactive oxygen species (ROS) leading to autophagic vacuole formation and downstream nuclear degradation. To elucidate the cytotoxic profile of Mn-Oda, a wide range of biomarkers specific to apoptosis and autophagy including caspase release, mitochondrial membrane integrity, fluorogenic probe localisation, and cell cycle analysis were employed. Through these techniques, the activity of Mn-Oda was compared directly to i.) the pro-apoptotic clinical anticancer drug doxorubicin, ii.) the multimodal histone deacetylase inhibitor suberoyanilide hydroxamic acid, and iii.) the autophagy inducer rapamycin. In conjunction with ROS-specific trapping agents and established inhibitors of autophagy, we have identified autophagy-induction linked to mitochondrial superoxide production, with confocal image analysis of SKOV3 cells further supporting autophagosome formation.
Collapse
Affiliation(s)
- Creina Slator
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Zara Molphy
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Vickie McKee
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Andrew Kellett
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
157
|
Felici R, Buonvicino D, Muzzi M, Cavone L, Guasti D, Lapucci A, Pratesi S, De Cesaris F, Luceri F, Chiarugi A. Post onset, oral rapamycin treatment delays development of mitochondrial encephalopathy only at supramaximal doses. Neuropharmacology 2017; 117:74-84. [PMID: 28161373 DOI: 10.1016/j.neuropharm.2017.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 01/16/2017] [Accepted: 01/31/2017] [Indexed: 10/20/2022]
Abstract
Mitochondrial encephalopathies are fatal, infantile neurodegenerative disorders caused by a deficit of mitochondrial functioning, for which there is urgent need to identify efficacious pharmacological treatments. Recent evidence shows that rapamycin administered both intraperitoneally or in the diet delays disease onset and enhances survival in the Ndufs4 null mouse model of mitochondrial encephalopathy. To delineate the clinical translatability of rapamycin in treatment of mitochondrial encephalopathy, we evaluated the drug's effects on disease evolution and mitochondrial parameters adopting treatment paradigms with fixed daily, oral doses starting at symptom onset in Ndufs4 knockout mice. Molecular mechanisms responsible for the pharmacodynamic effects of rapamycin were also evaluated. We found that rapamycin did not affect disease development at clinically-relevant doses (0.5 mg kg-1). Conversely, an oral dose previously adopted for intraperitoneal administration (8 mg kg-1) delayed development of neurological symptoms and increased median survival by 25%. Neurological improvement and lifespan were not further increased when the dose raised to 20 mg kg-1. Notably, rapamycin at 8 mg kg-1 did not affect the reduced expression of respiratory complex subunits, as well as mitochondrial number and mtDNA content. This treatment regimen however significantly ameliorated architecture of mitochondria cristae in motor cortex and cerebellum. However, reduction of mTOR activity by rapamycin was not consistently found within the brain of knockout mice. Overall, data show the ability of rapamycin to improve ultrastructure of dysfunctional mitochondria and corroborate its therapeutic potential in mitochondrial disorders. The non-clinical standard doses required, however, raise concerns about its rapid and safe clinical transferability.
Collapse
Affiliation(s)
- Roberta Felici
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Leonardo Cavone
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy
| | - Andrea Lapucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Sara Pratesi
- Department of Experimental and Clinical Medicine, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy
| | - Francesco De Cesaris
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Francesca Luceri
- General Laboratory Unit (Pharmacology Unit), Careggi University Hospital, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| |
Collapse
|
158
|
Induction of dormancy in hypoxic human papillomavirus-positive cancer cells. Proc Natl Acad Sci U S A 2017; 114:E990-E998. [PMID: 28115701 DOI: 10.1073/pnas.1615758114] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Oncogenic human papillomaviruses (HPVs) are closely linked to major human malignancies, including cervical and head and neck cancers. It is widely assumed that HPV-positive cancer cells are under selection pressure to continuously express the viral E6/E7 oncogenes, that their intracellular p53 levels are reconstituted on E6/E7 repression, and that E6/E7 inhibition phenotypically results in cellular senescence. Here we show that hypoxic conditions, as are often found in subregions of cervical and head and neck cancers, enable HPV-positive cancer cells to escape from these regulatory principles: E6/E7 is efficiently repressed, yet, p53 levels do not increase. Moreover, E6/E7 repression under hypoxia does not result in cellular senescence, owing to hypoxia-associated impaired mechanistic target of rapamycin (mTOR) signaling via the inhibitory REDD1/TSC2 axis. Instead, a reversible growth arrest is induced that can be overcome by reoxygenation. Impairment of mTOR signaling also interfered with the senescence response of hypoxic HPV-positive cancer cells toward prosenescent chemotherapy in vitro. Collectively, these findings indicate that hypoxic HPV-positive cancer cells can induce a reversible state of dormancy, with decreased viral antigen synthesis and increased therapeutic resistance, and may serve as reservoirs for tumor recurrence on reoxygenation.
Collapse
|
159
|
Villar VH, Nguyen TL, Delcroix V, Terés S, Bouchecareilh M, Salin B, Bodineau C, Vacher P, Priault M, Soubeyran P, Durán RV. mTORC1 inhibition in cancer cells protects from glutaminolysis-mediated apoptosis during nutrient limitation. Nat Commun 2017; 8:14124. [PMID: 28112156 PMCID: PMC5264013 DOI: 10.1038/ncomms14124] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 12/01/2016] [Indexed: 12/16/2022] Open
Abstract
A master coordinator of cell growth, mTORC1 is activated by different metabolic inputs, particularly the metabolism of glutamine (glutaminolysis), to control a vast range of cellular processes, including autophagy. As a well-recognized tumour promoter, inhibitors of mTORC1 such as rapamycin have been approved as anti-cancer agents, but their overall outcome in patients is rather poor. Here we show that mTORC1 also presents tumour suppressor features in conditions of nutrient restrictions. Thus, the activation of mTORC1 by glutaminolysis during nutritional imbalance inhibits autophagy and induces apoptosis in cancer cells. Importantly, rapamycin treatment reactivates autophagy and prevents the mTORC1-mediated apoptosis. We also observe that the ability of mTORC1 to activate apoptosis is mediated by the adaptor protein p62. Thus, the mTORC1-mediated upregulation of p62 during nutrient imbalance induces the binding of p62 to caspase 8 and the subsequent activation of the caspase pathway. Our data highlight the role of autophagy as a survival mechanism upon rapamycin treatment.
Collapse
Affiliation(s)
- Victor H. Villar
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Tra Ly Nguyen
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Vanessa Delcroix
- Institut Bergonié, INSERM U1218, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Silvia Terés
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Marion Bouchecareilh
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, 1 Rue Camille Saint-Saëns, Bordeaux 33077, France
| | - Bénédicte Salin
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, 1 Rue Camille Saint-Saëns, Bordeaux 33077, France
| | - Clément Bodineau
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| | - Pierre Vacher
- Institut Bergonié, INSERM U1218, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Muriel Priault
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, 1 Rue Camille Saint-Saëns, Bordeaux 33077, France
| | - Pierre Soubeyran
- Institut Bergonié, INSERM U1218, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Raúl V. Durán
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, 2 Rue Robert Escarpit, Pessac 33607, France
| |
Collapse
|
160
|
Mediani L, Gibellini F, Bertacchini J, Frasson C, Bosco R, Accordi B, Basso G, Bonora M, Calabrò ML, Mattiolo A, Sgarbi G, Baracca A, Pinton P, Riva G, Rampazzo E, Petrizza L, Prodi L, Milani D, Luppi M, Potenza L, De Pol A, Cocco L, Capitani S, Marmiroli S. Reversal of the glycolytic phenotype of primary effusion lymphoma cells by combined targeting of cellular metabolism and PI3K/Akt/ mTOR signaling. Oncotarget 2016; 7:5521-37. [PMID: 26575168 PMCID: PMC4868703 DOI: 10.18632/oncotarget.6315] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/27/2015] [Indexed: 12/12/2022] Open
Abstract
PEL is a B-cell non-Hodgkin lymphoma, occurring predominantly as a lymphomatous effusion in body cavities, characterized by aggressive clinical course, with no standard therapy. Based on previous reports that PEL cells display a Warburg phenotype, we hypothesized that the highly hypoxic environment in which they grow in vivo makes them more reliant on glycolysis, and more vulnerable to drugs targeting this pathway. We established here that indeed PEL cells in hypoxia are more sensitive to glycolysis inhibition. Furthermore, since PI3K/Akt/mTOR has been proposed as a drug target in PEL, we ascertained that pathway-specific inhibitors, namely the dual PI3K and mTOR inhibitor, PF-04691502, and the Akt inhibitor, Akti 1/2, display improved cytotoxicity to PEL cells in hypoxic conditions. Unexpectedly, we found that these drugs reduce lactate production/extracellular acidification rate, and, in combination with the glycolysis inhibitor 2-deoxyglucose (2-DG), they shift PEL cells metabolism from aerobic glycolysis towards oxidative respiration. Moreover, the associations possess strong synergistic cytotoxicity towards PEL cells, and thus may reduce adverse reaction in vivo, while displaying very low toxicity to normal lymphocytes. Finally, we showed that the association of 2-DG and PF-04691502 maintains its cytotoxic and proapoptotic effect also in PEL cells co-cultured with human primary mesothelial cells, a condition known to mimic the in vivo environment and to exert a protective and pro-survival action. All together, these results provide a compelling rationale for the clinical development of new therapies for the treatment of PEL, based on combined targeting of glycolytic metabolism and constitutively activated signaling pathways.
Collapse
Affiliation(s)
- Laura Mediani
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Federica Gibellini
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Jessika Bertacchini
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy.,Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Chiara Frasson
- Department of Woman's and Child's Health and Institute of Pediatric Research - Città della Speranza Foundation, University of Padova, Padova, Italy
| | - Raffaella Bosco
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Benedetta Accordi
- Department of Woman's and Child's Health and Institute of Pediatric Research - Città della Speranza Foundation, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Department of Woman's and Child's Health and Institute of Pediatric Research - Città della Speranza Foundation, University of Padova, Padova, Italy
| | - Massimo Bonora
- Department of Morphology, Surgery and Experimental Medicine Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Maria Luisa Calabrò
- Immunology and Molecular Oncology, Veneto Institute of Oncology, IOV IRCCS, Padova, Italy
| | - Adriana Mattiolo
- Immunology and Molecular Oncology, Veneto Institute of Oncology, IOV IRCCS, Padova, Italy
| | - Gianluca Sgarbi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Baracca
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Giovanni Riva
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Enrico Rampazzo
- Department of Chemistry, University of Bologna, Bologna, Italy
| | - Luca Petrizza
- Department of Chemistry, University of Bologna, Bologna, Italy
| | - Luca Prodi
- Department of Chemistry, University of Bologna, Bologna, Italy
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Mario Luppi
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Leonardo Potenza
- Department of Medical and Surgical Sciences, Section of Hematology, University of Modena and Reggio Emilia, AOU Policlinico, Modena, Italy
| | - Anto De Pol
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, Section of Anatomy and Histology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
161
|
Vernieri C, Casola S, Foiani M, Pietrantonio F, de Braud F, Longo V. Targeting Cancer Metabolism: Dietary and Pharmacologic Interventions. Cancer Discov 2016; 6:1315-1333. [PMID: 27872127 DOI: 10.1158/2159-8290.cd-16-0615] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 12/15/2022]
Abstract
Most tumors display oncogene-driven reprogramming of several metabolic pathways, which are crucial to sustain their growth and proliferation. In recent years, both dietary and pharmacologic approaches that target deregulated tumor metabolism are beginning to be considered for clinical applications. Dietary interventions exploit the ability of nutrient-restricted conditions to exert broad biological effects, protecting normal cells, organs, and systems, while sensitizing a wide variety of cancer cells to cytotoxic therapies. On the other hand, drugs targeting enzymes or metabolites of crucial metabolic pathways can be highly specific and effective, but must be matched with a responsive tumor, which might rapidly adapt. In this review, we illustrate how dietary and pharmacologic therapies differ in their effect on tumor growth, proliferation, and metabolism and discuss the available preclinical and clinical evidence in favor of or against each of them. We also indicate, when appropriate, how to optimize future investigations on metabolic therapies on the basis of tumor- and patient-related characteristics. SIGNIFICANCE To our knowledge, this is the first review article that comprehensively analyzes the preclinical and preliminary clinical experimental foundations of both dietary and pharmacologic metabolic interventions in cancer therapy. Among several promising therapies, we propose treatment personalization on the basis of tumor genetics, tumor metabolism, and patient systemic metabolism.Cancer Discov; 6(12); 1315-33. ©2016 AACR.
Collapse
Affiliation(s)
- Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy. .,Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - Stefano Casola
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - Marco Foiani
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy.,Universita' degli Studi di Milano, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.,Universita' degli Studi di Milano, Milan, Italy
| | - Valter Longo
- Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy. .,Longevity Institute, Davis School of Gerontology, University of Southern California, Los Angeles, California
| |
Collapse
|
162
|
Yoon S, Kim JH, Kim SE, Kim C, Tran PT, Ann J, Koh Y, Jang J, Kim S, Moon HS, Kim WK, Lee S, Lee J, Kim S, Lee J. Discovery of Leucyladenylate Sulfamates as Novel Leucyl-tRNA Synthetase (LRS)-Targeted Mammalian Target of Rapamycin Complex 1 (mTORC1) Inhibitors. J Med Chem 2016; 59:10322-10328. [PMID: 27933890 DOI: 10.1021/acs.jmedchem.6b01190] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent studies indicate that LRS may act as a leucine sensor for the mTORC1 pathway, potentially providing an alternative strategy to overcome rapamycin resistance in cancer treatments. In this study, we developed leucyladenylate sulfamate derivatives as LRS-targeted mTORC1 inhibitors. Compound 18 selectively inhibited LRS-mediated mTORC1 activation and exerted specific cytotoxicity against colon cancer cells with a hyperactive mTORC1, suggesting that 18 may offer a novel treatment option for human colorectal cancer.
Collapse
Affiliation(s)
- Suyoung Yoon
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Jong Hyun Kim
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Sung-Eun Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Changhoon Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Phuong-Thao Tran
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Jihyae Ann
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Yura Koh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Jayun Jang
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Sungmin Kim
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Hee-Sun Moon
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Won Kyung Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Sangkook Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| | - Jiyoun Lee
- Department of Global Medical Science, Sungshin University , Seoul 142-732, Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University , Seoul 151-742, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University , Seoul 151-742, Korea
| | - Jeewoo Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University , Seoul 151-742, Korea
| |
Collapse
|
163
|
Maiese K. Targeting molecules to medicine with mTOR, autophagy and neurodegenerative disorders. Br J Clin Pharmacol 2016; 82:1245-1266. [PMID: 26469771 PMCID: PMC5061806 DOI: 10.1111/bcp.12804] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 10/11/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders are significantly increasing in incidence as the age of the global population continues to climb with improved life expectancy. At present, more than 30 million individuals throughout the world are impacted by acute and chronic neurodegenerative disorders with limited treatment strategies. The mechanistic target of rapamycin (mTOR), also known as the mammalian target of rapamycin, is a 289 kDa serine/threonine protein kinase that offers exciting possibilities for novel treatment strategies for a host of neurodegenerative diseases that include Alzheimer's disease, Parkinson's disease, Huntington's disease, epilepsy, stroke and trauma. mTOR governs the programmed cell death pathways of apoptosis and autophagy that can determine neuronal stem cell development, precursor cell differentiation, cell senescence, cell survival and ultimate cell fate. Coupled to the cellular biology of mTOR are a number of considerations for the development of novel treatments involving the fine control of mTOR signalling, tumourigenesis, complexity of the apoptosis and autophagy relationship, functional outcome in the nervous system, and the intimately linked pathways of growth factors, phosphoinositide 3-kinase (PI 3-K), protein kinase B (Akt), AMP activated protein kinase (AMPK), silent mating type information regulation two homologue one (Saccharomyces cerevisiae) (SIRT1) and others. Effective clinical translation of the cellular signalling mechanisms of mTOR offers provocative avenues for new drug development in the nervous system tempered only by the need to elucidate further the intricacies of the mTOR pathway.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, Newark, New Jersey, 07101, USA.
| |
Collapse
|
164
|
Flaum N, Valle JW, Mansoor W, McNamara MG. Everolimus in the treatment of neuroendocrine tumors of the respiratory and gastroenteropancreatic systems. Future Oncol 2016; 12:2561-2578. [DOI: 10.2217/fon.16.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Neuroendocrine tumors (NETs) are a rare diverse group of malignancies occurring most commonly in the gastroenteropancreatic system and the lungs. The incidence of NETs is increasing worldwide; median survival for patients with metastatic NETs is 5–65 months. A growing body of evidence shows survival benefit in patients with advanced NETs (gastroenteropancreatic and lung) treated with mTOR inhibitor everolimus, with improvement in survival being demonstrated in the clinical trial and real-world setting. Everolimus has been shown to have a manageable safety profile, with the most common adverse events being stomatitis, rash, diarrhea, fatigue and infections. Due to the rarity of the condition, there are challenges in conducting clinical trials in these patients. Further research is required to clarify the role of adjuvant therapy, treatment sequencing and the use of multimodality treatments.
Collapse
Affiliation(s)
- Nicola Flaum
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Juan W Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - Wasat Mansoor
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Mairéad G McNamara
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Institute of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
165
|
Toda K, Kawada K, Iwamoto M, Inamoto S, Sasazuki T, Shirasawa S, Hasegawa S, Sakai Y. Metabolic Alterations Caused by KRAS Mutations in Colorectal Cancer Contribute to Cell Adaptation to Glutamine Depletion by Upregulation of Asparagine Synthetase. Neoplasia 2016; 18:654-665. [PMID: 27764698 PMCID: PMC5071549 DOI: 10.1016/j.neo.2016.09.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 12/28/2022] Open
Abstract
A number of clinical trials have shown that KRAS mutations of colorectal cancer (CRC) can predict a lack of responses to anti-epidermal growth factor receptor-based therapy. Recently, there have been several studies to elucidate metabolism reprogramming in cancer. However, it remains to be investigated how mutated KRAS can coordinate the metabolic shift to sustain CRC tumor growth. In this study, we found that KRAS mutation in CRC caused alteration in amino acid metabolism. KRAS mutation causes a marked decrease in aspartate level and an increase in asparagine level in CRC. Using several human CRC cell lines and clinical specimens of primary CRC, we demonstrated that the expression of asparagine synthetase (ASNS), an enzyme that synthesizes asparagine from aspartate, was upregulated by mutated KRAS and that ASNS expression was induced by KRAS-activated signaling pathway, in particular PI3K-AKT-mTOR pathway. Importantly, we demonstrated that KRAS-mutant CRC cells could become adaptive to glutamine depletion through asparagine biosynthesis by ASNS and that asparagine addition could rescue the inhibited growth and viability of cells grown under the glutamine-free condition in vitro. Notably, a pronounced growth suppression of KRAS-mutant CRC was observed upon ASNS knockdown in vivo. Furthermore, combination of L-asparaginase plus rapamycin markedly suppressed the growth of KRAS-mutant CRC xenografts in vivo, whereas either L-asparaginase or rapamycin alone was not effective. These results indicate ASNS might be a novel therapeutic target against CRCs with mutated KRAS.
Collapse
Affiliation(s)
- Kosuke Toda
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Masayoshi Iwamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Susumu Inamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | | | - Senji Shirasawa
- Departments of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Suguru Hasegawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
166
|
Dao V, Liu Y, Pandeswara S, Svatek RS, Gelfond JA, Liu A, Hurez V, Curiel TJ. Immune-Stimulatory Effects of Rapamycin Are Mediated by Stimulation of Antitumor γδ T Cells. Cancer Res 2016; 76:5970-5982. [PMID: 27569211 PMCID: PMC5065775 DOI: 10.1158/0008-5472.can-16-0091] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 08/01/2016] [Indexed: 11/16/2022]
Abstract
The FDA-approved mTOR inhibitor rapamycin mediates important immune effects, but its contributions to the anticancer effects of the drug are unclear. Here we report evidence that rapamycin-mediated cancer protection relies upon stimulation of γδ T cells. In a well-established mouse model of carcinogen and inflammation-driven skin carcinogenesis, IFNγ recruited γδ TCRmid T cells to the epidermis where rapamycin boosted their perforin-dependent antitumor properties. These antitumor cells were mostly Vγ5-Vγ4-Vγ1- in phenotype. IFNγ signals were required in both hematopoietic and nonhematopoietic cells for rapamycin to optimally promote epidermal infiltration of γδ TCRmid T cells, as mediated by CXCR3-CXCL10 interactions, along with the antitumor effects of these cells. In mouse xenograft models of human squamous cell carcinoma, rapamycin improved human γδ T-cell-mediated cancer cell killing. Our results identify immune mechanisms for the cancer prevention and treatment properties of rapamycin, challenging the paradigm that mTOR inhibition acts primarily by direct action on tumor cells. Cancer Res; 76(20); 5970-82. ©2016 AACR.
Collapse
Affiliation(s)
- Vinh Dao
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio (UTHSCSA), San Antonio, Texas. Department of Medicine, UTHSCSA, San Antonio, Texas
| | - Yang Liu
- Department of Medicine, UTHSCSA, San Antonio, Texas. Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | | | - Robert S Svatek
- Cancer Therapy & Research Center, UTHSCSA, San Antonio, Texas. Department of Urology, UTHSCSA, San Antonio, Texas
| | - Jonathan A Gelfond
- Department of Epidemiology and Biostatistics, UTHSCSA, San Antonio, Texas
| | - Aijie Liu
- Department of Medicine, UTHSCSA, San Antonio, Texas
| | | | - Tyler J Curiel
- Department of Microbiology and Immunology, University of Texas Health Science Center, San Antonio (UTHSCSA), San Antonio, Texas. Department of Medicine, UTHSCSA, San Antonio, Texas. Cancer Therapy & Research Center, UTHSCSA, San Antonio, Texas. Barshop Institute for Longevity and Aging Studies, UTHSCSA, San Antonio, Texas.
| |
Collapse
|
167
|
Tramutola A, Lanzillotta C, Di Domenico F. Targeting mTOR to reduce Alzheimer-related cognitive decline: from current hits to future therapies. Expert Rev Neurother 2016; 17:33-45. [PMID: 27690737 DOI: 10.1080/14737175.2017.1244482] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The mTOR pathway is involved in the regulation of a wide repertoire of cellular functions in the brain and its dysregulation is emerging as a leitmotif in a large number of neurological disorders. In AD, altered mTOR signaling contributes to the inhibition of autophagy deposition of Aβ and tau aggregates and to the alteration of several neuronal metabolic pathways. Areas covered: In this review, we report all the current findings on the use of mTOR inhibitors (rapamycin, rapalogues) in the treatment of AD. These results support the role of mTOR inhibitors as potential therapeutic agents able to reduce AD hallmarks and recover cognitive performances. Expert commentary: Despite mTOR inhibitors appearing to be ideal compounds to counteract AD, further studies are needed in order to gain knowledge on the involvement of aberrant mTOR in AD, and to standardize a valuable therapeutic approach that can be translated to humans.
Collapse
Affiliation(s)
- Antonella Tramutola
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| | - Chiara Lanzillotta
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| | - Fabio Di Domenico
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
168
|
Blau CA, Ramirez AB, Blau S, Pritchard CC, Dorschner MO, Schmechel SC, Martins TJ, Mahen EM, Burton KA, Komashko VM, Radenbaugh AJ, Dougherty K, Thomas A, Miller CP, Annis J, Fromm JR, Song C, Chang E, Howard K, Austin S, Schmidt RA, Linenberger ML, Becker PS, Senecal FM, Mecham BH, Lee SI, Madan A, Ronen R, Dutkowski J, Heimfeld S, Wood BL, Stilwell JL, Kaldjian EP, Haussler D, Zhu J. A Distributed Network for Intensive Longitudinal Monitoring in Metastatic Triple-Negative Breast Cancer. J Natl Compr Canc Netw 2016; 14:8-17. [PMID: 26733551 DOI: 10.6004/jnccn.2016.0003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Accelerating cancer research is expected to require new types of clinical trials. This report describes the Intensive Trial of OMics in Cancer (ITOMIC) and a participant with triple-negative breast cancer metastatic to bone, who had markedly elevated circulating tumor cells (CTCs) that were monitored 48 times over 9 months. A total of 32 researchers from 14 institutions were engaged in the patient's evaluation; 20 researchers had no prior involvement in patient care and 18 were recruited specifically for this patient. Whole-exome sequencing of 3 bone marrow samples demonstrated a novel ROS1 variant that was estimated to be present in most or all tumor cells. After an initial response to cisplatin, a hypothesis of crizotinib sensitivity was disproven. Leukapheresis followed by partial CTC enrichment allowed for the development of a differential high-throughput drug screen and demonstrated sensitivity to investigational BH3-mimetic inhibitors of BCL-2 that could not be tested in the patient because requests to the pharmaceutical sponsors were denied. The number and size of CTC clusters correlated with clinical status and eventually death. Focusing the expertise of a distributed network of investigators on an intensively monitored patient with cancer can generate high-resolution views of the natural history of cancer and suggest new opportunities for therapy. Optimization requires access to investigational drugs.
Collapse
Affiliation(s)
- C Anthony Blau
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington,Department of Medicine/Hematology, University of Washington, Seattle, Washington,Seattle Cancer Care Alliance, Seattle, Washington
| | - Arturo B Ramirez
- Center for Cancer Innovation, University of Washington, Seattle, Washington,RareCyte Inc., Seattle, Washington
| | - Sibel Blau
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Department of Medicine/Hematology, University of Washington, Seattle, Washington,Northwest Medical Specialities, Puyallup and Tacoma, Washington
| | - Colin C Pritchard
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Michael O Dorschner
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Department of Laboratory Medicine, University of Washington, Seattle, Washington,Department of Pathology, University of Washington, Seattle, Washington
| | - Stephen C Schmechel
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Department of Pathology, University of Washington, Seattle, Washington
| | - Timothy J Martins
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington,Quellos High Throughput Screening Core, Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington
| | - Elisabeth M Mahen
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington,Department of Medicine/Hematology, University of Washington, Seattle, Washington
| | - Kimberly A Burton
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington,Department of Medicine/Hematology, University of Washington, Seattle, Washington
| | - Vitalina M Komashko
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Trialomics LLC, Seattle, Washington
| | - Amie J Radenbaugh
- Center for Cancer Innovation, University of Washington, Seattle, Washington,University of California at Santa Cruz, Santa Cruz, California
| | - Katy Dougherty
- Seattle Cancer Care Alliance, Seattle, Washington,Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Anju Thomas
- Seattle Cancer Care Alliance, Seattle, Washington,Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Christopher P Miller
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington,Department of Medicine/Hematology, University of Washington, Seattle, Washington
| | - James Annis
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington,Quellos High Throughput Screening Core, Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington
| | - Jonathan R Fromm
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Chaozhong Song
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Elizabeth Chang
- Department of Medicine/Hematology, University of Washington, Seattle, Washington
| | | | | | - Rodney A Schmidt
- Department of Pathology, University of Washington, Seattle, Washington
| | - Michael L Linenberger
- Department of Medicine/Hematology, University of Washington, Seattle, Washington,Seattle Cancer Care Alliance, Seattle, Washington,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Pamela S Becker
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington,Department of Medicine/Hematology, University of Washington, Seattle, Washington,Seattle Cancer Care Alliance, Seattle, Washington,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Francis M Senecal
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Department of Medicine/Hematology, University of Washington, Seattle, Washington,Northwest Medical Specialities, Puyallup and Tacoma, Washington
| | - Brigham H Mecham
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Trialomics LLC, Seattle, Washington
| | - Su-In Lee
- School of Computer Science and Engineering, University of Washington, Seattle, Washington
| | - Anup Madan
- Covance/LabCorp Inc., Seattle, Washington
| | - Roy Ronen
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Data4Cure Inc., La Jolla, California
| | - Janusz Dutkowski
- Center for Cancer Innovation, University of Washington, Seattle, Washington,Data4Cure Inc., La Jolla, California
| | | | - Brent L Wood
- Seattle Cancer Care Alliance, Seattle, Washington,Department of Laboratory Medicine, University of Washington, Seattle, Washington,Department of Pathology, University of Washington, Seattle, Washington
| | - Jackie L Stilwell
- Center for Cancer Innovation, University of Washington, Seattle, Washington,RareCyte Inc., Seattle, Washington
| | - Eric P Kaldjian
- Center for Cancer Innovation, University of Washington, Seattle, Washington,RareCyte Inc., Seattle, Washington
| | - David Haussler
- Center for Cancer Innovation, University of Washington, Seattle, Washington,University of California at Santa Cruz, Santa Cruz, California
| | - Jingchun Zhu
- Center for Cancer Innovation, University of Washington, Seattle, Washington,University of California at Santa Cruz, Santa Cruz, California
| |
Collapse
|
169
|
Chen B, Zhao J, Zhang S, Zhang Y, Huang Z. HPIP promotes gastric cancer cell proliferation through activation of cap-dependent translation. Oncol Rep 2016; 36:3664-3672. [DOI: 10.3892/or.2016.5157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/07/2016] [Indexed: 11/05/2022] Open
|
170
|
The flavonoid fisetin as an anticancer agent targeting the growth signaling pathways. Eur J Pharmacol 2016; 789:8-16. [DOI: 10.1016/j.ejphar.2016.07.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 12/29/2022]
|
171
|
Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, Ferreri K, Kandeel FR, Lugea A, Li L, Pandol SJ. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol 2016; 311:G675-G687. [PMID: 27609771 PMCID: PMC5142202 DOI: 10.1152/ajpgi.00251.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/04/2016] [Indexed: 01/31/2023]
Abstract
Epidemiological studies support strong links between obesity, diabetes, and pancreatic disorders including pancreatitis and pancreatic adenocarcinoma (PDAC). Type 2 diabetes (T2DM) is associated with insulin resistance, hyperglycemia, and hyperinsulinemia, the latter due to increased insulin secretion by pancreatic beta-cells. We reported that high-fat diet-induced PDAC progression in mice is associated with hyperglycemia, hyperinsulinemia, and activation of pancreatic stellate cells (PaSC). We investigated here the effects of high concentrations of insulin and glucose on mouse and human PaSC growth and fibrosing responses. We found that compared with normal, pancreata from T2DM patients displayed extensive collagen deposition and activated PaSC in islet and peri-islet exocrine pancreas. Mice fed a high-fat diet for up to 12 mo similarly displayed increasing peri-islet fibrosis compared with mice fed control diet. Both quiescent and activated PaSC coexpress insulin (IR; mainly A type) and IGF (IGF-1R) receptors, and both insulin and glucose modulate receptor expression. In cultured PaSC, insulin induced rapid tyrosine autophosphorylation of IR/IGF-1R at specific kinase domain activation loop sites, activated Akt/mTOR/p70S6K signaling, and inactivated FoxO1, a transcription factor that restrains cell growth. Insulin did not promote activation of quiescent PaSC in either 5 mM or 25 mM glucose containing media. However, in activated PaSC, insulin enhanced cell proliferation and augmented production of extracellular matrix proteins, and these effects were abolished by specific inhibition of mTORC1 and mTORC2. In conclusion, our data support the concept that increased local glucose and insulin concentrations associated with obesity and T2DM promote PaSC growth and fibrosing responses.
Collapse
Affiliation(s)
- Jiayue Yang
- 1Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; ,2Department of Endocrinology, Zhongda Hospital Southeast University, China;
| | - Richard T. Waldron
- 1Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; ,4Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Hsin-Yuan Su
- 1Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California;
| | - Aune Moro
- 3Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California;
| | - Hui-Hua Chang
- 3Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California;
| | - Guido Eibl
- 3Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California;
| | - Kevin Ferreri
- 5Department of Translational Research & Cellular Therapeutics, City of Hope, Duarte, California
| | - Fouad R. Kandeel
- 5Department of Translational Research & Cellular Therapeutics, City of Hope, Duarte, California
| | - Aurelia Lugea
- 1Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; ,4Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| | - Ling Li
- 2Department of Endocrinology, Zhongda Hospital Southeast University, China;
| | - Stephen J. Pandol
- 1Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California; ,4Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California; and
| |
Collapse
|
172
|
Kager L, Tamamyan G, Bielack S. Novel insights and therapeutic interventions for pediatric osteosarcoma. Future Oncol 2016; 13:357-368. [PMID: 27651036 DOI: 10.2217/fon-2016-0261] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
High-grade osteosarcomas are the most common primary malignant tumors of bone. With complete surgical resection and multi-agent chemotherapy up to 70% of patients with high-grade osteosarcomas and localized extremity tumors can become long-term survivors. The prognosis, however, is poor for patients with nonresectable, primary metastatic or relapsed disease. Outcome is essentially unchanged for three decades. Herein, we describe selected novel insights into the genomics, biology and immunology of the disease and discuss selected strategies, which hold promise to overcome the current stagnation in the therapeutic success in childhood osteosarcoma.
Collapse
Affiliation(s)
- Leo Kager
- Department of Pediatrics, St Anna Children's Hospital, Medical University Vienna, Vienna, Austria.,Children's Cancer Research Institute, Vienna, Austria
| | - Gevorg Tamamyan
- Department of Oncology, Yerevan State Medical University, Yerevan, Armenia.,Clinic of Chemotherapy, Muratsan Hospital Complex of Yerevan State Medical University, Yerevan, Armenia
| | - Stefan Bielack
- Klinikum Stuttgart, Olgahospital, Pediatrics 5 - Oncology, Hematology, Immunology, Stuttgart, Germany
| |
Collapse
|
173
|
Beale G, Haagensen EJ, Thomas HD, Wang LZ, Revill CH, Payne SL, Golding BT, Hardcastle IR, Newell DR, Griffin RJ, Cano C. Combined PI3K and CDK2 inhibition induces cell death and enhances in vivo antitumour activity in colorectal cancer. Br J Cancer 2016; 115:682-90. [PMID: 27529512 PMCID: PMC5023777 DOI: 10.1038/bjc.2016.238] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/31/2016] [Accepted: 07/14/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway is commonly deregulated in human cancer, hence many PI3K and mTOR inhibitors have been developed and have now reached clinical trials. Similarly, CDKs have been investigated as cancer drug targets. METHODS We have synthesised and characterised a series of 6-aminopyrimidines identified from a kinase screen that inhibit PI3K and/or mTOR and/or CDK2. Kinase inhibition, tumour cell growth, cell cycle distribution, cytotoxicity and signalling experiments were undertaken in HCT116 and HT29 colorectal cancer cell lines, and in vivo HT29 efficacy studies. RESULTS 2,6-Diaminopyrimidines with an O(4)-cyclohexylmethyl substituent and a C-5-nitroso or cyano group (1,2,5) induced cell cycle phase alterations and were growth inhibitory (GI50<20 μM). Compound 1, but not 2 or 5, potently inhibits CDK2 (IC50=0.1 nM) as well as PI3K, and was cytotoxic at growth inhibitory concentrations. Consistent with kinase inhibition data, compound 1 reduced phospho-Rb and phospho-rS6 at GI50 concentrations. Combination of NU6102 (CDK2 inhibitor) and pictilisib (GDC-0941; pan-PI3K inhibitor) resulted in synergistic growth inhibition, and enhanced cytotoxicity in HT29 cells in vitro and HT29 tumour growth inhibition in vivo. CONCLUSIONS These studies identified a novel series of mixed CDK2/PI3K inhibitors and demonstrate that dual targeting of CDK2 and PI3K can result in enhanced antitumour activity.
Collapse
Affiliation(s)
- Gary Beale
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Emma J Haagensen
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Huw D Thomas
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Lan-Zhen Wang
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Charlotte H Revill
- Newcastle Cancer Centre, Northern Institute for Cancer Research, School of Chemistry, Newcastle University, Bedson Building, Newcastle NE1 7RU, UK
| | - Sara L Payne
- Newcastle Cancer Centre, Northern Institute for Cancer Research, School of Chemistry, Newcastle University, Bedson Building, Newcastle NE1 7RU, UK
| | - Bernard T Golding
- Newcastle Cancer Centre, Northern Institute for Cancer Research, School of Chemistry, Newcastle University, Bedson Building, Newcastle NE1 7RU, UK
| | - Ian R Hardcastle
- Newcastle Cancer Centre, Northern Institute for Cancer Research, School of Chemistry, Newcastle University, Bedson Building, Newcastle NE1 7RU, UK
| | - David R Newell
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Roger J Griffin
- Newcastle Cancer Centre, Northern Institute for Cancer Research, School of Chemistry, Newcastle University, Bedson Building, Newcastle NE1 7RU, UK
| | - Celine Cano
- Newcastle Cancer Centre, Northern Institute for Cancer Research, School of Chemistry, Newcastle University, Bedson Building, Newcastle NE1 7RU, UK
| |
Collapse
|
174
|
Wu M, Si S, Li Y, Schoen S, Xiao GQ, Li X, Teh BT, Wu G, Chen J. Flcn-deficient renal cells are tumorigenic and sensitive to mTOR suppression. Oncotarget 2016; 6:32761-73. [PMID: 26418749 PMCID: PMC4741728 DOI: 10.18632/oncotarget.5018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022] Open
Abstract
Deficiency of tumor suppressor FLCN leads to the activation of the mTOR signaling pathway in human BHD-associated renal cell carcinomas (RCC). We have previously developed a renal distal tubule-collecting duct-Henle's loop-specific Flcn knockout (KO) mouse model (Flcnflox/flox/Ksp-Cre). This mouse model can only survive for three weeks after birth due to the development of polycystic kidney and uremia. Whether these cystic solid hyperplasia changes seen in those KO mice are tumorigenic or malignant is unknown. In this study, we demonstrated that genetic disruption of Flcn in mouse kidney distal tubule cells could lead to tumorigenic transformation of these cells to develop allograft tumors with an aggressive histologic phenotype. Consistent with previous reports, we showed that the mTOR pathway plays an important role in the growth of these Flcn-deficient allograft and human UOK 257-1 xenograft tumors. We further demonstrated that the mTOR inhibitor, sirolimus, suppresses the tumor's growth, suggesting that mTOR inhibitors might be effective in control of FLCN-deficient RCC, especially in BHD renal tumorigenesis.
Collapse
Affiliation(s)
- Mingsong Wu
- Department of Cell Biology and Genetics, Zunyi Medical University, Zunyi 563099, China
| | - Shuhui Si
- Kidney Cancer Research Laboratory, Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yan Li
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Susan Schoen
- Kidney Cancer Research Laboratory, Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Guang-Qian Xiao
- Department of Pathology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Xueying Li
- Department of Cell Biology and Genetics, Zunyi Medical University, Zunyi 563099, China
| | - Bin Tean Teh
- NCCS-VARI Translational Cancer Research Laboratory, National Cancer Centre, 169610, Singapore
| | - Guan Wu
- Department of Cell Biology and Genetics, Zunyi Medical University, Zunyi 563099, China.,Department of Pathology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jindong Chen
- Department of Cell Biology and Genetics, Zunyi Medical University, Zunyi 563099, China.,Kidney Cancer Research Laboratory, Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
175
|
Downregulation of p70S6K Enhances Cell Sensitivity to Rapamycin in Esophageal Squamous Cell Carcinoma. J Immunol Res 2016; 2016:7828916. [PMID: 27595116 PMCID: PMC4993948 DOI: 10.1155/2016/7828916] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 07/13/2016] [Indexed: 12/26/2022] Open
Abstract
It has been demonstrated that mTOR/p70S6K pathway was abnormally activated in many cancers and rapamycin and its analogs can restrain tumor growth through inhibiting this pathway, but some tumors including esophageal squamous cell carcinoma (ESCC) appear to be insensitive to rapamycin in recent studies. In the present study, we explored the measures to improve the sensitivity of ESCC cells to rapamycin and identified the clinical significance of the expression of phosphorylated p70S6K (p-p70S6K). The results showed that, after downregulating the expression of p70S6K and p-p70S6K by p70S6K siRNA, the inhibitory effects of rapamycin on cell proliferation, cell cycle, and tumor growth were significantly enhanced in vitro and in vivo. Furthermore, p-p70S6K had strong positive expression in ESCC tissues and its expression was closely related to lymph node metastasis and the TNM staging. These results indicated that p-p70S6K may participate in the invasion and metastasis in the development of ESCC and downregulation of the expression of p-p70S6K could improve the sensitivity of cells to rapamycin in ESCC.
Collapse
|
176
|
Roles of mTOR complexes in the kidney: implications for renal disease and transplantation. Nat Rev Nephrol 2016; 12:587-609. [PMID: 27477490 DOI: 10.1038/nrneph.2016.108] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mTOR pathway has a central role in the regulation of cell metabolism, growth and proliferation. Studies involving selective gene targeting of mTOR complexes (mTORC1 and mTORC2) in renal cell populations and/or pharmacologic mTOR inhibition have revealed important roles of mTOR in podocyte homeostasis and tubular transport. Important advances have also been made in understanding the role of mTOR in renal injury, polycystic kidney disease and glomerular diseases, including diabetic nephropathy. Novel insights into the roles of mTORC1 and mTORC2 in the regulation of immune cell homeostasis and function are helping to improve understanding of the complex effects of mTOR targeting on immune responses, including those that impact both de novo renal disease and renal allograft outcomes. Extensive experience in clinical renal transplantation has resulted in successful conversion of patients from calcineurin inhibitors to mTOR inhibitors at various times post-transplantation, with excellent long-term graft function. Widespread use of this practice has, however, been limited owing to mTOR-inhibitor- related toxicities. Unique attributes of mTOR inhibitors include reduced rates of squamous cell carcinoma and cytomegalovirus infection compared to other regimens. As understanding of the mechanisms by which mTORC1 and mTORC2 drive the pathogenesis of renal disease progresses, clinical studies of mTOR pathway targeting will enable testing of evolving hypotheses.
Collapse
|
177
|
Mahon RN, Hafner R. Immune Cell Regulatory Pathways Unexplored as Host-Directed Therapeutic Targets for Mycobacterium tuberculosis: An Opportunity to Apply Precision Medicine Innovations to Infectious Diseases. Clin Infect Dis 2016; 61Suppl 3:S200-16. [PMID: 26409283 DOI: 10.1093/cid/civ621] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The lack of novel antimicrobial drugs in development for tuberculosis treatment has provided an impetus for the discovery of adjunctive host-directed therapies (HDTs). Several promising HDT candidates are being evaluated, but major advancement of tuberculosis HDTs will require understanding of the master or "core" cell signaling pathways that control intersecting immunologic and metabolic regulatory mechanisms, collectively described as "immunometabolism." Core regulatory pathways conserved in all eukaryotic cells include poly (ADP-ribose) polymerases (PARPs), sirtuins, AMP-activated protein kinase (AMPK), and mechanistic target of rapamycin (mTOR) signaling. Critical interactions of these signaling pathways with each other and their roles as master regulators of immunometabolic functions will be addressed, as well as how Mycobacterium tuberculosis is already known to influence various other cell signaling pathways interacting with them. Knowledge of these essential mechanisms of cell function regulation has led to breakthrough targeted treatment advances for many diseases, most prominently in oncology. Leveraging these exciting advances in precision medicine for the development of innovative next-generation HDTs may lead to entirely new paradigms for treatment and prevention of tuberculosis and other infectious diseases.
Collapse
Affiliation(s)
- Robert N Mahon
- Division of AIDS-Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Contractor to the National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Richard Hafner
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
178
|
Barnes PJ. Kinases as Novel Therapeutic Targets in Asthma and Chronic Obstructive Pulmonary Disease. Pharmacol Rev 2016; 68:788-815. [PMID: 27363440 DOI: 10.1124/pr.116.012518] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Multiple kinases play a critical role in orchestrating the chronic inflammation and structural changes in the respiratory tract of patients with asthma and chronic obstructive pulmonary disease (COPD). Kinases activate signaling pathways that lead to contraction of airway smooth muscle and release of inflammatory mediators (such as cytokines, chemokines, growth factors) as well as cell migration, activation, and proliferation. For this reason there has been great interest in the development of kinase inhibitors as anti-inflammatory therapies, particular where corticosteroids are less effective, as in severe asthma and COPD. However, it has proven difficult to develop selective kinase inhibitors that are both effective and safe after oral administration and this has led to a search for inhaled kinase inhibitors, which would reduce systemic exposure. Although many kinases have been implicated in inflammation and remodeling of airway disease, very few classes of drug have reached the stage of clinical studies in these diseases. The most promising drugs are p38 MAP kinases, isoenzyme-selective PI3-kinases, Janus-activated kinases, and Syk-kinases, and inhaled formulations of these drugs are now in development. There has also been interest in developing inhibitors that block more than one kinase, because these drugs may be more effective and with less risk of losing efficacy with time. No kinase inhibitors are yet on the market for the treatment of airway diseases, but as kinase inhibitors are improved from other therapeutic areas there is hope that these drugs may eventually prove useful in treating refractory asthma and COPD.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
179
|
Discovery of ( S )-4-isobutyloxazolidin-2-one as a novel leucyl-tRNA synthetase (LRS)-targeted mTORC1 inhibitor. Bioorg Med Chem Lett 2016; 26:3038-3041. [DOI: 10.1016/j.bmcl.2016.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 04/28/2016] [Accepted: 05/04/2016] [Indexed: 11/19/2022]
|
180
|
Schmidlin T, Garrigues L, Lane CS, Mulder TC, van Doorn S, Post H, de Graaf EL, Lemeer S, Heck AJR, Altelaar AFM. Assessment of SRM, MRM3, and DIA for the targeted analysis of phosphorylation dynamics in non-small cell lung cancer. Proteomics 2016; 16:2193-205. [DOI: 10.1002/pmic.201500453] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/12/2016] [Accepted: 05/20/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Thierry Schmidlin
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| | - Luc Garrigues
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| | | | - T. Celine Mulder
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| | - Sander van Doorn
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| | - Erik L. de Graaf
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
- Current address: Erik L. de Graaf, Fondazione Pisana per la Scienza ONLUS; Via Panfilo Castaldi 2; 56121 Pisa Italy
| | - Simone Lemeer
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| | - A. F. Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics; Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences; Utrecht University and Netherlands Proteomics Centre; Utrecht The Netherlands
| |
Collapse
|
181
|
Dong H, Jing W, Runpeng Z, Xuewei X, Min M, Ru C, Yingru X, Shengfa N, Rongbo Z. ESAT6 inhibits autophagy flux and promotes BCG proliferation through MTOR. Biochem Biophys Res Commun 2016; 477:195-201. [PMID: 27317487 DOI: 10.1016/j.bbrc.2016.06.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/09/2016] [Indexed: 12/31/2022]
Abstract
In recent years, increasing studies have found that pathogenic Mycobacterium tuberculosis (Mtb) inhibits autophagy, which mediates the anti-mycobacterial response, but the mechanism is not clear. We previously reported that secretory acid phosphatase (SapM) of Mtb can negatively regulate autophagy flux. Recently, another virulence factor of Mtb, early secretory antigenic target 6 (ESAT6), has been found to be involved in inhibiting autophagy, but the mechanism remains unclear. In this study, we show that ESAT6 hampers autophagy flux to boost bacillus Calmette-Guerin (BCG) proliferation and reveals a mechanism by which ESAT6 blocks autophagosome-lysosome fusion in a mammalian target of rapamycin (MTOR)-dependent manner. In both Raw264.7 cells and primary macrophages derived from the murine abdominal cavity (ACM), ESAT6 repressed autophagy flux by interfering with the autophagosome-lysosome fusion, which resulted in an increased load of BCG. Impaired degradation of LC3Ⅱ and SQSTM1 by ESAT6 was related to the upregulated activity of MTOR. Contrarily, inhibiting MTOR with Torin1 removed the ESAT6-induced autophagy block and lysosome dysfunction. Furthermore, in both Raw264.7 and ACM cells, MTOR inhibition significantly suppressed the survival of BCG. In conclusion, our study highlights how ESAT6 blocks autophagy and promotes BCG survival in a way that activates MTOR.
Collapse
Affiliation(s)
- Hu Dong
- Department of Medical Immunology, Medical School, Anhui University of Science and Technology, China; Medical Inspection Center, Anhui University of Science and Technology, Huainan, China.
| | - Wu Jing
- Department of Medical Immunology, Medical School, Anhui University of Science and Technology, China; Medical Inspection Center, Anhui University of Science and Technology, Huainan, China.
| | - Zhao Runpeng
- Department of Medical Immunology, Medical School, Anhui University of Science and Technology, China
| | - Xu Xuewei
- Department of Medical Immunology, Medical School, Anhui University of Science and Technology, China
| | - Mu Min
- Department of Medical Immunology, Medical School, Anhui University of Science and Technology, China
| | - Cai Ru
- Department of Medical Immunology, Medical School, Anhui University of Science and Technology, China
| | - Xing Yingru
- Affiliated Cancer Hospital, Anhui University of Science and Technology, China
| | - Ni Shengfa
- Affiliated Cancer Hospital, Anhui University of Science and Technology, China
| | - Zhang Rongbo
- Department of Medical Immunology, Medical School, Anhui University of Science and Technology, China; Medical Inspection Center, Anhui University of Science and Technology, Huainan, China
| |
Collapse
|
182
|
Abstract
Awareness that the metabolic phenotype of cells within tumours is heterogeneous - and distinct from that of their normal counterparts - is growing. In general, tumour cells metabolize glucose, lactate, pyruvate, hydroxybutyrate, acetate, glutamine, and fatty acids at much higher rates than their nontumour equivalents; however, the metabolic ecology of tumours is complex because they contain multiple metabolic compartments, which are linked by the transfer of these catabolites. This metabolic variability and flexibility enables tumour cells to generate ATP as an energy source, while maintaining the reduction-oxidation (redox) balance and committing resources to biosynthesis - processes that are essential for cell survival, growth, and proliferation. Importantly, experimental evidence indicates that metabolic coupling between cell populations with different, complementary metabolic profiles can induce cancer progression. Thus, targeting the metabolic differences between tumour and normal cells holds promise as a novel anticancer strategy. In this Review, we discuss how cancer cells reprogramme their metabolism and that of other cells within the tumour microenvironment in order to survive and propagate, thus driving disease progression; in particular, we highlight potential metabolic vulnerabilities that might be targeted therapeutically.
Collapse
|
183
|
Abstract
The movement toward precision medicine with targeted therapeutics for cancer treatment has been hindered by both innate and acquired resistance. Understanding the molecular wiring and plasticity of oncogenic signaling networks is essential to the development of therapeutic strategies to avoid or overcome resistance. The mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) represents a highly integrated signaling node that is dysregulated in the majority of human cancers. Several studies have revealed that sustained mTORC1 inhibition is essential to avoid resistance to targeted therapeutics against the driving oncogenic pathway in a given cancer. Here we discuss the role of mTORC1 in dictating the response of tumors to targeted therapeutics and review recent examples from lung cancer, breast cancer, and melanoma.
Collapse
Affiliation(s)
- Erika Ilagan
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
184
|
Karandish F, Mallik S. Biomarkers and Targeted Therapy in Pancreatic Cancer. BIOMARKERS IN CANCER 2016; 8:27-35. [PMID: 27147897 PMCID: PMC4847554 DOI: 10.4137/bic.s34414] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/06/2016] [Accepted: 03/11/2016] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) constitutes 90% of pancreatic cancers. PDAC is a complex and devastating disease with only 1%-3% survival rate in five years after the second stage. Treatment of PDAC is complicated due to the tumor microenvironment, changing cell behaviors to the mesenchymal type, altered drug delivery, and drug resistance. Considering that pancreatic cancer shows early invasion and metastasis, critical research is needed to explore different aspects of the disease, such as elaboration of biomarkers, specific signaling pathways, and gene aberration. In this review, we highlight the biomarkers, the fundamental signaling pathways, and their importance in targeted drug delivery for pancreatic cancers.
Collapse
Affiliation(s)
- Fataneh Karandish
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA
| |
Collapse
|
185
|
Xia Y, Chen J, Gong C, Chen H, Sun J. α-Mangostin, a Natural Agent, Enhances the Response of NRAS Mutant Melanoma to Retinoic Acid. Med Sci Monit 2016; 22:1360-7. [PMID: 27104669 PMCID: PMC4844330 DOI: 10.12659/msm.898204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The identification and use of novel compounds alone or in combination hold promise for the fight against NRAS mutant melanoma. MATERIAL AND METHODS We screened a kinase-specific inhibitor library through combining it with α-Mangostin in NRAS mutant melanoma cell line, and verified the enhancing effect of α-Mangostin through inhibition of the tumorigenesis pathway. RESULTS Within the kinase inhibitors, retinoic acid showed a significant synergistic effect with α-Mangostin. α-Mangostin also can reverse the drug resistance of retinoic acid in RARa siRNA-transduced sk-mel-2 cells. Colony assay, TUNEL staining, and the expressions of several apoptosis-related genes revealed that a-Mangostin enhanced the effect of retinoic acid-induced apoptosis. The combination treatment resulted in marked induction of ROS generation and inhibition of the AKT/S6 pathway. CONCLUSIONS These results indicate that the combination of these novel natural agents with retinoid acid may be clinically effective in NRAS mutant melanoma.
Collapse
Affiliation(s)
- Yun Xia
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jing Chen
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Chongwen Gong
- Department of Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (mainland)
| |
Collapse
|
186
|
Reinwald M, Boch T, Hofmann WK, Buchheidt D. Risk of Infectious Complications in Hemato-Oncological Patients Treated with Kinase Inhibitors. Biomark Insights 2016; 10:55-68. [PMID: 27127405 PMCID: PMC4841329 DOI: 10.4137/bmi.s22430] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/13/2015] [Accepted: 09/15/2015] [Indexed: 12/30/2022] Open
Abstract
Infectious complications are a major cause of morbidity and mortality in patients with hemato-oncological diseases. Although disease-related immunosuppression represents one factor, aggressive treatment regimens, such as chemotherapy, stem cell transplantation, or antibody treatment, account for a large proportion of infectious side effects. With the advent of targeted therapies affecting specific kinases in malignant diseases, the outcome of patients has further improved. Nonetheless, dependent on the specific pathway targeted or off-target activity of the kinase inhibitor, therapy-associated infectious complications may occur. We review the most common and approved kinase inhibitors targeting a variety of hemato-oncological malignancies for their immunosuppressive potential and evaluate their risk of infectious side effects based on preclinical evidence and clinical data in order to raise awareness of the potential risks involved.
Collapse
Affiliation(s)
- Mark Reinwald
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Tobias Boch
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Dieter Buchheidt
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
187
|
Baretić D, Berndt A, Ohashi Y, Johnson CM, Williams RL. Tor forms a dimer through an N-terminal helical solenoid with a complex topology. Nat Commun 2016; 7:11016. [PMID: 27072897 PMCID: PMC4833857 DOI: 10.1038/ncomms11016] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/11/2016] [Indexed: 01/18/2023] Open
Abstract
The target of rapamycin (Tor) is a Ser/Thr protein kinase that regulates a range of anabolic and catabolic processes. Tor is present in two complexes, TORC1 and TORC2, in which the Tor–Lst8 heterodimer forms a common sub-complex. We have determined the cryo-electron microscopy (EM) structure of Tor bound to Lst8. Two Tor–Lst8 heterodimers assemble further into a dyad-symmetry dimer mediated by Tor–Tor interactions. The first 1,300 residues of Tor form a HEAT repeat-containing α-solenoid with four distinct segments: a highly curved 800-residue N-terminal 'spiral', followed by a 400-residue low-curvature 'bridge' and an extended ‘railing' running along the bridge leading to the 'cap' that links to FAT region. This complex topology was verified by domain insertions and offers a new interpretation of the mTORC1 structure. The spiral of one TOR interacts with the bridge of another, which together form a joint platform for the Regulatory Associated Protein of TOR (RAPTOR) regulatory subunit. The target of rapamycin (Tor) is a Ser/Thr protein kinase that regulates a wide range of anabolic and catabolic processes. Here the authors describe a sub-nanometer cryo-EM structure of a yeast Tor–Lst8 complex and propose an overall topology that differs from that previously suggested for mTORC1.
Collapse
Affiliation(s)
| | - Alex Berndt
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Yohei Ohashi
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | | |
Collapse
|
188
|
Gao Y, Gao J, Li M, Zheng Y, Wang Y, Zhang H, Wang W, Chu Y, Wang X, Xu M, Cheng T, Ju Z, Yuan W. Rheb1 promotes tumor progression through mTORC1 in MLL-AF9-initiated murine acute myeloid leukemia. J Hematol Oncol 2016; 9:36. [PMID: 27071307 PMCID: PMC4830070 DOI: 10.1186/s13045-016-0264-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/03/2016] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The constitutive hyper-activation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathways has frequently been associated with acute myeloid leukemia (AML). While many inhibitors targeting these pathways have been developed, the anti-leukemic effect was not as robust as expected. As part of the molecular link between PI3K/Akt and mTOR kinase, the role of Rheb1 in AML remains unexplored. Our study aims to explore the role of Rheb1 in AML and estimate whether Rheb1 could be a potential target of AML treatment. METHODS The expressions of Rheb1 and other indicated genes were analyzed using real-time PCR. AML mouse model was established by retrovirus transduction. Leukemia cell properties and related signaling pathways were dissected by in vitro and in vivo studies. The transcriptional changes were analyzed via gene chip analysis. Molecular reagents including mTOR inhibitor and mTOR activator were used to evaluate the function of related signaling pathway in the mouse model. RESULTS We observed that Rheb1 is overexpressed in AML patients and the change of Rheb1 level in AML patients is associated with their median survival. Using a Rheb1-deficient MLL-AF9 murine AML model, we revealed that Rheb1 deletion prolonged the survival of AML mice by weakening LSC function. In addition, Rheb1 deletion arrested cell cycle progression and enhanced apoptosis of AML cells. Furthermore, while Rheb1 deletion reduced mTORC1 activity in AML cells, additional rapamycin treatment further decreased mTORC1 activity and increased the apoptosis of Rheb1 (Δ/Δ) AML cells. The mTOR activator 3BDO partially rescued mTORC1 signaling and inhibited apoptosis in Rheb1 (Δ/Δ) AML cells. CONCLUSIONS Our data suggest that Rheb1 promotes AML progression through mTORC1 signaling pathway and combinational drug treatments targeting Rheb1 and mTOR might have a better therapeutic effect on leukemia.
Collapse
Affiliation(s)
- Yanan Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Juan Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Minghao Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Yawei Zheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Yajie Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Hongyan Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Weili Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Yajing Chu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Xiaomin Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China.
| | - Mingjiang Xu
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, USA
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China
| | - Zhenyu Ju
- Institute of Aging, Hangzhou Normal University, Hangzhou, 310036, China
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Center for Stem Cell Medicine, CAMS & PUMC, Beijing, China.
| |
Collapse
|
189
|
Sato T, Ishii J, Ota Y, Sasaki E, Shibagaki Y, Hattori S. Mammalian target of rapamycin (mTOR) complex 2 regulates filamin A-dependent focal adhesion dynamics and cell migration. Genes Cells 2016; 21:579-93. [DOI: 10.1111/gtc.12366] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/01/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Tatsuhiro Sato
- Division of Biochemistry; School of Pharmaceutical Sciences; Kitasato University; 5-9-1 Shirokane Minato-ku Tokyo 108-8641 Japan
| | - Junko Ishii
- Division of Biochemistry; School of Pharmaceutical Sciences; Kitasato University; 5-9-1 Shirokane Minato-ku Tokyo 108-8641 Japan
| | - Yuki Ota
- Division of Biochemistry; School of Pharmaceutical Sciences; Kitasato University; 5-9-1 Shirokane Minato-ku Tokyo 108-8641 Japan
| | - Eri Sasaki
- Division of Biochemistry; School of Pharmaceutical Sciences; Kitasato University; 5-9-1 Shirokane Minato-ku Tokyo 108-8641 Japan
| | - Yoshio Shibagaki
- Division of Biochemistry; School of Pharmaceutical Sciences; Kitasato University; 5-9-1 Shirokane Minato-ku Tokyo 108-8641 Japan
| | - Seisuke Hattori
- Division of Biochemistry; School of Pharmaceutical Sciences; Kitasato University; 5-9-1 Shirokane Minato-ku Tokyo 108-8641 Japan
| |
Collapse
|
190
|
4EBP1/c-MYC/PUMA and NF-κB/EGR1/BIM pathways underlie cytotoxicity of mTOR dual inhibitors in malignant lymphoid cells. Blood 2016; 127:2711-22. [PMID: 26917778 DOI: 10.1182/blood-2015-02-629485] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 02/13/2016] [Indexed: 12/14/2022] Open
Abstract
The mammalian target of rapamycin (mTOR), a kinase that regulates proliferation and apoptosis, has been extensively evaluated as a therapeutic target in multiple malignancies. Rapamycin analogs, which partially inhibit mTOR complex 1 (mTORC1), exhibit immunosuppressive and limited antitumor activity, but sometimes activate survival pathways through feedback mechanisms involving mTORC2. Thus, attention has turned to agents targeting both mTOR complexes by binding the mTOR active site. Here we show that disruption of either mTOR-containing complex is toxic to acute lymphocytic leukemia (ALL) cells and identify 2 previously unrecognized pathways leading to this cell death. Inhibition of mTORC1-mediated 4EBP1 phosphorylation leads to decreased expression of c-MYC and subsequent upregulation of the proapoptotic BCL2 family member PUMA, whereas inhibition of mTORC2 results in nuclear factor-κB-mediated expression of the Early Growth Response 1 (EGR1) gene, which encodes a transcription factor that binds and transactivates the proapoptotic BCL2L11 locus encoding BIM. Importantly, 1 or both pathways contribute to death of malignant lymphoid cells after treatment with dual mTORC1/mTORC2 inhibitors. Collectively, these observations not only provide new insight into the survival roles of mTOR in lymphoid malignancies, but also identify alterations that potentially modulate the action of mTOR dual inhibitors in ALL.
Collapse
|
191
|
Blockage of glutaminolysis enhances the sensitivity of ovarian cancer cells to PI3K/mTOR inhibition involvement of STAT3 signaling. Tumour Biol 2016; 37:11007-15. [PMID: 26894601 DOI: 10.1007/s13277-016-4984-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/07/2016] [Indexed: 12/30/2022] Open
Abstract
The PI3K/Akt/mTOR axis in ovarian cancer is frequently activated and implicated in tumorigenesis. Specific targeting of this pathway is therefore an attractive therapeutic approach for ovarian cancer. However, ovarian cancer cells are resistant to PP242, a dual inhibitor of mTORC1 and mTORC2. Interestingly, blockage of GLS1 with a selective inhibitor, CB839, or siRNA dramatically sensitized the PP242-induced cell death, as evident from increased PARP cleavage. The anti-cancer activity of CB-839 and PP242 was abrogated by the addition of the TCA cycle product α-ketoglutarate, indicating the critical function of GLS1 in ovarian cancer cell survival. Finally, glutaminolysis inhibition activated apoptosis and synergistically sensitized ovarian cancer cells to priming with the mTOR inhibitor PP242. GLS1 inhibition significantly reduced phosphorylated STAT3 expression in ovarian cancer cells. These findings show that targeting glutamine addiction via GLS1 inhibition offers a potential novel therapeutic strategy to overcome resistance to PI3K/Akt/mTOR inhibition.
Collapse
|
192
|
Bridgeman BB, Wang P, Ye B, Pelling JC, Volpert OV, Tong X. Inhibition of mTOR by apigenin in UVB-irradiated keratinocytes: A new implication of skin cancer prevention. Cell Signal 2016; 28:460-468. [PMID: 26876613 DOI: 10.1016/j.cellsig.2016.02.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 12/18/2022]
Abstract
Ultraviolet B (UVB) radiation is the major environmental risk factor for developing skin cancer, the most common cancer worldwide, which is characterized by aberrant activation of Akt/mTOR (mammalian target of rapamycin). Importantly, the link between UV irradiation and mTOR signaling has not been fully established. Apigenin is a naturally occurring flavonoid that has been shown to inhibit UV-induced skin cancer. Previously, we have demonstrated that apigenin activates AMP-activated protein kinase (AMPK), which leads to suppression of basal mTOR activity in cultured keratinocytes. Here, we demonstrated that apigenin inhibited UVB-induced mTOR activation, cell proliferation and cell cycle progression in mouse skin and in mouse epidermal keratinocytes. Interestingly, UVB induced mTOR signaling via PI3K/Akt pathway, however, the inhibition of UVB-induced mTOR signaling by apigenin was not Akt-dependent. Instead, it was driven by AMPK activation. In addition, mTOR inhibition by apigenin in keratinocytes enhanced autophagy, which was responsible, at least in part, for the decreased proliferation in keratinocytes. In contrast, apigenin did not alter UVB-induced apoptosis. Taken together, our results indicate the important role of mTOR inhibition in UVB protection by apigenin, and provide a new target and strategy for better prevention of UV-induced skin cancer.
Collapse
Affiliation(s)
- Bryan B Bridgeman
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Pu Wang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Boping Ye
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jill C Pelling
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olga V Volpert
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xin Tong
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
193
|
Apoptosis, autophagy and unfolded protein response pathways in Arbovirus replication and pathogenesis. Expert Rev Mol Med 2016; 18:e1. [PMID: 26781343 PMCID: PMC4836210 DOI: 10.1017/erm.2015.19] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Arboviruses are pathogens that widely affect the health of people in different communities around the world. Recently, a few successful approaches toward production of effective vaccines against some of these pathogens have been developed, but treatment and prevention of the resulting diseases remain a major health and research concern. The arbovirus infection and replication processes are complex, and many factors are involved in their regulation. Apoptosis, autophagy and the unfolded protein response (UPR) are three mechanisms that are involved in pathogenesis of many viruses. In this review, we focus on the importance of these pathways in the arbovirus replication and infection processes. We provide a brief introduction on how apoptosis, autophagy and the UPR are initiated and regulated, and then discuss the involvement of these pathways in regulation of arbovirus pathogenesis.
Collapse
|
194
|
Fraser C, Carragher NO, Unciti-Broceta A. eCF309: a potent, selective and cell-permeable mTOR inhibitor. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00493d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Kinase inhibitors capable of blocking the phosphorylation of protein substrates with high selectivity are essential to probe and elucidate the etiological role of such molecules and their signalling pathways.
Collapse
Affiliation(s)
- Craig Fraser
- Edinburgh Cancer Research UK Centre
- Institute of Genetics and Molecular Medicine
- University of Edinburgh
- Edinburgh EH4 2XR
- UK
| | - Neil O. Carragher
- Edinburgh Cancer Research UK Centre
- Institute of Genetics and Molecular Medicine
- University of Edinburgh
- Edinburgh EH4 2XR
- UK
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research UK Centre
- Institute of Genetics and Molecular Medicine
- University of Edinburgh
- Edinburgh EH4 2XR
- UK
| |
Collapse
|
195
|
Abstract
Mechanistic target of rapamycin (mTOR, also known as mammalian target of rapamycin) is a ubiquitous serine/threonine kinase that regulates cell growth, proliferation and survival. These effects are cell-type-specific, and are elicited in response to stimulation by growth factors, hormones and cytokines, as well as to internal and external metabolic cues. Rapamycin was initially developed as an inhibitor of T-cell proliferation and allograft rejection in the organ transplant setting. Subsequently, its molecular target (mTOR) was identified as a component of two interacting complexes, mTORC1 and mTORC2, that regulate T-cell lineage specification and macrophage differentiation. mTORC1 drives the proinflammatory expansion of T helper (TH) type 1, TH17, and CD4(-)CD8(-) (double-negative, DN) T cells. Both mTORC1 and mTORC2 inhibit the development of CD4(+)CD25(+)FoxP3(+) T regulatory (TREG) cells and, indirectly, mTORC2 favours the expansion of T follicular helper (TFH) cells which, similarly to DN T cells, promote B-cell activation and autoantibody production. In contrast to this proinflammatory effect of mTORC2, mTORC1 favours, to some extent, an anti-inflammatory macrophage polarization that is protective against infections and tissue inflammation. Outside the immune system, mTORC1 controls fibroblast proliferation and chondrocyte survival, with implications for tissue fibrosis and osteoarthritis, respectively. Rapamycin (which primarily inhibits mTORC1), ATP-competitive, dual mTORC1/mTORC2 inhibitors and upstream regulators of the mTOR pathway are being developed to treat autoimmune, hyperproliferative and degenerative diseases. In this regard, mTOR blockade promises to increase life expectancy through treatment and prevention of rheumatic diseases.
Collapse
Affiliation(s)
- Andras Perl
- Division of Rheumatology, Departments of Medicine, Microbiology and Immunology, and Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, 750 East Adams Street, Syracuse, New York 13210, USA
| |
Collapse
|
196
|
Cai H, Dong LQ, Liu F. Recent Advances in Adipose mTOR Signaling and Function: Therapeutic Prospects. Trends Pharmacol Sci 2015; 37:303-317. [PMID: 26700098 DOI: 10.1016/j.tips.2015.11.011] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/19/2015] [Accepted: 11/20/2015] [Indexed: 12/11/2022]
Abstract
The increasing epidemic of obesity and its comorbidities has spurred research interest in adipose biology and its regulatory functions. Recent studies have revealed that the mechanistic target of rapamycin (mTOR) signaling pathway has a critical role in the regulation of adipose tissue function, including adipogenesis, lipid metabolism, thermogenesis, and adipokine synthesis and/or secretion. Given the importance of mTOR signaling in controlling energy homeostasis, it is not unexpected that deregulated mTOR signaling is associated with obesity and related metabolic disorders. In this review, we highlight current advances in understanding the roles of the mTOR signaling pathway in adipose tissue. We also provide a more nuanced view of how the mTOR signaling pathway regulates adipose tissue biology and function. Finally, we describe approaches to modulate the activity and tissue-specific function of mTOR that may pave the way towards counteracting obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Huan Cai
- Institute of Metabolism and Endocrinology, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Pharmacology, UTHSCSA, San Antonio, TX, USA
| | - Lily Q Dong
- Departments of Cellular Structural Biology, UTHSCSA, San Antonio, TX, USA
| | - Feng Liu
- Institute of Metabolism and Endocrinology, Metabolic Syndrome Research Center, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Pharmacology, UTHSCSA, San Antonio, TX, USA.
| |
Collapse
|
197
|
Feng J, Yan PF, Zhao HY, Zhang FC, Zhao WH, Feng M. SIRT6 suppresses glioma cell growth via induction of apoptosis, inhibition of oxidative stress and suppression of JAK2/STAT3 signaling pathway activation. Oncol Rep 2015; 35:1395-402. [PMID: 26648570 DOI: 10.3892/or.2015.4477] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/01/2015] [Indexed: 11/06/2022] Open
Abstract
Sirtuin 6 (SIRT6) is a member of the mammalian NAD+‑dependent deacetylase sirtuin family that acts to maintain genomic stability and to repress genes. SIRT6 has recently been reported to be a tumor suppressor that controls cancer metabolism, although this effect of SIRT6 is still in dispute. Moreover, the role of SIRT6 in glioma is largely unknown. In the present study, we found that overexpression of SIRT6 using an adenovirus inhibited glioma cell growth and induced marked cell injury in two glioma cell lines (U87‑MG and T98G). Fluorescent terminal deoxyribonucleotidyl transferase (TdT)‑mediated biotin‑16‑dUTP nick‑end labelling (TUNEL) assay showed that SIRT6 overexpression induced obvious apoptosis in the T98G glioma cells. Immunoblotting and immunofluorescent staining demonstrated that SIRT6 overexpression promoted the mitochondrial-to‑nuclear translocation of apoptosis‑inducing factor (AIF), a potent apoptosis inducer. Moreover, we found that SIRT6 overexpression largely reduced oxidative stress and suppressed the activation of the JAK2/STAT3 signaling pathway in glioma cells. Finally, we showed that SIRT6 mRNA and protein levels in human glioblastoma multiforme tissues were significantly lower than the levels in peritumor tissues. In summary, our data suggest that SIRT6 suppresses glioma cell growth via induction of apoptosis, inhibition of oxidative stress and inhibition of the activation of the JAK2/STAT3 signaling pathway. These results indicate that SIRT6 may be a promising therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Jun Feng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Peng-Fei Yan
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hong-Yang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Fang-Cheng Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wo-Hua Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Min Feng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
198
|
Eiden AM, Zhang S, Gary JM, Simmons JK, Mock BA. Molecular Pathways: Increased Susceptibility to Infection Is a Complication of mTOR Inhibitor Use in Cancer Therapy. Clin Cancer Res 2015; 22:277-83. [PMID: 26607598 DOI: 10.1158/1078-0432.ccr-14-3239] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022]
Abstract
As one of the earliest examples of "chemical biology," the M: echanistic T: arget of R: apamycin (mTOR) protein and its chemical inhibitors have been extensively studied across a spectrum of physiologic and pathologic processes at the molecular, organismal, and patient population levels. There are several FDA-approved mTOR inhibitors (sirolimus, everolimus, and temsirolimus) with indications for cancer treatment and for prevention of solid organ rejection. Dozens of mTOR inhibitors are currently being evaluated in hundreds of ongoing clinical trials across a spectrum of diseases, including numerous cancer indications, autoimmune diseases, and a number of congenital disorders. As many of the approved and investigational indications for mTOR inhibitors require long-term treatment, the magnitude and incidence of particular side effects differ from those observed in shorter-term treatments. Here, we focus on the increased risk of infections in patients being treated with mTOR inhibitors. While increased infection rates might be expected from a class of drugs approved as posttransplant immunosuppressants, we review reports from clinical, mechanistic, and genetically engineered mouse model studies detailing a much more nuanced view of mTOR inhibitor drug action and target biology.
Collapse
Affiliation(s)
- Adrian M Eiden
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Joy M Gary
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - John K Simmons
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
199
|
Chen L, Liu T, Tu Y, Rong D, Cao Y. Cul1 promotes melanoma cell proliferation by promoting DEPTOR degradation and enhancing cap-dependent translation. Oncol Rep 2015; 35:1049-56. [PMID: 26717892 DOI: 10.3892/or.2015.4442] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 09/15/2015] [Indexed: 11/06/2022] Open
Abstract
Cullin1 (Cul1) serves as a rigid scaffold in the SCF (Skp1/Cullin/Rbx1/F-box protein) E3 ubiquitin ligase complex and has been found to be overexpressed in melanoma and to enhance melanoma cell proliferation by promoting G1-S phase transition. However, the underlying mechanisms involved in the regulation of melanoma cell proliferation by Cul1 remain poorly understood. In the present study, we found that Cul1 promoted mTORC1 activity and cap-dependent translation by enhancing the ubiquitination and degradation of DEPTOR. We further showed that suppression of the eIF4F complex assembly profoundly inhibited the promoting effect of Cul1 on melanoma cell proliferation, while enhancement of the eIF4F complex activity reversed the inhibitory effect of Cul1 depletion on melanoma cell proliferation, indicating that Cul1 contributes to melanoma cell proliferation by activating cap‑dependent translation. These data elucidate the role of Cul1 in cap-dependent translation and improves our understanding of the underlying mechanisms involved in the regulation of melanoma cell proliferation by Cul1.
Collapse
Affiliation(s)
- Lan Chen
- Department of Dermatology, The Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Tianyu Liu
- Department of Dermatology, The Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yunhua Tu
- Department of Dermatology, The Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Dongyun Rong
- Department of Dermatology, The Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yu Cao
- Department of Dermatology, The Affiliated Hospital of Guiyang Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
200
|
Malley CO, Pidgeon GP. The mTOR pathway in obesity driven gastrointestinal cancers: Potential targets and clinical trials. BBA CLINICAL 2015; 5:29-40. [PMID: 27051587 PMCID: PMC4802403 DOI: 10.1016/j.bbacli.2015.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 11/03/2015] [Accepted: 11/11/2015] [Indexed: 12/20/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a crucial point of convergence between growth factor signalling, metabolism, nutrient status and cellular proliferation. The mTOR pathway is heavily implicated in the progression of many cancers and is emerging as an important driver of gastrointestinal (GI) malignancies. Due to its central role in adapting metabolism to environmental conditions, mTOR signalling is also believed to be critical in the development of obesity. Recent research has delineated that excessive nutrient intake can promote signalling through the mTOR pathway and possibly evoke changes to cellular metabolism that could accelerate obesity related cancers. Acting through its two effector complexes mTORC1 and mTORC2, mTOR dictates the transcription of genes important in glycolysis, lipogenesis, protein translation and synthesis and has recently been defined as a central mediator of the Warburg effect in cancer cells. Activation of the mTOR pathway is involved in both the pathogenesis of GI malignancies and development of resistance to conventional chemotherapy and radiotherapy. The use of mTOR inhibitors is a promising therapeutic option in many GI malignancies, with greatest clinical efficacy seen in combination regimens. Recent research has also provided insight into crosstalk between mTOR and other pathways which could potentially expand the list of therapeutic targets in the mTOR pathway. Here we review the available strategies for targeting the mTOR pathway in GI cancers. We discuss current clinical trials of both established and novel mTOR inhibitors, with particular focus on combinations of these drugs with conventional chemotherapy, radiotherapy and targeted therapies.
Collapse
Affiliation(s)
- Cian O Malley
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - Graham P Pidgeon
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|