1
|
Al-Othman R, Al-Jarallah A, Babiker F. High-density lipoprotein protects normotensive and hypertensive rats against ischemia-reperfusion injury through differential regulation of mTORC1 and mTORC2 signaling. Front Pharmacol 2024; 15:1398630. [PMID: 39611167 PMCID: PMC11603114 DOI: 10.3389/fphar.2024.1398630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Background High-density lipoprotein (HDL) protects against myocardial ischemia-reperfusion (I/R) injury. Mammalian target of rapamycin complexes 1 and 2 (mTORC1 and mTORC2) play opposing roles in protecting against I/R injury, whereby mTORC1 appears to be detrimental while mTORC2 is protective. However, the role of HDL and mTORC signaling in protecting against I/R in hypertensive rodents is not clearly understood. In this study, we investigated the involvement of mTORC1 and mTORC2 in HDL-mediated protection against myocardial I/R injury in normotensive Wistar Kyoto (WKY) rats and spontaneously hypertensive rats (SHR). Methods Hearts from WKY and SHR were subjected to I/R injury using a modified Langendorff system. Hemodynamics data were collected, and infarct size was measured. Rapamycin and JR-AB2-011 were used to test the role of mTORC1 and mTORC2, respectively. MK-2206 was used to test the role of Akt in HDL-mediated cardiac protection. The expression levels and the activation states of mediators of mTORC1 and mTORC2 signaling and myocardial apoptosis were measured by immunoblotting and/or enzyme-linked immunosorbent assay (ELISA). Results HDL protected hearts from WKY and SHR against I/R injury as indicated by significant improvements in cardiac hemodynamics and reduction in infarct size. HDL induced greater protection in WKY compared to SHR. HDL treatment attenuated mTORC1 signaling in WKY by reducing the phosphorylation of P70S6K (mTORC1 substrate). In SHR however, HDL attenuated mTORC1 signaling by reducing the levels of phospho-mTORC1, Rag C (mTORC1 activator), and phospho-PRAS40 (mTORC1 inhibitor). HDL increased the phosphorylation of mTORC2 substrate Akt, specifically the Akt2 isoform in SHR and to a greater extent in WKY. HDL-induced protection was abolished in the presence of Akt antagonist and involved attenuation of GSK, caspases 7 and 8 activation, and cytochrome C release. Conclusion HDL mediates cardiac protection via attenuation of mTORC1, activation of mTORC2-Akt2, and inhibition of myocardial apoptosis. HDL regulates mTORC1 and mTORC2 signaling via distinct mechanisms in normotensive and hypertensive rats. HDL attenuation of mTORC1 and activation of mTORC2-Akt2 signaling could be a mechanism by which HDL protects against myocardial I/R injury in hypertension.
Collapse
Affiliation(s)
- Reham Al-Othman
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Aishah Al-Jarallah
- Department of Biochemistry, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Fawzi Babiker
- Department of Physiology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
2
|
Kohvakka A, Sattari M, Nättinen J, Aapola U, Gregorová P, Tammela TLJ, Uusitalo H, Sarin LP, Visakorpi T, Latonen L. Long noncoding RNA EPCART regulates translation through PI3K/AKT/mTOR pathway and PDCD4 in prostate cancer. Cancer Gene Ther 2024; 31:1536-1546. [PMID: 39147845 PMCID: PMC11489079 DOI: 10.1038/s41417-024-00822-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
While hundreds of cancer-associated long noncoding RNAs (lncRNAs) have been discovered, their functional role in cancer cells is still largely a mystery. An increasing number of lncRNAs are recognized to function in the cytoplasm, e.g., as modulators of translation. Here, we investigated the detailed molecular identity and functional role of EPCART, a lncRNA we previously discovered to be a potential oncogene in prostate cancer (PCa). First, we interrogated the transcript structure of EPCART and then confirmed EPCART to be a non-peptide-coding lncRNA using in silico methods. Pathway analysis of differentially expressed protein-coding genes in EPCART knockout cells implied that EPCART modulates the translational machinery of PCa cells. EPCART was also largely located in the cytoplasm and at the sites of translation. With quantitative proteome analysis on EPCART knockout cells we discovered PDCD4, an inhibitor of protein translation, to be increased by EPCART reduction. Further studies indicated that the inhibitory effect of EPCART silencing on translation was mediated by reduced activation of AKT and inhibition of the mTORC1 pathway. Together, our findings identify EPCART as a translation-associated lncRNA that functions via modulation of the PI3K/AKT/mTORC1 pathway in PCa cells. Furthermore, we provide evidence for the prognostic potential of PDCD4 in PCa tumors in connection with EPCART.
Collapse
Affiliation(s)
- Annika Kohvakka
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, 33520, Tampere, Finland
| | - Mina Sattari
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, 33520, Tampere, Finland
| | - Janika Nättinen
- Eye and Vision Research Group, Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland
| | - Ulla Aapola
- Eye and Vision Research Group, Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland
| | - Pavlína Gregorová
- RNAcious Laboratory, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Teuvo L J Tammela
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, 33520, Tampere, Finland
- Department of Urology, Tampere University Hospital, Tampere, Finland
| | - Hannu Uusitalo
- Eye and Vision Research Group, Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland
- Tays Eye Centre, Tampere University Hospital, 33520, Tampere, Finland
| | - L Peter Sarin
- RNAcious Laboratory, Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
- HiLIFE Helsinki Institute of Life Science, University of Helsinki, 00014, Helsinki, Finland
| | - Tapio Visakorpi
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, 33520, Tampere, Finland.
- Fimlab Laboratories Ltd, Tampere University Hospital, 00014, Tampere, Finland.
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, 70211, Kuopio, Finland.
| |
Collapse
|
3
|
Prosdocimi E, Carpanese V, Todesca LM, Varanita T, Bachmann M, Festa M, Bonesso D, Perez-Verdaguer M, Carrer A, Velle A, Peruzzo R, Muccioli S, Doni D, Leanza L, Costantini P, Stein F, Rettel M, Felipe A, Edwards MJ, Gulbins E, Cendron L, Romualdi C, Checchetto V, Szabo I. BioID-based intact cell interactome of the Kv1.3 potassium channel identifies a Kv1.3-STAT3-p53 cellular signaling pathway. SCIENCE ADVANCES 2024; 10:eadn9361. [PMID: 39231216 PMCID: PMC11373599 DOI: 10.1126/sciadv.adn9361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024]
Abstract
Kv1.3 is a multifunctional potassium channel implicated in multiple pathologies, including cancer. However, how it is involved in disease progression is not fully clear. We interrogated the interactome of Kv1.3 in intact cells using BioID proximity labeling, revealing that Kv1.3 interacts with STAT3- and p53-linked pathways. To prove the relevance of Kv1.3 and of its interactome in the context of tumorigenesis, we generated stable melanoma clones, in which ablation of Kv1.3 remodeled gene expression, reduced proliferation and colony formation, yielded fourfold smaller tumors, and decreased metastasis in vivo in comparison to WT cells. Kv1.3 deletion or pharmacological inhibition of mitochondrial Kv1.3 increased mitochondrial Reactive Oxygen Species release, decreased STAT3 phosphorylation, stabilized the p53 tumor suppressor, promoted metabolic switch, and altered the expression of several BioID-identified Kv1.3-networking proteins in tumor tissues. Collectively, our work revealed the tumor-promoting Kv1.3-interactome landscape, thus opening the way to target Kv1.3 not only as an ion-conducting entity but also as a signaling hub.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Andrea Carrer
- Department of Biology, University of Padova, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Angelo Velle
- Department of Biology, University of Padova, Padova, Italy
| | | | | | - Davide Doni
- Department of Biology, University of Padova, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | | | | | | | - Antonio Felipe
- Molecular Physiology Laboratory, Department de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | | | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Laura Cendron
- Department of Biology, University of Padova, Padova, Italy
| | | | | | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
4
|
Delenko J, Xue X, Chatterjee PK, Hyman N, Shih AJ, Adelson RP, Safaric Tepes P, Gregersen PK, Metz CN. Quercetin enhances decidualization through AKT-ERK-p53 signaling and supports a role for senescence in endometriosis. Reprod Biol Endocrinol 2024; 22:100. [PMID: 39118090 PMCID: PMC11308242 DOI: 10.1186/s12958-024-01265-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Patients with endometriosis suffer with chronic pelvic pain and infertility, and from the lack of pharmacologic therapies that consistently halt disease progression. Differences in the endometrium of patients with endometriosis vs. unaffected controls are well-documented. Specifically, shed endometrial tissues (delivered to the pelvic cavity via retrograde menstruation) reveal that a subset of stromal cells exhibiting pro-inflammatory, pro-fibrotic, and pro-senescence-like phenotypes is enhanced in endometriosis patients compared to controls. Additionally, cultured biopsy-derived endometrial stromal cells from endometriosis patients exhibit impaired decidualization, a defined differentiation process required for human embryo implantation and pregnancy. Quercetin, a senolytic agent, shows therapeutic potential for pulmonary fibrosis, a disorder attributed to senescent pulmonary fibroblasts. In rodent models of endometriosis, quercetin shows promise, and quercetin improves decidualization in vitro. However, the exact mechanisms are not completely understood. Therefore, we investigated the effects of quercetin on menstrual effluent-derived endometrial stromal cells from endometriosis patients and unaffected controls to define the signaling pathways underlying quercetin's effects on endometrial stromal cells. METHODS Menstrual effluent-derived endometrial stromal cells were collected and cultured from unaffected controls and endometriosis patients and then, low passage cells were treated with quercetin (25 µM) under basal or standard decidualization conditions. Decidualization responses were analyzed by measuring the production of IGFBP1 and PRL. Also, the effects of quercetin on intracellular cAMP levels and cellular oxidative stress responses were measured. Phosphokinase arrays, western blotting, and flow cytometry methods were performed to define the effects of quercetin on various signaling pathways and the potential mechanistic roles of quercetin. RESULTS Quercetin significantly promotes decidualization of control- and endometriosis-endometrial stromal cells. Quercetin substantially reduces the phosphorylation of multiple signaling molecules in the AKT and ERK1/2 pathways, while enhancing the phosphorylation of p53 and total p53 levels. Furthermore, p53 inhibition blocks decidualization while p53 activation promotes decidualization. Finally, we provide evidence that quercetin increases apoptosis of endometrial stromal cells with a senescent-like phenotype. CONCLUSIONS These data provide insight into the mechanisms of action of quercetin on endometrial stromal cells and warrant future clinical trials to test quercetin and other senolytics for treating endometriosis.
Collapse
Affiliation(s)
- Julia Delenko
- The Donald and Barbara Zucker School of Medicine, Hempstead, NY, 11549, USA
| | - Xiangying Xue
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Prodyot K Chatterjee
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Nathaniel Hyman
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Andrew J Shih
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Robert P Adelson
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Polona Safaric Tepes
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Peter K Gregersen
- The Donald and Barbara Zucker School of Medicine, Hempstead, NY, 11549, USA.
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
| | - Christine N Metz
- The Donald and Barbara Zucker School of Medicine, Hempstead, NY, 11549, USA.
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
| |
Collapse
|
5
|
Kase AM, Gleba J, Miller JL, Miller E, Petit J, Barrett MT, Zhou Y, Parent EE, Cai H, Knight JA, Orme J, Reynolds J, Durham WF, Metz TM, Meurice N, Edenfield B, Alasonyalilar Demirer A, Bilgili A, Hickman PG, Pawlush ML, Marlow L, Wickland DP, Tan W, Copland JA. Patient-Derived Tumor Xenograft Study with CDK4/6 Inhibitor Plus AKT Inhibitor for the Management of Metastatic Castration-Resistant Prostate Cancer. Mol Cancer Ther 2024; 23:823-835. [PMID: 38442920 DOI: 10.1158/1535-7163.mct-23-0296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 12/04/2023] [Accepted: 02/28/2024] [Indexed: 03/07/2024]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is an aggressive malignancy with poor outcomes. To investigate novel therapeutic strategies, we characterized three new metastatic prostate cancer patient derived-tumor xenograft (PDTX) models and developed 3D spheroids from each to investigate molecular targeted therapy combinations including CDK4/6 inhibitors (CDK4/6i) with AKT inhibitors (ATKi). Metastatic prostate cancer tissue was collected and three PDTX models were established and characterized using whole-exome sequencing. PDTX 3D spheroids were developed from these three PDTXs to show resistance patterns and test novel molecular-targeted therapies. CDK4/6i's were combined with AKTi's to assess synergistic antitumor response to prove our hypothesis that blockade of AKT overcomes drug resistance to CDK4/6i. This combination was evaluated in PDTX three-dimensional (3D) spheroids and in vivo experiments with responses measured by tumor volumes, PSA, and Ga-68 PSMA-11 PET-CT imaging. We demonstrated CDK4/6i's with AKTi's possess synergistic antitumor activity in three mCRPC PDTX models. These models have multiple unique pathogenic and deleterious genomic alterations with resistance to single-agent CDK4/6i's. Despite this, combination therapy with AKTi's was able to overcome resistance mechanisms. The IHC and Western blot analysis confirmed on target effects, whereas tumor volume, serum PSA ELISA, and radionuclide imaging demonstrated response to therapy with statistically significant SUV differences seen with Ga-68 PSMA-11 PET-CT. These preclinical data demonstrating antitumor synergy by overcoming single-agent CDK 4/6i as well as AKTi drug resistance provide the rational for a clinical trial combining a CDK4/6i with an AKTi in patients with mCRPC whose tumor expresses wild-type retinoblastoma 1.
Collapse
Affiliation(s)
- Adam M Kase
- Division of Hematology-Oncology, Mayo Clinic Jacksonville, Florida
| | - Justyna Gleba
- Cancer Biology Department, Mayo Clinic Jacksonville, Florida
| | - James L Miller
- Cancer Biology Department, Mayo Clinic Jacksonville, Florida
| | - Erin Miller
- Cancer Biology Department, Mayo Clinic Jacksonville, Florida
| | - Joachim Petit
- Division of Hematology-Oncology, Mayo Clinic Scottsdale, Arizona
| | | | - Yumei Zhou
- Division of Hematology-Oncology, Mayo Clinic Scottsdale, Arizona
| | | | - Hancheng Cai
- Radiology Department, Mayo Clinic Jacksonville, Florida
| | - Joshua A Knight
- Cancer Biology Department, Mayo Clinic Jacksonville, Florida
| | - Jacob Orme
- Division of Hematology-Oncology, Mayo Clinic Rochester, Minnesota
| | - Jordan Reynolds
- Department of Laboratory Medicine and Pathology, Mayo Clinic Jacksonville, Florida
| | | | - Thomas M Metz
- Charles River Discovery Research Services Germany, Freiburg, Germany
| | - Nathalie Meurice
- Division of Hematology-Oncology, Mayo Clinic Scottsdale, Arizona
| | | | | | - Ahmet Bilgili
- Cancer Biology Department, Mayo Clinic Jacksonville, Florida
| | | | | | - Laura Marlow
- Cancer Biology Department, Mayo Clinic Jacksonville, Florida
| | - Daniel P Wickland
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic Jacksonville, Florida
| | - Winston Tan
- Division of Hematology-Oncology, Mayo Clinic Jacksonville, Florida
| | - John A Copland
- Cancer Biology Department, Mayo Clinic Jacksonville, Florida
| |
Collapse
|
6
|
Malla A, Gupta S, Sur R. Glycolytic enzymes in non-glycolytic web: functional analysis of the key players. Cell Biochem Biophys 2024; 82:351-378. [PMID: 38196050 DOI: 10.1007/s12013-023-01213-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/26/2023] [Indexed: 01/11/2024]
Abstract
To survive in the tumour microenvironment, cancer cells undergo rapid metabolic reprograming and adaptability. One of the key characteristics of cancer is increased glycolytic selectivity and decreased oxidative phosphorylation (OXPHOS). Apart from ATP synthesis, glycolysis is also responsible for NADH regeneration and macromolecular biosynthesis, such as amino acid biosynthesis and nucleotide biosynthesis. This allows cancer cells to survive and proliferate even in low-nutrient and oxygen conditions, making glycolytic enzymes a promising target for various anti-cancer agents. Oncogenic activation is also caused by the uncontrolled production and activity of glycolytic enzymes. Nevertheless, in addition to conventional glycolytic processes, some glycolytic enzymes are involved in non-canonical functions such as transcriptional regulation, autophagy, epigenetic changes, inflammation, various signaling cascades, redox regulation, oxidative stress, obesity and fatty acid metabolism, diabetes and neurodegenerative disorders, and hypoxia. The mechanisms underlying the non-canonical glycolytic enzyme activities are still not comprehensive. This review summarizes the current findings on the mechanisms fundamental to the non-glycolytic actions of glycolytic enzymes and their intermediates in maintaining the tumor microenvironment.
Collapse
Affiliation(s)
- Avirup Malla
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata, India
| | - Suvroma Gupta
- Department of Aquaculture Management, Khejuri college, West Bengal, Baratala, India.
| | - Runa Sur
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata, India.
| |
Collapse
|
7
|
Yun HM, Kwon HS, Lee JY, Park KR. Vitexicarpin Induces Apoptosis and Inhibits Metastatic Properties via the AKT-PRAS40 Pathway in Human Osteosarcoma. Int J Mol Sci 2024; 25:3582. [PMID: 38612399 PMCID: PMC11012096 DOI: 10.3390/ijms25073582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Osteosarcoma, which has poor prognosis after metastasis, is the most common type of bone cancer in children and adolescents. Therefore, plant-derived bioactive compounds are being actively developed for cancer therapy. Artemisia apiacea Hance ex Walp. is a traditional medicinal plant native to Eastern Asia, including China, Japan, and Korea. Vitexicarpin (Vitex), derived from A. apiacea, has demonstrated analgesic, anti-inflammatory, antitumour, and immunoregulatory properties; however, there are no published studies on Vitex isolated from the aerial parts of A. apiacea. Thus, this study aimed to evaluate the antitumour activity of Vitex against human osteosarcoma cells. In the present study, Vitex (>99% purity) isolated from A. apiacea induced significant cell death in human osteosarcoma MG63 cells in a dose- and time-dependent manner; cell death was mediated by apoptosis, as evidenced by the appearance of cleaved-PARP, cleaved-caspase 3, anti-apoptotic proteins (Survivin and Bcl-2), pro-apoptotic proteins (Bax), and cell cycle-related proteins (Cyclin D1, Cdk4, and Cdk6). Additionally, a human phosphokinase array proteome profiler revealed that Vitex suppressed AKT-dependent downstream kinases. Further, Vitex reduced the phosphorylation of PRAS40, which is associated with autophagy and metastasis, induced autophagosome formation, and suppressed programmed cell death and necroptosis. Furthermore, Vitex induced antimetastatic activity by suppressing the migration and invasion of MMP13, which is the primary protease that degrades type I collagen for tumour-induced osteolysis in bone tissues and preferential metastasis sites. Taken together, our results suggest that Vitex is an attractive target for treating human osteosarcoma.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyun Sook Kwon
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (H.S.K.); (J.Y.L.)
| | - Joon Yeop Lee
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (H.S.K.); (J.Y.L.)
| | - Kyung-Ran Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
| |
Collapse
|
8
|
Martins C, Rasbach E, Heppt MV, Singh P, Kulcsar Z, Holzgruber J, Chakraborty A, Mucciarone K, Kleffel S, Brandenburg A, Hoetzenecker W, Rahbari NN, DeCaprio JA, Thakuria M, Murphy GF, Ramsey MR, Posch C, Barthel SR, Schatton T. Tumor cell-intrinsic PD-1 promotes Merkel cell carcinoma growth by activating downstream mTOR-mitochondrial ROS signaling. SCIENCE ADVANCES 2024; 10:eadi2012. [PMID: 38241371 PMCID: PMC10798567 DOI: 10.1126/sciadv.adi2012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024]
Abstract
Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer. Inhibitors targeting the programmed cell death 1 (PD-1) immune checkpoint have improved MCC patient outcomes by boosting antitumor T cell immunity. Here, we identify PD-1 as a growth-promoting receptor intrinsic to MCC cells. In human MCC lines and clinical tumors, RT-PCR-based sequencing, immunoblotting, flow cytometry, and immunofluorescence analyses demonstrated PD-1 gene and protein expression by MCC cells. MCC-PD-1 ligation enhanced, and its inhibition or silencing suppressed, in vitro proliferation and in vivo tumor xenograft growth. Consistently, MCC-PD-1 binding to PD-L1 or PD-L2 induced, while antibody-mediated PD-1 blockade inhibited, protumorigenic mTOR signaling, mitochondrial (mt) respiration, and ROS generation. Last, pharmacologic inhibition of mTOR or mtROS reversed MCC-PD-1:PD-L1-dependent proliferation and synergized with PD-1 checkpoint blockade in suppressing tumorigenesis. Our results identify an MCC-PD-1-mTOR-mtROS axis as a tumor growth-accelerating mechanism, the blockade of which might contribute to clinical response in patients with MCC.
Collapse
Affiliation(s)
- Christina Martins
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Erik Rasbach
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Surgery, University Hospital Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Markus V. Heppt
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University (FAU), 91054 Erlangen, Germany
| | - Praveen Singh
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zsofi Kulcsar
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, University Hospital Bonn, 53127 Bonn, Germany
| | - Julia Holzgruber
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology and Venerology, Johannes Kepler University, 4020 Linz, Austria
| | - Asmi Chakraborty
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kyla Mucciarone
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sonja Kleffel
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anne Brandenburg
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, University Hospital Bonn, 53127 Bonn, Germany
| | - Wolfram Hoetzenecker
- Department of Dermatology and Venerology, Johannes Kepler University, 4020 Linz, Austria
| | - Nuh N. Rahbari
- Department of Surgery, University Hospital Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - James A. DeCaprio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA
- Merkel Cell Carcinoma Center of Excellence, Dana-Farber/Brigham and Women’s Hospital Cancer Center, Boston, MA 02115, USA
| | - Manisha Thakuria
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Merkel Cell Carcinoma Center of Excellence, Dana-Farber/Brigham and Women’s Hospital Cancer Center, Boston, MA 02115, USA
| | - George F. Murphy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew R. Ramsey
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christian Posch
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, Vienna Healthcare Group, 1130 Vienna, Austria
- Faculty of Medicine, Sigmund Freud University Vienna, 1020 Vienna, Austria
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Steven R. Barthel
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tobias Schatton
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program of Glyco-Immunology and Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Wang B, Shen XY, Pan LY, Li Z, Chen CJ, Yao YS, Tang DF, Gao W. The HDAC2-MTA3 interaction induces nonsmall cell lung cancer cell migration and invasion by targeting c-Myc and cyclin D1. Mol Carcinog 2023; 62:1630-1644. [PMID: 37401867 DOI: 10.1002/mc.23604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/05/2023]
Abstract
Genome-wide association studies have identified numerous single-nucleotide polymorphisms (SNPs) associated with lung cancer; however, the functions of histone deacetylase 2 (HDAC2) rs13213007 and HDAC2 in nonsmall cell lung cancer (NSCLC) remain unclear. Here we identified HDAC2 rs13213007 as a risk SNP and showed that HDAC2 was upregulated in both peripheral blood mononuclear cells (PBMCs) and NSCLC tissues with the rs13213007 A/A genotype compared with those with the rs13213007 G/G or G/A genotype. Patient clinical data indicated strong associations between rs13213007 genotype and N classification. Immunohistochemical staining confirmed that higher expression of HDAC2 was associated with NSCLC progression. Furthermore, we generated 293T cells with the rs13213007 A/A genotype using CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 gene editing. Chromatin immunoprecipitation sequencing followed by motif analysis showed that HDAC2 can bind to c-Myc in rs13213007 A/A 293T cells. Cell Counting Kit-8, colony formation, wound-healing, and Transwell assays revealed that HDAC2 upregulates c-Myc and cyclin D1 expression and promotes NSCLC cell proliferation, migration, and invasion. Co-immunoprecipitation, quantitative reverse transcription-polymerase chain reaction, and western blot analysis assays showed that MTA3 interacts with HDAC2, decreases HDAC2 expression, and rescues the migration and invasion abilities of NSCLC cells. Taken together, these findings identify HDAC2 as a potential therapeutic biomarker in NSCLC.
Collapse
Affiliation(s)
- Bin Wang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Xiao-Yong Shen
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Lin-Yue Pan
- Department of Respiration, The Affiliated Zhongshan Hospital of Fudan University, Shanghai, China
| | - Zheng Li
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Chun-Ji Chen
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Yuan-Shan Yao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Dong-Fang Tang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| | - Wen Gao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Department of Thoracic Surgery, The Affiliated Huadong Hospital of Fudan University, Shanghai, China
| |
Collapse
|
10
|
Vignesh R, Aradhyam GK. Calnuc-derived nesfatin-1-like peptide is an activator of tumor cell proliferation and migration. FEBS Lett 2023; 597:2288-2300. [PMID: 37539786 DOI: 10.1002/1873-3468.14712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 08/05/2023]
Abstract
Calnuc (nucleobindin-1, nucb1) is a Ca2+ -binding protein involved in the etiology of many human diseases. To understand the functions of calnuc, we have identified a nesfatin-1-like peptide (NLP) in its N terminus that is proteolyzed by a convertase enzyme in the secretory granules of cells. Mutational studies confirm the presence of a proteolytic cleavage site for proprotein convertase subtilisin/kexin type 1 (PCSK1). We demonstrate that NLP regulates Gαq-mediated intracellular Ca2+ dynamics, likely via a G-protein-coupled receptor. NLP treatment to carcinoma cell lines (SCC131 cells) promotes the expression of regulators of cell cycle, proliferation, and clonogenicity by the AKT/mTOR pathway. NLP is causative of augmented migration and epithelial-mesenchymal transition (EMT), illustrating its metastatic propensity and establishing its tumor promotion ability.
Collapse
Affiliation(s)
- Ravichandran Vignesh
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Gopala Krishna Aradhyam
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| |
Collapse
|
11
|
Prasad S, Kumar V, Singh C, Singh A. Crosstalk between phytochemicals and inflammatory signaling pathways. Inflammopharmacology 2023; 31:1117-1147. [PMID: 37022574 DOI: 10.1007/s10787-023-01206-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023]
Abstract
Novel bioactive constituents from natural sources are actively being investigated. The phytochemicals in these phenolic compounds are believed to have a variety of beneficial effects on human health. Several phenolic compounds have been found in plants. The antioxidant potential of phenols has been discussed in numerous studies along with their anti-inflammatory effects on pro-inflammatory cytokine, inducible cyclooxygenase-2, and nitric oxide synthase. Through current study, an attempt is made to outline and highlight a wide variety of inflammation-associated signaling pathways that have been modified by several natural compounds. These signaling pathways include nuclear factor-kappa B (NF-кB), activator protein (AP)-1, protein tyrosine kinases (PTKs), mitogen-activated protein kinases (MAPKs), nuclear factor erythroid 2-related factor 2 (Nrf2) transcription factors, tyrosine phosphatidylinositol 3-kinase (PI3K)/AKT, and the ubiquitin-proteasome system. In light of the influence of natural substances on signaling pathways, their impact on the production of inflammatory mediator is highlighted in this review.
Collapse
Affiliation(s)
- Sonima Prasad
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, H.N.B. Garhwal University, Srinagar, Garhwal, 246174, Uttarakhand, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India.
| |
Collapse
|
12
|
Gao J, Yao M, Chang D, Liu J. mTOR (Mammalian Target of Rapamycin): Hitting the Bull's Eye for Enhancing Neurogenesis After Cerebral Ischemia? Stroke 2023; 54:279-285. [PMID: 36321454 DOI: 10.1161/strokeaha.122.040376] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ischemic stroke remains a leading cause of morbidity and disability around the world. The sequelae of serious neurological damage are irreversible due to body's own limited repair capacity. However, endogenous neurogenesis induced by cerebral ischemia plays a critical role in the repair and regeneration of impaired neural cells after ischemic brain injury. mTOR (mammalian target of rapamycin) kinase has been suggested to regulate neural stem cells ability to self-renew and differentiate into proliferative daughter cells, thus leading to improved cell growth, proliferation, and survival. In this review, we summarized the current evidence to support that mTOR signaling pathways may enhance neurogenesis, angiogenesis, and synaptic plasticity following cerebral ischemia, which could highlight the potential of mTOR to be a viable therapeutic target for the treatment of ischemic brain injury.
Collapse
Affiliation(s)
- Jiale Gao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, China (J.G., M.Y., J.L.)
| | - Mingjiang Yao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, China (J.G., M.Y., J.L.)
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, Australia (D.C.)
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Medica, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, China (J.G., M.Y., J.L.)
| |
Collapse
|
13
|
mTOR Complex 1 Content and Regulation Is Adapted to Animal Longevity. Int J Mol Sci 2022; 23:ijms23158747. [PMID: 35955882 PMCID: PMC9369240 DOI: 10.3390/ijms23158747] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Decreased content and activity of the mechanistic target of rapamycin (mTOR) signalling pathway, as well as the mTOR complex 1 (mTORC1) itself, are key traits for animal species and human longevity. Since mTORC1 acts as a master regulator of intracellular metabolism, it is responsible, at least in part, for the longevous phenotype. Conversely, increased content and activity of mTOR signalling and mTORC1 are hallmarks of ageing. Additionally, constitutive and aberrant activity of mTORC1 is also found in age-related diseases such as Alzheimer’s disease (AD) and cancer. The downstream processes regulated through this network are diverse, and depend upon nutrient availability. Hence, multiple nutritional strategies capable of regulating mTORC1 activity and, consequently, delaying the ageing process and the development of age-related diseases, are under continuous study. Among these, the restriction of calories is still the most studied and robust intervention capable of downregulating mTOR signalling and feasible for application in the human population.
Collapse
|
14
|
Mohamed HT, El-Sharkawy AA, El-Shinawi M, Schneider RJ, Mohamed MM. Inflammatory Breast Cancer: The Secretome of HCMV+ Tumor-Associated Macrophages Enhances Proliferation, Invasion, Colony Formation, and Expression of Cancer Stem Cell Markers. Front Oncol 2022; 12:899622. [PMID: 35847899 PMCID: PMC9281473 DOI: 10.3389/fonc.2022.899622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a highly aggressive phenotype of breast cancer that is characterized by a high incidence early metastasis. We previously reported a significant association of human cytomegalovirus (HCMV) DNA in the carcinoma tissues of IBC patients but not in the adjacent normal tissues. HCMV-infected macrophages serve as “mobile vectors” for spreading and disseminating virus to different organs, and IBC cancer tissues are highly infiltrated by tumor-associated macrophages (TAMs) that enhance IBC progression and promote breast cancer stem cell (BCSC)-like properties. Therefore, there is a need to understand the role of HCMV-infected TAMs in IBC progression. The present study aimed to test the effect of the secretome (cytokines and secreted factors) of TAMs derived from HCMV+ monocytes isolated from IBC specimens on the proliferation, invasion, and BCSC abundance when tested on the IBC cell line SUM149. HCMV+ monocytes were isolated from IBC patients during modified radical mastectomy surgery and tested in vitro for polarization into TAMs using the secretome of SUM149 cells. MTT, clonogenic, invasion, real-time PCR arrays, PathScan Intracellular Signaling array, and cytokine arrays were used to characterize the secretome of HCMV+ TAMs for their effect on the progression of SUM149 cells. The results showed that the secretome of HCMV+ TAMs expressed high levels of IL-6, IL-8, and MCP-1 cytokines compared to HCMV- TAMs. In addition, the secretome of HCMV+ TAMs induced the proliferation, invasion, colony formation, and expression of BCSC-related genes in SUM149 cells compared to mock untreated cells. In addition, the secretome of HCMV+ TAMs activated the phosphorylation of intracellular signaling molecules p-STAT3, p-AMPKα, p-PRAS40, and p-SAPK/JNK in SUM149 cells. In conclusion, this study shows that the secretome of HCMV+ TAMs enhances the proliferation, invasion, colony formation, and BCSC properties by activating the phosphorylation of p-STAT3, p-AMPKα, p-PRAS40, and p-SAPK/JNK intracellular signaling molecules in IBC cells.
Collapse
Affiliation(s)
- Hossam Taha Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
- *Correspondence: Hossam Taha Mohamed,
| | | | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Galala University, Suez, Egypt
| | - Robert J. Schneider
- Department of Microbiology, School of Medicine, New York University, New York, NY, United States
| | - Mona Mostafa Mohamed
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
- Sector of International Cooperation, Galala University, Suez, Egypt
| |
Collapse
|
15
|
Breedon SA, Gupta A, Storey KB. Regulation of Apoptosis and Autophagy During Anoxia in the Freshwater Crayfish, Faxonius virilis. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:626-639. [PMID: 35567599 DOI: 10.1007/s10126-022-10132-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
The ability of an animal to survive prolonged periods of oxygen deprivation is a critical area of study, both in terms of its importance to better understanding the physiology of these incredible animals and to its potential applicability to medical fields. The freshwater crayfish, Faxonius virilis, is one such animal capable of resisting anoxia, but it remains understudied and much of the metabolic mechanisms underlying this anoxia tolerance remain largely unprofiled. This study examines the activity and regulation of apoptosis and autophagy in F. virilis in response to 20-h anoxia. Apoptosis signaling was assessed through pro- and anti-apoptosis targets, whereas autophagy was assessed via expression response of multiple autophagy proteins. An anoxia-triggered, tissue-specific result arose, potentially based on the importance of individual organ integrity through hypometabolism. Tail muscle, which showed increased expression profiles of all three target groups, contrasted with hepatopancreas, which appeared to not be susceptible to either apoptotic or autophagic signaling during anoxia. This is likely due to the importance of the hepatopancreas, given that apoptosis or autophagy of this organ at any significant level could be fatal to the organism. The data provides a comprehensive overview of the responses and integration of multiple stress-responsive signaling pathways in F. virilis that provide a novel contribution to our understanding of pro-survival mechanisms supporting invertebrate anoxia resistance.
Collapse
Affiliation(s)
- Sarah A Breedon
- Institute of Biochemistry and Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Aakriti Gupta
- Institute of Biochemistry and Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Kenneth B Storey
- Institute of Biochemistry and Department of Biology, Carleton University, Ottawa, ON, Canada.
| |
Collapse
|
16
|
Huang Y, Motta E, Nanvuma C, Kuhrt LD, Yuan Y, Xia P, Lubas M, Zhu S, Schnauss M, Qazi N, Hu F, Zhang H, Lei T, Synowitz M, Flüh C, Kettenmann H. Microglia/macrophage-derived human CCL18 promotes glioma progression via CCR8-ACP5 axis analyzed in humanized slice model. Cell Rep 2022; 39:110670. [PMID: 35417708 DOI: 10.1016/j.celrep.2022.110670] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 02/21/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
Factors released from glioma-associated microglia/macrophages (GAMs) play a crucial role in glioblastoma multiforme (GBM) progression. Here, we study the importance of CCL18, a cytokine expressed in human but not in rodent GAMs, as a modulator of glioma growth. Since CCL18 signaling could not be studied in classical mouse glioma models, we developed an approach by transplanting induced pluripotent stem cell-derived human microglia and human glioma cells into mouse brain slices depleted of their intrinsic microglia. We observe that CCL18 promotes glioma cell growth and invasion. Chemokine (C-C motif) receptor 8 (CCR8) is identified as a functional receptor for CCL18 on glioma cells, and ACP5 (acid phosphatase 5) is revealed as an important part of the downstream signaling cascade for mediating glioma growth. We conclude, based on the results from an in vitro, ex vivo humanized glioma model and an in vivo GBM model that microglia/macrophage-derived CCL18 promotes glioma growth.
Collapse
Affiliation(s)
- Yimin Huang
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany; Charité-Universitätsmedizin, 10117 Berlin, Germany; Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Edyta Motta
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | - Cynthia Nanvuma
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany; Department of Neurosurgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Leonard D Kuhrt
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany; Charité-Universitätsmedizin, 10117 Berlin, Germany
| | - Yang Yuan
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | - Pengfei Xia
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | - Malgorzata Lubas
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | - Shuai Zhu
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | - Marina Schnauss
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | - Niyeti Qazi
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany
| | - Feng Hu
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Michael Synowitz
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Charlotte Flüh
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany; Department of Neurosurgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany.
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert Roessle Strasse 10, 13125 Berlin, Germany; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
17
|
Pan Q, Wang L, Liu Y, Li M, Zhang Y, Peng W, Deng T, Peng ML, Jiang JQ, Tang J, Wang J, Duan HX, Fan SS. Knockdown of POLQ interferes the development and progression of hepatocellular carcinoma through regulating cell proliferation, apoptosis and migration. Cancer Cell Int 2021; 21:482. [PMID: 34517891 PMCID: PMC8436534 DOI: 10.1186/s12935-021-02178-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Background DNA Polymerase Theta (POLQ) is a DNA polymerase involved in error-prone translesion DNA synthesis (TLS) and error-prone repair of DNA double-strand breaks (DSBs), whose function in hepatocellular carcinoma has not been investigated. Methods In the present study, both the data collected from the Cancer Genome Atlas (TCGA) and our group’s results showed higher POLQ expression in HCC tissues than the para-cancerous tissues, which was associated with higher malignancy and poor prognosis. POLQ knockdown HCC cell model (shPOLQ) was constructed along with the corresponding negative control (shCtrl) through lentivirus infection for loss-of-function study. Results We found that, upon knockdown of POLQ, the proliferation and migration of HCC cells decreased and apoptosis percentage increased. Moreover, the percentage of cells in G2 phase significantly increased in shPOLQ group compared with shCtrl group. Xenografts in mice grafted with shPOLQ cells grew much slower than that transplanted with shCtrl cells, and expressed lower Ki67 level. Furthermore, an apoptosis-related signaling array was used to explore the involvement of downstream signaling pathways, suggesting the enhanced phosphorylation of HSP27 and JNK, and the de-activation of mTOR, PRAS40, ERK1/2 and STAT3 pathways. Conclusions Collectively, our study revealed that POLQ may participate in the development of HCC, depletion of which may be a promising treatment strategy for HCC.
Collapse
Affiliation(s)
- Qi Pan
- Department of Hepatic Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lu Wang
- Department of Hepatic Surgery, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yu Liu
- Department of Pathology, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Changsha, 410000, Hunan, China
| | - Min Li
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China
| | - Yao Zhang
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China
| | - Wei Peng
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China
| | - Tan Deng
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China
| | - Mei-Ling Peng
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China
| | - Jin-Qiong Jiang
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China
| | - Jiao Tang
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China
| | - Jingjing Wang
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, 410000, Hunan, China
| | - Hua-Xin Duan
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China.
| | - Sha-Sha Fan
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, 410000, Hunan, China.
| |
Collapse
|
18
|
Zhou Q, Tang S, Zhang X, Chen L. Targeting PRAS40: a novel therapeutic strategy for human diseases. J Drug Target 2021; 29:703-715. [PMID: 33504218 DOI: 10.1080/1061186x.2021.1882470] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proline-rich Akt substrate of 40 kD (PRAS40) is not only the substrate of protein kinase B (PKB/Akt), but also the binding protein of 14-3-3 protein. PRAS40 is expressed in a variety of tissues in vivo and has multiple phosphorylation sites, which its activity is closely related to phosphorylation. Studies have shown that PRAS40 is involved in regulating cell growth, cell apoptosis, oxidative stress, autophagy and angiogenesis, as well as various of signalling pathways such as mammalian target of mammalian target rapamycin (mTOR), protein kinase B (PKB/Akt), nuclear factor kappa-B(NF-κB), proto-oncogene serine/threonine-protein kinase PIM-1(PIM1) and pyruvate kinase M2 (PKM2). The interactive roles between PRAS40 and these signal proteins were analysed by bioinformatics in this paper. Moreover, it is of great necessity for analyse the important roles of PRAS40 in some human diseases including cardiovascular disease, ischaemia-reperfusion injury, neurodegenerative disease, cancer, diabetes and other metabolic diseases. Finally, the effects of miRNA on the regulation of PRAS40 function and the occurrence and development of PRAS40-related diseases are also discussed. Overall, PRAS40 is expected to be a drug target and provide a new treatment strategy for human diseases.
Collapse
Affiliation(s)
- Qun Zhou
- Hunan Province Key Laboratory for Antibody- Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody- Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, China
| | - Xianhui Zhang
- Orthopedics Department, Dongkou People's Hospital, Dongkou, China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target, New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| |
Collapse
|
19
|
Wang W, Sindrewicz-Goral P, Chen C, Duckworth CA, Pritchard DM, Rhodes JM, Yu LG. Appearance of peanut agglutinin in the blood circulation after peanut ingestion promotes endothelial secretion of metastasis-promoting cytokines. Carcinogenesis 2021; 42:1079-1088. [PMID: 34223877 PMCID: PMC8643467 DOI: 10.1093/carcin/bgab059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/07/2021] [Accepted: 07/02/2021] [Indexed: 11/18/2022] Open
Abstract
Peanut agglutinin (PNA) is a carbohydrate-binding protein in peanuts that accounts for ~0.15% peanut weight. PNA is highly resistant to cooking and digestion and is rapidly detectable in the blood after peanut consumption. Our previous studies have shown that circulating PNA mimics the actions of endogenous galactoside-binding protein galectin-3 by interaction with tumour cell-associated MUC1 and promotes circulating tumour cell metastatic spreading. The present study shows that circulating PNA interacts with micro- as well as macro-vascular endothelial cells and induces endothelial secretion of cytokines MCP-1 (CCL2) and IL-6 in vitro and in vivo. The increased secretion of these cytokines autocrinely/paracrinely enhances the expression of endothelial cell surface adhesion molecules including integrins, VCAM and selectin, leading to increased tumour cell-endothelial adhesion and endothelial tubule formation. Binding of PNA to endothelial surface MCAM (CD146), via N-linked glycans, and subsequent activation of PI3K-AKT-PREAS40 signalling is here shown responsible for PNA-induced secretion of MCP-1 and IL-6 by vascular endothelium. Thus, in addition to its influence on promoting tumour cell spreading by interaction with tumour cell-associated MUC1, circulating PNA might also influence metastasis by enhancing the secretion of metastasis-promoting MCP-1 and IL-6 from the vascular endothelium.
Collapse
Affiliation(s)
- Weikun Wang
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Paulina Sindrewicz-Goral
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Chen Chen
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Carrie A Duckworth
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - David Mark Pritchard
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Jonathan M Rhodes
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| | - Lu-Gang Yu
- The Henry Wellcome Laboratory of Molecular and Cellular Gastroenterology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, United Kingdom
| |
Collapse
|
20
|
Wu JH, Limmer AL, Narayanan D, Doan HQ, Simonette RA, Rady PL, Tyring SK. The novel AKT inhibitor afuresertib suppresses human Merkel cell carcinoma MKL-1 cell growth. Clin Exp Dermatol 2021; 46:1551-1554. [PMID: 34115902 DOI: 10.1111/ced.14798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/29/2022]
Abstract
Merkel cell carcinoma (MCC) is a highly aggressive neuroendocrine neoplasm of the skin, which has an exceedingly poor prognosis. The AKT/mammalian target of rapamycin (mTOR) signalling pathway, which plays a pivotal role in the modulation of protein synthesis and cell survival, has been shown to be extremely important for Merkel cell carcinogenesis. In the current study, we found that AKT has important regulatory functions in MCC cells and that inhibition of AKT with the novel ATP-competitive AKT inhibitor, afuresertib, has widespread effects on proliferative pathways. In particular, we found that treatment of MCC cells with afuresertib led to deactivation of mTOR and glycogen synthase kinase 3 pathway proteins while increasing activation of proapoptotic pathways through the upregulation of p16 expression and phosphomodulation of the B-cell lymphoma-2-associated death promoter. Overall, afuresertib treatment led to significant and robust inhibition of MCC cell proliferation, thus raising intriguing questions regarding the potential efficacy of AKT inhibition for the future clinical management of MCC.
Collapse
Affiliation(s)
- J H Wu
- Department of Dermatology, McGovern Medical School at The University of Texas Health Science Center, Houston, TX, USA.,Ronald O. Perelman Department of Dermatology, New York University, New York, NY, USA
| | - A L Limmer
- Department of Dermatology, McGovern Medical School at The University of Texas Health Science Center, Houston, TX, USA
| | - D Narayanan
- Department of Dermatology, McGovern Medical School at The University of Texas Health Science Center, Houston, TX, USA
| | - H Q Doan
- Department of Dermatology, McGovern Medical School at The University of Texas Health Science Center, Houston, TX, USA.,Department of Dermatology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - R A Simonette
- Department of Dermatology, McGovern Medical School at The University of Texas Health Science Center, Houston, TX, USA
| | - P L Rady
- Department of Dermatology, McGovern Medical School at The University of Texas Health Science Center, Houston, TX, USA
| | - S K Tyring
- Department of Dermatology, McGovern Medical School at The University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
21
|
Gonzalez de Mejia E, Castañeda-Reyes ED, Mojica L, Dia V, Wang H, Wang T, Johnson LA. Potential Health Benefits Associated with Lunasin Concentration in Dietary Supplements and Lunasin-Enriched Soy Extract. Nutrients 2021; 13:1618. [PMID: 34065911 PMCID: PMC8150303 DOI: 10.3390/nu13051618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/22/2022] Open
Abstract
Lunasin has demonstrated antioxidative, anti-inflammatory, and chemopreventive properties. The objectives were to evaluate the concentration of lunasin in different lunasin-based commercial dietary supplements, to produce a lunasin-enriched soy extract (LESE) using a two-step pilot-plant-based ultrafiltration process, and to evaluate their biological potential in vitro. LESE was produced using 30 and 1 kDa membranes in a custom-made ultrafiltration skid. Lunasin was quantified in eight products and LESE. Lunasin concentrations of the lunasin-based products ranged from 9.2 ± 0.6 to 25.7 ± 1.1 mg lunasin/g protein. The LESE extract contained 58.2 mg lunasin/g protein, up to 6.3-fold higher lunasin enrichment than lunasin-based dietary supplements. Antioxidant capacity ranged from 121.5 mmol Trolox equivalents (TE)/g in Now® Kids to 354.4 mmol TE/g in LESE. Histone acetyltransferase (HAT) inhibition ranged from 5.3% on Soy Sentials® to 38.3% on synthetic lunasin. ORAC and lunasin concentrations were positively correlated, and HAT and lunasin concentrations were negatively correlated (p < 0.05). Melanoma B16-F10 and A375 cells treated with lunasin showed dose-dependent inhibitory potential (IC50 equivalent to 330 and 370 μM lunasin, respectively). Lunasin showed protein kinase B expression (57 ± 14%) compared to the control (100%) in B16-F10. Lunasin concentration found in commercial products and lunasin-enriched soy extract could exert benefits to consumers.
Collapse
Affiliation(s)
- Elvira Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (E.D.C.-R.); (L.M.); (V.D.)
| | - Erick Damian Castañeda-Reyes
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (E.D.C.-R.); (L.M.); (V.D.)
| | - Luis Mojica
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (E.D.C.-R.); (L.M.); (V.D.)
- Tecnología Alimentaria, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A. C., CIATEJ, Guadalajara 44270, Mexico
| | - Vermont Dia
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (E.D.C.-R.); (L.M.); (V.D.)
| | - Hui Wang
- Center for Crops Utilization Research, Iowa State University, Ames, IA 50011, USA; (H.W.); (T.W.); (L.A.J.)
| | - Toni Wang
- Center for Crops Utilization Research, Iowa State University, Ames, IA 50011, USA; (H.W.); (T.W.); (L.A.J.)
| | - Lawrence A. Johnson
- Center for Crops Utilization Research, Iowa State University, Ames, IA 50011, USA; (H.W.); (T.W.); (L.A.J.)
| |
Collapse
|
22
|
Yin S, Liu L, Gan W. The Roles of Post-Translational Modifications on mTOR Signaling. Int J Mol Sci 2021; 22:ijms22041784. [PMID: 33670113 PMCID: PMC7916890 DOI: 10.3390/ijms22041784] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a master regulator of cell growth, proliferation, and metabolism by integrating various environmental inputs including growth factors, nutrients, and energy, among others. mTOR signaling has been demonstrated to control almost all fundamental cellular processes, such as nucleotide, protein and lipid synthesis, autophagy, and apoptosis. Over the past fifteen years, mapping the network of the mTOR pathway has dramatically advanced our understanding of its upstream and downstream signaling. Dysregulation of the mTOR pathway is frequently associated with a variety of human diseases, such as cancers, metabolic diseases, and cardiovascular and neurodegenerative disorders. Besides genetic alterations, aberrancies in post-translational modifications (PTMs) of the mTOR components are the major causes of the aberrant mTOR signaling in a number of pathologies. In this review, we summarize current understanding of PTMs-mediated regulation of mTOR signaling, and also update the progress on targeting the mTOR pathway and PTM-related enzymes for treatment of human diseases.
Collapse
|
23
|
Kaur H, Moreau R. mTORC1 silencing during intestinal epithelial Caco-2 cell differentiation is mediated by the activation of the AMPK/TSC2 pathway. Biochem Biophys Res Commun 2021; 545:183-188. [PMID: 33561653 DOI: 10.1016/j.bbrc.2021.01.070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) signaling is the prototypical pathway regulating protein synthesis and cell proliferation. The level of mTORC1 activity is high in intestinal stem cells located at the base of the crypts and thought to gradually decrease as transit-amplifying cells migrate out of the crypts and differentiate into enterocytes, goblet cells or enteroendocrine cells along the epithelium. The unknown mechanism responsible for the silencing of intestinal epithelium mTORC1 during cell differentiation was investigated in Caco-2 cells, which spontaneously differentiate into enterocytes in standard growth medium. The results show that TSC2, an upstream negative regulator of mTORC1 was central to mTORC1 silencing in differentiated Caco-2 cells. AMPK-mediated activation of TSC2 (Ser1387) and repression of Raptor (Ser792), an essential component of mTORC1, were stimulated in differentiated Caco-2 cells. ERK1/2-mediated repression of TSC2 (Ser664) seen in undifferentiated Caco-2 cells was lifted in differentiated cells. IRS-1-mediated activation of AKT (Thr308) phosphorylation was stimulated in differentiated Caco-2 cells and may be involved in cross-pathway repression of ERK1/2. Additionally, PRAS40 (Thr246) phosphorylation was decreased in differentiated Caco-2 cells compared to undifferentiated cells allowing dephosphorylated PRAS40 to displace Raptor thereby repressing mTORC1 kinase activity.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
24
|
Jeitany M, Prabhu A, Dakle P, Pathak E, Madan V, Kanojia D, Mukundan V, Jiang YY, Landesman Y, Tam WL, Kappei D, Koeffler HP. Novel carfilzomib-based combinations as potential therapeutic strategies for liposarcomas. Cell Mol Life Sci 2021; 78:1837-1851. [PMID: 32851475 PMCID: PMC7904719 DOI: 10.1007/s00018-020-03620-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/19/2020] [Accepted: 08/07/2020] [Indexed: 01/09/2023]
Abstract
Proteasome inhibitors, such as bortezomib and carfilzomib, have shown efficacy in anti-cancer therapy in hematological diseases but not in solid cancers. Here, we found that liposarcomas (LPS) are susceptible to proteasome inhibition, and identified drugs that synergize with carfilzomib, such as selinexor, an inhibitor of XPO1-mediated nuclear export. Through quantitative nuclear protein profiling and phospho-kinase arrays, we identified potential mode of actions of this combination, including interference with ribosome biogenesis and inhibition of pro-survival kinase PRAS40. Furthermore, by assessing global protein levels changes, FADS2, a key enzyme regulating fatty acids synthesis, was found down-regulated after proteasome inhibition. Interestingly, SC26196, an inhibitor of FADS2, synergized with carfilzomib. Finally, to identify further combinational options, we performed high-throughput drug screening and uncovered novel drug interactions with carfilzomib. For instance, cyclosporin A, a known immunosuppressive agent, enhanced carfilzomib's efficacy in vitro and in vivo. Altogether, these results demonstrate that carfilzomib and its combinations could be repurposed for LPS clinical management.
Collapse
Affiliation(s)
- Maya Jeitany
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| | - Aishvaryaa Prabhu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Pushkar Dakle
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Elina Pathak
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Vikas Madan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Deepika Kanojia
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Vineeth Mukundan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yan Yi Jiang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | | | - Wai Leong Tam
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Cedars-Sinai Medical Center, Division of Hematology/Oncology, UCLA School of Medicine, Los Angeles, CA, USA
- Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), National University Hospital, Singapore, Singapore
| |
Collapse
|
25
|
Moosavi F, Giovannetti E, Peters GJ, Firuzi O. Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol 2021; 160:103234. [PMID: 33497758 DOI: 10.1016/j.critrevonc.2021.103234] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
MET receptor has emerged as a druggable target across several human cancers. Agents targeting MET and its ligand hepatocyte growth factor (HGF) including small molecules such as crizotinib, tivantinib and cabozantinib or antibodies including rilotumumab and onartuzumab have proven their values in different tumors. Recently, capmatinib was approved for treatment of metastatic lung cancer with MET exon 14 skipping. In this review, we critically examine the current evidence on how HGF/MET combination therapies may take advantage of synergistic effects, overcome primary or acquired drug resistance, target tumor microenvironment, modulate drug metabolism or tackle pharmacokinetic issues. Preclinical and clinical studies on the combination of HGF/MET-targeted agents with conventional chemotherapeutics or molecularly targeted treatments (including EGFR, VEGFR, HER2, RAF/MEK, and PI3K/Akt targeting agents) and also the value of biomarkers are examined. Our deeper understanding of molecular mechanisms underlying successful pharmacological combinations is crucial to find the best personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
26
|
Vatseba TS, Sokolova LK, Pushkarev VM. Activation of the PI3K/AKT/MTOR/P70S6K1 signaling cascade in peripheral blood mononuclear cells in patients with type 2 diabetes. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.06.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
27
|
Merecz-Sadowska A, Sitarek P, Śliwiński T, Zajdel R. Anti-Inflammatory Activity of Extracts and Pure Compounds Derived from Plants via Modulation of Signaling Pathways, Especially PI3K/AKT in Macrophages. Int J Mol Sci 2020; 21:ijms21249605. [PMID: 33339446 PMCID: PMC7766727 DOI: 10.3390/ijms21249605] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
The plant kingdom is a source of important therapeutic agents. Therefore, in this review, we focus on natural compounds that exhibit efficient anti-inflammatory activity via modulation signaling transduction pathways in macrophage cells. Both extracts and pure chemicals from different species and parts of plants such as leaves, roots, flowers, barks, rhizomes, and seeds rich in secondary metabolites from various groups such as terpenes or polyphenols were included. Selected extracts and phytochemicals control macrophages biology via modulation signaling molecules including NF-κB, MAPKs, AP-1, STAT1, STAT6, IRF-4, IRF-5, PPARγ, KLF4 and especially PI3K/AKT. Macrophages are important immune effector cells that take part in antigen presentation, phagocytosis, and immunomodulation. The M1 and M2 phenotypes are related to the production of pro- and anti-inflammatory agents, respectively. The successful resolution of inflammation mediated by M2, or failed resolution mediated by M1, may lead to tissue repair or chronic inflammation. Chronic inflammation is strictly related to several disorders. Thus, compounds of plant origin targeting inflammatory response may constitute promising therapeutic strategies.
Collapse
Affiliation(s)
- Anna Merecz-Sadowska
- Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland
- Correspondence: (A.M.-S.); (T.Ś.)
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, 90-151 Lodz, Poland;
| | - Tomasz Śliwiński
- Laboratory of Medical Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
- Correspondence: (A.M.-S.); (T.Ś.)
| | - Radosław Zajdel
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| |
Collapse
|
28
|
Characterization of BRCA1-deficient premalignant tissues and cancers identifies Plekha5 as a tumor metastasis suppressor. Nat Commun 2020; 11:4875. [PMID: 32978388 PMCID: PMC7519681 DOI: 10.1038/s41467-020-18637-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 09/03/2020] [Indexed: 12/20/2022] Open
Abstract
Single-cell whole-exome sequencing (scWES) is a powerful approach for deciphering intratumor heterogeneity and identifying cancer drivers. So far, however, simultaneous analysis of single nucleotide variants (SNVs) and copy number variations (CNVs) of a single cell has been challenging. By analyzing SNVs and CNVs simultaneously in bulk and single cells of premalignant tissues and tumors from mouse and human BRCA1-associated breast cancers, we discover an evolution process through which the tumors initiate from cells with SNVs affecting driver genes in the premalignant stage and malignantly progress later via CNVs acquired in chromosome regions with cancer driver genes. These events occur randomly and hit many putative cancer drivers besides p53 to generate unique genetic and pathological features for each tumor. Upon this, we finally identify a tumor metastasis suppressor Plekha5, whose deficiency promotes cancer metastasis to the liver and/or lung.
Collapse
|
29
|
Sainani SR, Pansare PA, Rode K, Bhalchim V, Doke R, Desai S. Emendation of autophagic dysfuction in neurological disorders: a potential therapeutic target. Int J Neurosci 2020; 132:466-482. [PMID: 32924706 DOI: 10.1080/00207454.2020.1822356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Neurological disorders have been continuously contributing to the global disease burden and affect millions of people worldwide. Researchers strive hard to extract out the ultimate cure and serve for the betterment of the society, and yet the treatments available provide only symptomatic relief. Aging and abnormal mutations seem to be the major culprits responsible for neurotoxicity and neuronal death. One of the major causes of these neurological disorders that has been paid utmost attention recently, is Autophagic Dysfunction. AIM The aim of the study was to understand the autophagic process, its impairment in neurological disorders and targeting the impairments as a therapeutic option for the said disorders. METHODS For the purpose of review, we carried out an extensive literature study to excerpt the series of steps involved in autophagy and to understand the mechanism of autophagic impairment occurring in a range of neurodegenerative and neuropsychiatric disorders like Parkinson, Alzheimer, Depression, Schizophrenia, Autism etc. The review also involved the exploration of certain molecules that can help in triggering the compromised autophagic members. RESULTS We found that, a number of genes, proteins, receptors and transcription factors interplay to bring about autophagy and plethora of neurological disorders are associated with the diminished expression of one or more autophagic member leading to inhibition of autophagy. CONCLUSION Autophagy is a significant process for the removal of misfolded, abnormal, damaged protein aggregates and nonfunctional cell organelles in order to suppress neurodegeneration. Therefore, triggering autophagy could serve as an important therapeutic target to treat neurological disorders.
Collapse
Affiliation(s)
- Shivani R Sainani
- Department of Pharmacology, Dr D Y Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | - Prajakta A Pansare
- Department of Pharmacology, Dr D Y Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | - Ketki Rode
- Department of Pharmacology, Dr D Y Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | - Vrushali Bhalchim
- Department of Pharmacology, Dr D Y Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | - Rohit Doke
- Department of Pharmacology, Dr D Y Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| | - Shivani Desai
- Department of Pharmacology, Dr D Y Patil Institute of Pharmaceutical Sciences and Research, Pune, India
| |
Collapse
|
30
|
Zhou J, Zhu Y, Liu Y, Niu P, Chen H, Deng J, Shi D. High PRAS40 mRNA expression and its role in prognosis of clear cell renal cell carcinoma. Transl Androl Urol 2020; 9:1650-1660. [PMID: 32944526 PMCID: PMC7475688 DOI: 10.21037/tau-20-741] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is one of the most common type of kidney malignancy. The proline-rich Akt substrate of 40 kDa (PRAS40) plays an important role in tumor growth. The present study aimed to analysis the prognostic value of PRAS40 mRNA expression in ccRCC. Methods We analyzed the PRAS40 mRNA expression using the data from TCGA-KIRC cohort. A receiver operating characteristic (ROC) curve was performed to assessed the diagnostic value of PRAS40 mRNA expression in ccRCC. Chi-square test was used to analyzed the correlation between clinical characteristics and PRAS40 mRNA expression. Kaplan-Meier analysis and Cox analysis were performed to determine the prognostic value of PRAS40 mRNA expression in ccRCC. Gene set enrichment analysis (GSEA) was conducted using TCGA database. Results Our results revealed that PRAS40 mRNA expression was higher in ccRCC tissues than in normal tissues. PRAS40 presented a moderate diagnostic value in ccRCC. High PRAS40 mRNA expression was correlated with histological grade, clinical stage, T classification, distant metastasis and vital status of ccRCC. High PRAS40 mRNA expression was associated with poor overall survival. Furthermore, Multivariate analysis revealed that PRAS40 was an independent risk factor for ccRCC patients. Myc targets, DNA repair, oxidative phosphorylation, glycolysis, adipogenesis, p53 pathway, reactive oxygen species pathway, myogenesis were differentially enriched in the phenotype that positively correlated with PRAS40. Conclusions In conclusion, our results suggest that PRAS40 was a promising diagnostic and prognostic biomarker for ccRCC.
Collapse
Affiliation(s)
- Jintuo Zhou
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yanting Zhu
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ying Liu
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Peiguang Niu
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Huajiao Chen
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jie Deng
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Daohua Shi
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
31
|
Luszczak S, Simpson BS, Stopka-Farooqui U, Sathyadevan VK, Echeverria LMC, Kumar C, Costa H, Haider A, Freeman A, Jameson C, Ratynska M, Ben-Salha I, Sridhar A, Shaw G, Kelly JD, Pye H, Gately KA, Whitaker HC, Heavey S. Co-targeting PIM and PI3K/mTOR using multikinase inhibitor AUM302 and a combination of AZD-1208 and BEZ235 in prostate cancer. Sci Rep 2020; 10:14380. [PMID: 32873828 PMCID: PMC7463239 DOI: 10.1038/s41598-020-71263-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
PIM and PI3K/mTOR pathways are often dysregulated in prostate cancer, and may lead to decreased survival, increased metastasis and invasion. The pathways are heavily interconnected and act on a variety of common effectors that can lead to the development of resistance to drug inhibitors. Most current treatments exhibit issues with toxicity and resistance. We investigated the novel multikinase PIM/PI3K/mTOR inhibitor, AUM302, versus a combination of the PIM inhibitor, AZD-1208, and the PI3K/mTOR inhibitor BEZ235 (Dactolisib) to determine their impact on mRNA and phosphoprotein expression, as well as their functional efficacy. We have determined that around 20% of prostate cancer patients overexpress the direct targets of these drugs, and this cohort are more likely to have a high Gleason grade tumour (≥ Gleason 8). A co-targeted inhibition approach offered broader inhibition of genes and phosphoproteins in the PI3K/mTOR pathway, when compared to single kinase inhibition. The preclinical inhibitor AUM302, used at a lower dose, elicited a comparable or superior functional outcome compared with combined AZD-1208 + BEZ235, which have been investigated in clinical trials, and could help to reduce treatment toxicity in future trials. We believe that a co-targeting approach is a viable therapeutic strategy that should be developed further in pre-clinical studies.
Collapse
Affiliation(s)
- Sabina Luszczak
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Benjamin S Simpson
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | | | | | | | - Christopher Kumar
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Helena Costa
- Research Department of Pathology, University College London, London, UK
| | - Aiman Haider
- Research Department of Pathology, University College London, London, UK
| | - Alex Freeman
- Research Department of Pathology, University College London, London, UK
| | - Charles Jameson
- Research Department of Pathology, University College London, London, UK
| | - Marzena Ratynska
- Research Department of Pathology, University College London, London, UK
| | - Imen Ben-Salha
- Research Department of Pathology, University College London, London, UK
| | - Ashwin Sridhar
- Department of Uro-Oncology, UCLH NHS Foundation Trust, London, UK
| | - Greg Shaw
- Department of Uro-Oncology, UCLH NHS Foundation Trust, London, UK
| | - John D Kelly
- Department of Uro-Oncology, UCLH NHS Foundation Trust, London, UK
| | - Hayley Pye
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Kathy A Gately
- Trinity Translational Medicine Institute, St. James's Hospital Dublin, Dublin 8, Ireland
| | - Hayley C Whitaker
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK.
| |
Collapse
|
32
|
Di Desidero T, Orlandi P, Gentile D, Banchi M, Alì G, Kusmic C, Armanetti P, Cayme GJ, Menichetti L, Fontanini G, Francia G, Bocci G. Pharmacological effects of vinorelbine in combination with lenvatinib in anaplastic thyroid cancer. Pharmacol Res 2020; 158:104920. [PMID: 32461187 PMCID: PMC8011355 DOI: 10.1016/j.phrs.2020.104920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/29/2020] [Accepted: 05/10/2020] [Indexed: 12/27/2022]
Abstract
Anaplastic thyroid cancer (ATC) is a rare neoplasia with a poor prognosis. Proliferation and apoptosis assays were performed on ATC cell lines (8305C, 8505C) exposed to vinorelbine, lenvatinib, as well as to concomitant combinations. ABCB1, ABCG2 and CSF-1 mRNA expression was evaluated by real time PCR. The relative levels of pospho Akt were investigated as part of a human phospho-kinase array analysis, and CSF-1 and VEGFR-2 protein levels were measured by ELISA. The intracellular concentration of lenvatinib in ATC cells was measured by combined reversed-phase liquid chromatography-tandem mass spectrometry. An ATC subcutaneous xenograft tumor model in nude mice was treated with vinorelbine, lenvatinib, or vinorelbine plus lenvatinib. After treatment with vinorelbine, lenvatinib, a significant antiproliferative effect in ATC cell lines was observed. The concomitant treatment of vinorelbine and lenvatinib revealed synergism for all the fractions of affected cells. A decrease in ABCB1 expression was reported in both ATC cell lines treated with the lenvatinib plus vinorelbine combination, as was an increase in the intracellular concentration of lenvatinib. The combination caused a decrease in Akt, GSK3α/β, PRAS40 and Src phosphorylation, and in both CSF-1 mRNA and protein levels. In the subcutaneous tumor model, the combination reduced the tumor volume during the treatment period. Our results establish the synergistic ATC antitumor activity of a vinorelbine and lenvatinib combination.
Collapse
Key Words
- Anaplastic thyroid cancer
- Lenvatinib
- Lenvatinib - IUPAC name: 4-[3-chloro-4-(cyclopropylcarbamoylamino)phenoxy]-7-methoxyquinoline-6-carboxamide - PubChem CID: 9823820
- Sorafenib
- Synergism
- Vinorelbine
- Vinorelbine - IUPAC namemethyl (1R, 9R, 10S, 11R, 12R, 19R)-11-acetyloxy-12-ethyl-4-[(12S, 14R,)-16-ethyl-12-methoxycarbonyl-1, 10-diazatetracyclo[12.3.1.0, (3), (11).0, (4), (9),]octadeca-3(11), 4, 6, 8, 15-pentaen-12-yl]-10-hydroxy-5-methoxy-8-methyl-8, 16-diazapentacyclo[10.6.1.0, (1), (9).0, (2), (7).0, (16), (19),]nonadeca-2, 4, 6, 13-tetraene-10-carboxylate- PubChem CID 5311497
Collapse
Affiliation(s)
- Teresa Di Desidero
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Paola Orlandi
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Daniela Gentile
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Marta Banchi
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| | - Greta Alì
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università di Pisa, Pisa, Italy
| | - Claudia Kusmic
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Paolo Armanetti
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Ginelle J Cayme
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Luca Menichetti
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Gabriella Fontanini
- Dipartimento di Patologia Chirurgica, Medica, Molecolare e Dell'Area Critica, Università di Pisa, Pisa, Italy
| | - Giulio Francia
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, USA
| | - Guido Bocci
- Dipartimento Di Medicina Clinica e Sperimentale, Università Di Pisa, Pisa, Italy
| |
Collapse
|
33
|
Lee H, Chin H, Kim H, Jung H, Lee D. STAT3-mediated MLST8 gene expression regulates cap-dependent translation in cancer cells. Mol Oncol 2020; 14:1850-1867. [PMID: 32495998 PMCID: PMC7400782 DOI: 10.1002/1878-0261.12735] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 05/15/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) regulates cell growth, cell survival, angiogenesis, metastasis of cancer cells, and cancer immune evasion by regulating gene expression as a transcription factor. However, the effect of STAT3 on translation is almost unknown. We demonstrated that STAT3 acts as a trans‐acting factor for MLST8 gene expression and the protein level of mLST8, a core component of mechanistic target of rapamycin complex 1 and 2 (mTORC1/2), positively regulates the mTORC1/2 downstream pathways. Suppression of STAT3 by siRNA attenuated 4E‐BP1 phosphorylation, cap‐dependent translation, and cell proliferation in a variety of cancer cells. In HCT116 cells, STAT3 knockdown‐induced decreases in 4E‐BP1 and AKT phosphorylation levels were further attenuated by MLST8 knockdown or recovered by mLST8 overexpression. STAT3 knockdown‐induced G2/M phase arrest was partially restored by co‐knockdown of 4EBP1, and the attenuation of cell proliferation was enhanced by the expression of an mTORC1‐mediated phosphorylation‐defective mutant of 4E‐BP1. ChIP and promoter mapping using a luciferase reporter assay showed that the −951 to −894 bp of MLST8 promoter seems to include STAT3‐binding site. Overall, these results suggest that STAT3‐driven MLST8 gene expression regulates cap‐dependent translation through 4E‐BP1 phosphorylation in cancer cells.
Collapse
Affiliation(s)
- Hyunji Lee
- Department of Life Science, Ewha Womans University Ewhayeodae-gil 52, Seodaemun-gu, Seoul, South Korea
| | - Hyunjung Chin
- Department of Life Science, Ewha Womans University Ewhayeodae-gil 52, Seodaemun-gu, Seoul, South Korea
| | - Hyeyoung Kim
- Department of Anatomy, and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Hosung Jung
- Department of Anatomy, and Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Daekee Lee
- Department of Life Science, Ewha Womans University Ewhayeodae-gil 52, Seodaemun-gu, Seoul, South Korea
| |
Collapse
|
34
|
Wang C, Chen Y, Chen K, Zhang L. Long Noncoding RNA LINC01134 Promotes Hepatocellular Carcinoma Metastasis via Activating AKT1S1 and NF-κB Signaling. Front Cell Dev Biol 2020; 8:429. [PMID: 32656205 PMCID: PMC7325970 DOI: 10.3389/fcell.2020.00429] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies with poor outcomes. The main causes of HCC-related deaths are recurrence and metastasis. Long noncoding RNAs (lncRNAs) are recently identified as critical regulators in cancers. However, the lncRNAs involved in HCC recurrence and metastasis are poorly understood. In this study, via analyzing The Cancer Genome Atlas Liver Hepatocellular Carcinoma dataset, we identified a novel lncRNA LINC01134, which is highly expressed in HCC tissues and correlated with microvascular invasion, macrovascular invasion, recurrence, and poor overall survival of HCC patients. Functional experiments revealed that ectopic expression of LINC01134 promotes HCC cell migration and invasion in vitro and HCC liver metastasis and lung metastasis in vivo. Knockdown of LINC01134 represses HCC cell migration and invasion in vitro and HCC liver metastasis and lung metastasis in vivo. Mechanistically, we found that LINC01134 directly binds the promoter of AKT1S1 and activates AKT1S1 expression. Via activating AKT1S1, LINC01134 further activates NF-κB signaling. The expression of LINC01134 is significantly positively correlated with that of AKT1S1 in HCC tissues. In line with LINC01134, AKT1S1 is also highly expressed in HCC tissues and correlated with poor survival of HCC patients. Functional rescue experiments showed that repressing AKT1S1 or NF-κB signaling abrogates the roles of LINC01134 in HCC. Taken together, these findings recognized LINC01134 as a novel oncogenic lncRNA, which indicates vascular invasion, recurrence, and poor overall survival of HCC patients. LINC01134 promotes HCC metastasis via activating AKT1S1 expression and subsequently activating NF-κB signaling. This study suggested LINC01134 as a potential prognostic biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Chao Wang
- Department of General Surgery, Clinical Research Center of Geriatric Diseases in Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kunlun Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Zhang
- Hepatic Surgery Center, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Effect of Electroacupuncture Treatment at Dazhui (GV14) and Mingmen (GV4) Modulates the PI3K/AKT/mTOR Signaling Pathway in Rats after Spinal Cord Injury. Neural Plast 2020; 2020:5474608. [PMID: 32399023 PMCID: PMC7201674 DOI: 10.1155/2020/5474608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/08/2019] [Accepted: 12/26/2019] [Indexed: 12/18/2022] Open
Abstract
Electroacupuncture (EA) is widely recognized as clinical treatment of spinal cord injury (SCI). The purpose of this study is to elucidate whether and how the PI3K/AKT/mTOR signaling pathway plays any role in EA treating SCI. Rats were randomly divided into four equal groups: Control Group, Sham-operation Group, Model Group, and EA Group, then further randomly divided into the following subgroups: 1-day (n = 12), 1-day rapamycin (n = 6), 14-day (n = 18), and 28-day (n = 18). A rat model of SCI was established by a modified Allen's weight-drop method. In the EA Group, rats were stimulated on Dazhui (GV14) and Mingmen (GV4) for 20 min by sterilized stainless steel needles. In the EA Group, the Basso, Beattie, and Bresnahan locomotor rating scale showed obvious improved locomotor function, and hematoxylin-eosin staining and magnetic resonance imaging showed that the histological morphology change of injured spinal cord tissue was obviously alleviated. Also, blocking spinal mTOR by injection of rapamycin showed that mTOR existed in the injured spinal cord, and EA could significantly activate mTOR in SCI rats. And immunohistochemistry and western blot analysis on the PI3K/AKT/mTOR signaling pathway showed that levels of PI3K, AKT, mTOR, and p70S6K in the injured spinal cord tissue were greatly increased in the EA Group, while the levels of PTEN and caspase 3 were decreased. The present study suggests that EA could affect cell growth, apoptosis, and autophagy through the PI3K/AKT/mTOR signaling pathway.
Collapse
|
36
|
Kurman Y, Kiliccioglu I, Dikmen AU, Esendagli G, Bilen CY, Sozen S, Konac E. Cucurbitacin B and cisplatin induce the cell death pathways in MB49 mouse bladder cancer model. Exp Biol Med (Maywood) 2020; 245:805-814. [PMID: 32252554 DOI: 10.1177/1535370220917367] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT Alternative agents that will increase the effectiveness of cisplatin, which are widely used in the advanced stage and metastatic bladder cancer, are being investigated. In previous studies, Cucurbitacin B (CuB), which is a natural compound from the Cucurbitaceae family has been shown to inhibit the proliferation of tumor cells and create synergistic effects with cisplatin. In this study, we investigated the synergistic effect of CuB with cisplatin for the first time in bladder cancer in vitro and in vivo models. Our findings showed that CuB treatment with cisplatin reduced cell proliferation, and reduced tumor development through activating apoptosis and autophagy via PI3K/AKT/mTOR signaling pathway. Our results showed that CuB may be a new agent that can support conventional treatment in bladder cancer. Our study is important in terms of enlightening new pathways and developing new treatment methods in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Yener Kurman
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ilker Kiliccioglu
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey.,Department of Medical Biology, Faculty of Medicine, Duzce University, Duzce 81620, Turkey
| | - Asiye U Dikmen
- Department of Public Health, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Guldal Esendagli
- Department of Pathology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Cenk Y Bilen
- Department of Urology, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Sinan Sozen
- Department of Urology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ece Konac
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| |
Collapse
|
37
|
Secondary Metabolites from the Culture of the Marine-derived Fungus Paradendryphiella salina PC 362H and Evaluation of the Anticancer Activity of Its Metabolite Hyalodendrin. Mar Drugs 2020; 18:md18040191. [PMID: 32260204 PMCID: PMC7230232 DOI: 10.3390/md18040191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/24/2022] Open
Abstract
High-throughput screening assays have been designed to identify compounds capable of inhibiting phenotypes involved in cancer aggressiveness. However, most studies used commercially available chemical libraries. This prompted us to explore natural products isolated from marine-derived fungi as a new source of molecules. In this study, we established a chemical library from 99 strains corresponding to 45 molecular operational taxonomic units and evaluated their anticancer activity against the MCF7 epithelial cancer cell line and its invasive stem cell-like MCF7-Sh-WISP2 counterpart. We identified the marine fungal Paradendryphiella salina PC 362H strain, isolated from the brown alga Pelvetia caniculata (PC), as one of the most promising fungi which produce active compounds. Further chemical and biological characterizations of the culture of the Paradendryphiella salina PC 362H strain identified (-)-hyalodendrin as the active secondary metabolite responsible for the cytotoxic activity of the crude extract. The antitumor activity of (-)-hyalodendrin was not only limited to the MCF7 cell lines, but also prominent on cancer cells with invasive phenotypes including colorectal cancer cells resistant to chemotherapy. Further investigations showed that treatment of MCF7-Sh-WISP2 cells with (-)-hyalodendrin induced changes in the phosphorylation status of p53 and altered expression of HSP60, HSP70 and PRAS40 proteins. Altogether, our study reveals that this uninvestigated marine fungal crude extract possesses a strong therapeutic potential against tumor cells with aggressive phenotypes and confirms that members of the epidithiodioxopiperazines are interesting fungal toxins with anticancer activities.
Collapse
|
38
|
Raglan O, Assi N, Nautiyal J, Lu H, Gabra H, Gunter MJ, Kyrgiou M. Proteomic analysis of malignant and benign endometrium according to obesity and insulin-resistance status using Reverse Phase Protein Array. Transl Res 2020; 218:57-72. [PMID: 31954096 DOI: 10.1016/j.trsl.2019.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/21/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022]
Abstract
Obesity and hyperinsulinemia are known risk factors for endometrial cancer, yet the biological pathways underlying this relationship are incompletely understood. This study investigated protein expression in endometrial cancer and benign tissue and its correlation with obesity and insulin resistance. One hundred and seven women undergoing hysterectomy for endometrial cancer or benign conditions provided a fasting blood sample and endometrial tissue. We performed proteomic expression according to body mass index, insulin resistance, and serum marker levels. We used linear regression and independent t test for statistical analysis. Proteomic data from 560 endometrial cancer cases from The Cancer Genome Atlas (TCGA) databank were used to assess reproducibility of results. One hundred and twenty seven proteins were significantly differentially expressed between 66 cancer and 26 benign patients. Protein expression involved in cell cycle progression, impacting cytoskeletal dynamics (PAK1) and cell survival (Rab 25), were most significantly altered. Obese women with cancer had increased PRAS40_pT246; a downstream marker of increased PI3K-AKT signaling. Obese women without cancer had increased mitogenic and antiapoptotic signaling by way of upregulation of Mcl-1, DUSP4, and Insulin Receptor-b. This exploratory study identified a number of candidate proteins specific to endometrioid endometrial cancer and benign endometrial tissues. Obesity and insulin resistance in women with benign endometrium leads to specific upregulation of proteins involved in insulin and driver oncogenic signaling pathways such as the PI3K-AKT-mTOR and growth factor signaling pathways which are mitogenic and also disruptive to metabolism.
Collapse
Affiliation(s)
- Olivia Raglan
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, UK; Queen Charlotte's and Chelsea - Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Nada Assi
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), Lyon, France
| | - Jaya Nautiyal
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, UK
| | - Haonan Lu
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, UK
| | - Hani Gabra
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, UK; Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC), Lyon, France
| | - Maria Kyrgiou
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, UK; Queen Charlotte's and Chelsea - Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK.
| |
Collapse
|
39
|
Protein Kinase B Inactivation Is Associated with Magnolol-Enhanced Therapeutic Efficacy of Sorafenib in Hepatocellular Carcinoma In Vitro and In Vivo. Cancers (Basel) 2019; 12:cancers12010087. [PMID: 31905887 PMCID: PMC7017147 DOI: 10.3390/cancers12010087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 12/03/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022] Open
Abstract
Although sorafenib, an oral multikinase inhibitor, was approved as a treatment drug of advance hepatocellular carcinoma (HCC), treatment efficacy still requires improvement. Searching for the adjuvant reagent for enhancing sorafenib efficacy remains as a critical issue. Sorafenib has been proved to suppress extracellular signal-regulated kinases (ERK) in HCC; however, protein kinase B (AKT) was not affected by it. Targeting AKT in combination with sorafenib could be an important breakthrough point of HCC treatment. Many herbal compounds and composite formulas have been shown to enhance anti-HCC activity of sorafenib. Magnolol is a bioactive compound extracted from the bark of the Magnolia officinalis and has been shown to induce apoptosis and inhibit cell invasion in HCC in vitro. However, whether magnolol sensitizes HCC to sorafenib is ambiguous. In this study, we indicated that magnolol significantly enhanced sorafenib-diminished tumor cell growth, expression of anti-apoptotic proteins, and migration/invasion ability compared to sorafenib alone. Magnolol significantly boosted sorafenib-induced extrinsic/intrinsic dependent apoptosis pathways in HCC. Notably sorafenib could not reduce protein level of AKT (Ser473), but expression of AKT (Ser473) was significantly decreased by magnolol or magnolol combined with sorafenib. LY294002 as specific AKT inhibitor was used to confirm that AKT inactivation may promote anticancer effect of sorafenib. Taken together, AKT inhibition is associated with magnolol-enhanced the therapeutic effect of sorafenib in HCC. We suggested magnolol as the potential adjuvant which may enhance therapeutic benefits of sorafenib in patients with HCC.
Collapse
|
40
|
Xu Q, Wu N, Li X, Guo C, Li C, Jiang B, Wang H, Shi D. Inhibition of PTP1B blocks pancreatic cancer progression by targeting the PKM2/AMPK/mTOC1 pathway. Cell Death Dis 2019; 10:874. [PMID: 31745071 PMCID: PMC6864061 DOI: 10.1038/s41419-019-2073-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/27/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is a highly malignant cancer and lacks effective therapeutic targets. Protein-tyrosine phosphatase 1B (PTP1B), a validated therapeutic target for diabetes and obesity, also plays a critical positive or negative role in tumorigenesis. However, the role of PTP1B in pancreatic cancer remains elusive. Here, we initially demonstrated that PTP1B was highly expressed in pancreatic tumors, and was positively correlated with distant metastasis and tumor staging, and indicated poor survival. Then, inhibition of PTP1B either by shRNA or by a specific small-molecule inhibitor significantly suppressed pancreatic cancer cell growth, migration and colony formation with cell cycle arrest in vitro and inhibited pancreatic cancer progression in vivo. Mechanism studies revealed that PTP1B targeted the PKM2/AMPK/mTOC1 signaling pathway to regulate cell growth. PTP1B inhibition directly increased PKM2 Tyr-105 phosphorylation to further result in significant activation of AMPK, which decreased mTOC1 activity and led to inhibition of p70S6K. Meanwhile, the decreased phosphorylation of PRAS40 caused by decreased PKM2 activity also helped to inhibit mTOC1. Collectively, these findings support the notion of PTP1B as an oncogene and a promising therapeutic target for PDAC.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/antagonists & inhibitors
- AMP-Activated Protein Kinases/metabolism
- Animals
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Disease Progression
- Female
- Humans
- Male
- Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors
- Mechanistic Target of Rapamycin Complex 1/metabolism
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Random Allocation
- Signal Transduction/drug effects
- Small Molecule Libraries/pharmacology
- Thyroid Hormones/metabolism
- Xenograft Model Antitumor Assays
- Thyroid Hormone-Binding Proteins
Collapse
Affiliation(s)
- Qi Xu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Ning Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiangqian Li
- State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, Shandong, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Chuanlong Guo
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Chao Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Bo Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
- The University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
41
|
Vatseba TS. Influence of pathogenetic factors of type 2 diabetes on activation of PI3K/AkT/mTOR pathway and on the development of endometrial and breast cancer. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Type 2 diabetes (T2D), which is an epidemic of the 20th century, increases mortality, caused not only by cardiovascular diseases but also cancer. Pathogenetic factors of T2D lead to dysfunction of intracellular regulatory systems, particularly of PI3K/Akt/mTOR signaling pathway, which is involved in development of breast and endometrial cancer. During the study, the activation of this pathway by cancer and T2D was examined by determining the content of phosphorylated PRAS40 and p70S6K1. We studied the link between these kinases and pathogenetic factors of T2D. 65 women were examined. Patients were divided into four groups: І – healthy, ІІ – women with T2D, ІІІ – women with cancer without diabetes, IV – women with cancer and T2D. Level of HbA1c was determined by the ion-exchange chromatography. Levels of insulin, IGF-1, phospho-PRAS40, phospho-p70S6K1 were determined in PBMCs by immune-enzymatic methods. According to research results, significant hyperinsulinemia was detected in both groups with T2D. The highest level of insulin was in group II. A significantly higher level of IGF-1 was found only in patients with cancer of group III. The content of phospho-PRAS40 and phospho-p70S6K1 was higher in women with T2D of group II and in women with cancer of group III. Patients in group IV with combination of cancer and T2D had a lower level of phospho-PRAS40 in comparison to other groups. Phospho-PRAS40 in group II correlates with insulin, IGF-1 and HbA1c; in groups III and IV only with BMI. Phospho-p70S6K1 correlates with IGF-1 and with HbA1c in group II. Pathogenetic factors of T2D activate the signal path PI3K/Akt/mTOR, which is involved in the regulation of oncogenesis and metabolism. Phosphorylation of PRAS40 and p70S6K1 reflects the activation of P13K/Akt/mTOR pathway in women with T2D. Increased levels the phospho-PRAS40 and phospho-p70S6K1 can be used as early markers of oncogenesis in women with T2D.
Collapse
|
42
|
Boscolo E, Pastura P, Glaser K, Goines J, Hammill AM, Adams DM, Dickie P, Dickie BH, Le Cras TD. Signaling pathways and inhibitors of cells from patients with kaposiform lymphangiomatosis. Pediatr Blood Cancer 2019; 66:e27790. [PMID: 31045327 PMCID: PMC6588438 DOI: 10.1002/pbc.27790] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/20/2019] [Accepted: 04/17/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Kaposiform lymphangiomatosis (KLA) is a rare lymphatic anomaly with significant morbidity and mortality. KLA is characterized by diffuse multifocal lesions comprised of focal areas of "kaposiform" spindled cells accompanying malformed lymphatic channels. The goal of this study was to identify activated signaling pathways in cells isolated from three KLA patients for the purpose of testing new therapies. PROCEDURE Cells were obtained from the lungs of one patient isolated at autopsy and the spleen of two patients removed in surgery due to disease complications. A protein kinase array was performed on the KLA cell lysates and normal lymphatic endothelial cells. RESULTS Higher activation of key signaling pathways in the KLA cells, including PRAS40, AKT1/2/3, and ERK-1/2, was identified by protein kinase array and confirmed by Western blot analysis. This indicated a role for highly activated PI3K-AKT and MAPK-ERK-1/2 signaling pathways in KLA cells. Cell proliferation studies assessed PI3K inhibitors (LY294002; BYL719), AKT inhibitor ARQ092, mTOR inhibitor rapamycin, and MAPK inhibitor U0126. These studies demonstrated that PI3K-AKT-mTOR and MAPK signaling are important mediators of KLA cell proliferation. BYL719 and rapamycin were more effective at inhibiting KLA cell proliferation than U0126. CONCLUSIONS Our studies using cells from KLA patient lesions demonstrate that these cells are highly proliferative and the PI3K-AKT-mTOR and MAPK pathways are promising therapeutic targets. Development and clinical trials of PI3K, AKT, and MAPK inhibitors for cancer treatment and the data in this study lend support for early clinical trials assessing the efficacy of these inhibitors in KLA patients.
Collapse
Affiliation(s)
- Elisa Boscolo
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Patricia Pastura
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Division of Pulmonary Biology, Cincinnati Children’s Hospital, Cincinnati, OH, USA
| | - Kathryn Glaser
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jillian Goines
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Adrienne M. Hammill
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Hemangioma & Vascular Anomalies Center & Division of Hematology, Cincinnati Children’s Hospital, Cincinnati, OH, USA
| | - Denise M. Adams
- Vascular Anomalies Center, Boston, MA, USA,Boston Children’s Hospital, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Peter Dickie
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Belinda Hsi Dickie
- Boston Children’s Hospital, Boston, MA, USA,Colorectal and Pelvic Malformation Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Timothy D. Le Cras
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Division of Pulmonary Biology, Cincinnati Children’s Hospital, Cincinnati, OH, USA
| |
Collapse
|
43
|
Javid H, Soltani A, Mohammadi F, Hashemy SI. Emerging roles of microRNAs in regulating the mTOR signaling pathway during tumorigenesis. J Cell Biochem 2019; 120:10874-10883. [PMID: 30719752 DOI: 10.1002/jcb.28401] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/13/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
The mammalian target of rapamycin (mTOR) is a large Ser/Thr protein kinase that belongs to the phosphoinositide 3-kinase (PI3K) family and mediates various physiological and pathological processes, especially cell proliferation, protein synthesis, autophagy, and cancer development. The mTOR expression is transient and tightly regulated in normal cells, but it is overactivated in cancer cells. Recently, several studies have indicated that microRNAs (miRNAs) play a critical role in the regulation of mTOR and mTOR-associated processes, some acting as inhibitors and the others as activators. Although it is still in infancy, the strategy of combining both miRNAs and mTOR inhibitors might provide an approach to selectively sensitizing tumor cells to chemotherapy-induced DNA damage and subsequently attenuating the tumor cell growth and apoptosis.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Soltani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Mohammadi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
44
|
Lisi L, Ciotti GMP, Chiavari M, Pizzoferrato M, Mangiola A, Kalinin S, Feinstein DL, Navarra P. Phospho-mTOR expression in human glioblastoma microglia-macrophage cells. Neurochem Int 2019; 129:104485. [PMID: 31195027 DOI: 10.1016/j.neuint.2019.104485] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/01/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022]
Abstract
The glioblastoma (GBM) immune microenvironment is highly heterogeneous, and microglia may represent 30-70% of the entire tumor. However, the role of microglia and other specific immune populations is poorly characterized. Activation of mTOR signaling occurs in numerous human cancers and has roles in microglia-glioma cell interactions. We now show in human tumor specimens (42 patients), that 39% of tumor-associated microglial (TAM) cells express mTOR phosphorylated at Ser-2448; and similar mTOR activation is observed using a human microglia-glioma interaction paradigm. In addition, we confirm previous studies that microglia express urea and ARG1 (taken as M2 marker) in the presence of glioma cells, and this phenotype is down-regulated in the presence of a mTOR inhibitor. These results suggest that mTOR suppression in GBM patients might induce a reduction of the M2 phenotype expression in up to 40% of all TAMs. Since the M2 profile of microglial activation is believed to be associated with tumor progression, reductions in that phenotype may represent an additional anti-tumor mechanism of action of mTOR inhibitors, along with direct anti-proliferative activities.
Collapse
Affiliation(s)
- Lucia Lisi
- Institute of Farmacologia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, Rome, Italy.
| | | | - Marta Chiavari
- Institute of Farmacologia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, Rome, Italy
| | - Michela Pizzoferrato
- Institute of Farmacologia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, Rome, Italy
| | - Annunziato Mangiola
- Department of Neuroscience, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio Chieti-Pescara, via Colle dell'Ara 100, Chieti, Italy
| | - Sergey Kalinin
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL, USA; Department of Veterans Affairs, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pierluigi Navarra
- Institute of Farmacologia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, Rome, Italy; Fondazione Policlinico Universitario Agostino Gemelli, L.go F. Vito 1, Rome, Italy
| |
Collapse
|
45
|
Llerena S, García-Díaz N, Curiel-Olmo S, Agraz-Doblas A, García-Blanco A, Pisonero H, Varela M, Santibáñez M, Almaraz C, Cereceda L, Martínez N, Arias-Loste MT, Puente Á, Martín-Ramos L, de Lope CR, Castillo-Suescun F, Cagigas-Fernandez C, Isidro P, Lopez-López C, Lopez-Hoyos M, Llorca J, Agüero J, Crespo-Facorro B, Varela I, Piris MÁ, Crespo J, Vaqué JP. Applied diagnostics in liver cancer. Efficient combinations of sorafenib with targeted inhibitors blocking AKT/mTOR. Oncotarget 2018; 9:30869-30882. [PMID: 30112114 PMCID: PMC6089396 DOI: 10.18632/oncotarget.25766] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/22/2018] [Indexed: 12/31/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related deaths worldwide. There is increasing interest in developing specific markers to serve as predictors of response to sorafenib and to guide targeted therapy. Using a sequencing platform designed to study somatic mutations in a selection of 112 genes (HepatoExome), we aimed to characterize lesions from HCC patients and cell lines, and to use the data to study the biological and mechanistic effects of case-specific targeted therapies used alone or in combination with sorafenib. We characterized 331 HCC cases in silico and 32 paired samples obtained prospectively from primary tumors of HCC patients. Each case was analyzed in a time compatible with the requirements of the clinic (within 15 days). In 53% of the discovery cohort cases, we detected unique mutational signatures, with up to 34% of them carrying mutated genes with the potential to guide therapy. In a panel of HCC cell lines, each characterized by a specific mutational signature, sorafenib elicited heterogeneous mechanistic and biological responses, whereas targeted therapy provoked the robust inhibition of cell proliferation and DNA synthesis along with the blockage of AKT/mTOR signaling. The combination of sorafenib with targeted therapies exhibited synergistic anti-HCC biological activity concomitantly with highly effective inhibition of MAPK and AKT/mTOR signaling. Thus, somatic mutations may lead to identify case-specific mechanisms of disease in HCC lesions arising from multiple etiologies. Moreover, targeted therapies guided by molecular characterization, used alone or in combination with sorafenib, can effectively block important HCC disease mechanisms.
Collapse
Affiliation(s)
- Susana Llerena
- Gastroenterology and Hepatology Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain
| | - Nuria García-Díaz
- Translational Hematopathology Group, IDIVAL, Instituto de Investigación Marqués de Valdecilla, Santander, Spain.,Departamento de Biología Molecular, Universidad de Cantabria (UC-IBBTEC), Santander, Spain
| | - Soraya Curiel-Olmo
- Translational Hematopathology Group, IDIVAL, Instituto de Investigación Marqués de Valdecilla, Santander, Spain
| | - Antonio Agraz-Doblas
- Departamento de Biología Molecular, Universidad de Cantabria (UC-IBBTEC), Santander, Spain.,Josep Carreras Leukemia Research Institute and School of Medicine, University of Barcelona, Barcelona, Spain
| | - Agustín García-Blanco
- Gastroenterology and Hepatology Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain
| | - Helena Pisonero
- Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain.,Departamento de Biología Molecular, Universidad de Cantabria (UC-IBBTEC), Santander, Spain
| | - María Varela
- Digestive Service, Hepatology Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Carmen Almaraz
- Translational Hematopathology Group, IDIVAL, Instituto de Investigación Marqués de Valdecilla, Santander, Spain
| | - Laura Cereceda
- Translational Hematopathology Group, IDIVAL, Instituto de Investigación Marqués de Valdecilla, Santander, Spain
| | - Nerea Martínez
- Translational Hematopathology Group, IDIVAL, Instituto de Investigación Marqués de Valdecilla, Santander, Spain
| | - María Teresa Arias-Loste
- Gastroenterology and Hepatology Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain
| | - Ángela Puente
- Gastroenterology and Hepatology Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain
| | - Luis Martín-Ramos
- Gastroenterology and Hepatology Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain
| | - Carlos Rodríguez de Lope
- Gastroenterology and Hepatology Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain
| | - Federico Castillo-Suescun
- General and Digestive Tract Surgery Service, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Carmen Cagigas-Fernandez
- General and Digestive Tract Surgery Service, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Pablo Isidro
- Biobanco-Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Carlos Lopez-López
- Oncology Service, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Marcos Lopez-Hoyos
- Immunology Service, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Javier Llorca
- Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, Santander, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Jesús Agüero
- Microbiology Service, University Hospital Marques de Valdecilla-IDIVAL, Santander, Spain
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, Marqués de Valdecilla University Hospital-IDIVAL, Santander, Spain.,CIBERSAM, Centro de Investigación Biomédica en Red Salud Mental, Madrid, Spain
| | - Ignacio Varela
- Departamento de Biología Molecular, Universidad de Cantabria (UC-IBBTEC), Santander, Spain
| | | | - Javier Crespo
- Gastroenterology and Hepatology Unit, Hospital Universitario Marqués de Valdecilla, Santander, Spain.,Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain
| | - José Pedro Vaqué
- Infection, Immunity and Digestive Pathology Group, IDIVAL, Santander, Spain.,Departamento de Biología Molecular, Universidad de Cantabria (UC-IBBTEC), Santander, Spain
| |
Collapse
|
46
|
Neurofibromin level directs RAS pathway signaling and mediates sensitivity to targeted agents in malignant peripheral nerve sheath tumors. Oncotarget 2018; 9:22571-22585. [PMID: 29854299 PMCID: PMC5978249 DOI: 10.18632/oncotarget.25181] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a type of soft-tissue sarcoma strongly associated with dysfunction in neurofibromin; an inhibitor of the RAS pathway. We performed high-throughput screening of an array of FDA approved and promising agents in clinical development both alone and in combination at physiologically achievable concentrations against a panel of established MPNST cell line models. We found that drugs targeting a variety of factors in the RAS pathway can effectively lead to cell death in vitro with considerable drug combination synergy in regimens that target MEK or mTOR. We observed that the degree of relative sensitivity to chemotherapeutic agents was associated with the status of neurofibromin in these cell line models. Using a combination of agents that target MEK and mTORC1/2, we effectively silenced RAS/PI3K/MEK/mTOR signaling in vitro. Moreover, we employed RNAi against NF1 to establish that MPNST drug sensitivity is directly proportional to relative level of intracellular neurofibromin. Thus, two-drug combinations that target MEK and mTORC1/2 are most effective in halting the RAS signaling cascade, and the relative success of this and related small molecule interventions in MPNSTs may be predicated upon the molecular status of neurofibromin.
Collapse
|