1
|
Song F, Zhang Z, Liu W, Xu T, Hu X, Wang Q, Zhang W, Ge L, Zhang C, Hu Q, Qin H, Zhang S, Ren X, Fan W, Zhang Y, Huang P. Peptide Transporter 1-Mediated Dipeptide Transport Promotes Hepatocellular Carcinoma Metastasis by Activating MAP4K4/G3BP2 Signaling Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306671. [PMID: 38639383 PMCID: PMC11200092 DOI: 10.1002/advs.202306671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/18/2024] [Indexed: 04/20/2024]
Abstract
Cancer metastasis is the leading cause of mortality in patients with hepatocellular carcinoma (HCC). To meet the rapid malignant growth and transformation, tumor cells dramatically increase the consumption of nutrients, such as amino acids. Peptide transporter 1 (PEPT1), a key transporter for small peptides, has been found to be an effective and energy-saving intracellular source of amino acids that are required for the growth of tumor cells. Here, the role of PEPT1 in HCC metastasis and its underlying mechanisms is explored. PEPT1 is upregulated in HCC cells and tissues, and high PEPT1 expression is associated with poor prognosis in patients with HCC. PEPT1 overexpression dramatically promoted HCC cell migration, invasion, and lung metastasis, whereas its knockdown abolished these effects both in vitro and in vivo. Mechanistic analysis revealed that high PEPT1 expression increased cellular dipeptides in HCC cells that are responsible for activating the MAP4K4/G3BP2 signaling pathway, ultimately facilitating the phosphorylation of G3BP2 at Thr227 and enhancing HCC metastasis. Taken together, these findings suggest that PEPT1 acts as an oncogene in promoting HCC metastasis through dipeptide-induced MAP4K4/G3BP2 signaling and that the PEPT1/MAP4K4/G3BP2 axis can serve as a promising therapeutic target for metastatic HCC.
Collapse
Affiliation(s)
- Feifeng Song
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014China
- Zhejiang Provincial Clinical Research Center for malignant tumorHangzhou310014China
| | - Zhentao Zhang
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Weifeng Liu
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou310009China
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014China
- Zhejiang Provincial Clinical Research Center for malignant tumorHangzhou310014China
| | - Xiaoping Hu
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Qiyue Wang
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Wanli Zhang
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Luqi Ge
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Chengwu Zhang
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasion SurgeryZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Qing Hu
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Hui Qin
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Song Zhang
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014China
- Zhejiang Provincial Clinical Research Center for malignant tumorHangzhou310014China
| | - Xinxin Ren
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Weijiao Fan
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014China
- Zhejiang Provincial Clinical Research Center for malignant tumorHangzhou310014China
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer CenterDepartment of PharmacyZhejiang Provincial People's Hospital (Affiliated People's Hospital)Hangzhou Medical CollegeHangzhou310014China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014China
- Zhejiang Provincial Clinical Research Center for malignant tumorHangzhou310014China
| |
Collapse
|
2
|
Heberle A, Cappuccio E, Andric A, Kuen T, Simonini A, Weiss AKH. Mitochondrial enzyme FAHD1 reduces ROS in osteosarcoma. Sci Rep 2024; 14:9231. [PMID: 38649439 PMCID: PMC11035622 DOI: 10.1038/s41598-024-60012-x] [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: 08/20/2023] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
This study investigated the impact of overexpressing the mitochondrial enzyme Fumarylacetoacetate hydrolase domain-containing protein 1 (FAHD1) in human osteosarcoma epithelial cells (U2OS) in vitro. While the downregulation or knockdown of FAHD1 has been extensively researched in various cell types, this study aimed to pioneer the exploration of how increased catalytic activity of human FAHD1 isoform 1 (hFAHD1.1) affects human cell metabolism. Our hypothesis posited that elevation in FAHD1 activity would lead to depletion of mitochondrial oxaloacetate levels. This depletion could potentially result in a decrease in the flux of the tricarboxylic acid (TCA) cycle, thereby accompanied by reduced ROS production. In addition to hFAHD1.1 overexpression, stable U2OS cell lines were established overexpressing a catalytically enhanced variant (T192S) and a loss-of-function variant (K123A) of hFAHD1. It is noteworthy that homologs of the T192S variant are present in animals exhibiting increased resistance to oxidative stress and cancer. Our findings demonstrate that heightened activity of the mitochondrial enzyme FAHD1 decreases cellular ROS levels in U2OS cells. However, these results also prompt a series of intriguing questions regarding the potential role of FAHD1 in mitochondrial metabolism and cellular development.
Collapse
Affiliation(s)
- Anne Heberle
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Elia Cappuccio
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Andreas Andric
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Tatjana Kuen
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Anna Simonini
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Alexander K H Weiss
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
3
|
Dhakne P, Pillai M, Mishra S, Chatterjee B, Tekade RK, Sengupta P. Refinement of safety and efficacy of anti-cancer chemotherapeutics by tailoring their site-specific intracellular bioavailability through transporter modulation. Biochim Biophys Acta Rev Cancer 2023; 1878:188906. [PMID: 37172652 DOI: 10.1016/j.bbcan.2023.188906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/20/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
Low intracellular bioavailability, off-site toxicities, and multi drug resistance (MDR) are the major constraints involved in cancer chemotherapy. Many anticancer molecules fail to become a good lead in drug discovery because of their poor site-specific bioavailability. Concentration of a molecule at target sites is largely varied because of the wavering expression of transporters. Recent anticancer drug discovery strategies are paying high attention to enhance target site bioavailability by modulating drug transporters. The level of genetic expression of transporters is an important determinant to understand their ability to facilitate drug transport across the cellular membrane. Solid carrier (SLC) transporters are the major influx transporters involved in the transportation of most anti-cancer drugs. In contrast, ATP-binding cassette (ABC) superfamily is the most studied class of efflux transporters concerning cancer and is significantly involved in efflux of chemotherapeutics resulting in MDR. Balancing SLC and ABC transporters is essential to avoid therapeutic failure and minimize MDR in chemotherapy. Unfortunately, comprehensive literature on the possible approaches of tailoring site-specific bioavailability of anticancer drugs through transporter modulation is not available till date. This review critically discussed the role of different specific transporter proteins in deciding the intracellular bioavailability of anticancer molecules. Different strategies for reversal of MDR in chemotherapy by incorporation of chemosensitizers have been proposed in this review. Targeted strategies for administration of the chemotherapeutics to the intracellular site of action through clinically relevant transporters employing newer nanotechnology-based formulation platforms have been explained. The discussion embedded in this review is timely considering the current need of addressing the ambiguity observed in pharmacokinetic and clinical outcomes of the chemotherapeutics in anti-cancer treatment regimens.
Collapse
Affiliation(s)
- Pooja Dhakne
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India
| | - Megha Pillai
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India
| | - Sonam Mishra
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India
| | - Bappaditya Chatterjee
- SVKM's NMIMS School of Pharmacy and Management, Department of Pharmaceutics, Vaikunthlal Mehta Road, Vile Parle West, Mumbai, Maharashtra 400056, India
| | - Rakesh K Tekade
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India
| | - Pinaki Sengupta
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar 382355, Gujarat, India.
| |
Collapse
|
4
|
Schniers BK, Rajasekaran D, Korac K, Sniegowski T, Ganapathy V, Bhutia YD. PEPT1 is essential for the growth of pancreatic cancer cells: a viable drug target. Biochem J 2021; 478:3757-3774. [PMID: 34569600 PMCID: PMC8589330 DOI: 10.1042/bcj20210377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/17/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022]
Abstract
PEPT1 is a proton-coupled peptide transporter that is up-regulated in PDAC cell lines and PDXs, with little expression in the normal pancreas. However, the relevance of this up-regulation to cancer progression and the mechanism of up-regulation have not been investigated. Herein, we show that PEPT1 is not just up-regulated in a large panel of PDAC cell lines and PDXs but is also functional and transport-competent. PEPT2, another proton-coupled peptide transporter, is also overexpressed in PDAC cell lines and PDXs, but is not functional due to its intracellular localization. Using glibenclamide as a pharmacological inhibitor of PEPT1, we demonstrate in cell lines in vitro and mouse xenografts in vivo that inhibition of PEPT1 reduces the proliferation of the cancer cells. These findings are supported by genetic knockdown of PEPT1 with shRNA, wherein the absence of the transporter significantly attenuates the growth of cancer cells, both in vitro and in vivo, suggesting that PEPT1 is critical for the survival of cancer cells. We also establish that the tumor-derived lactic acid (Warburg effect) in the tumor microenvironment supports the transport function of PEPT1 in the maintenance of amino acid nutrition in cancer cells by inducing MMPs and DPPIV to generate peptide substrates for PEPT1 and by generating a H+ gradient across the plasma membrane to energize PEPT1. Taken collectively, these studies demonstrate a functional link between PEPT1 and extracellular protein breakdown in the tumor microenvironment as a key determinant of pancreatic cancer growth, thus identifying PEPT1 as a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Bradley K. Schniers
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, U.S.A
| | - Devaraja Rajasekaran
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, U.S.A
| | - Ksenija Korac
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, U.S.A
| | - Tyler Sniegowski
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, U.S.A
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, U.S.A
| | - Yangzom D. Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, U.S.A
| |
Collapse
|
5
|
Ayers J, Milner RJ, Cortés-Hinojosa G, Riva A, Bechtel S, Sahay B, Cascio M, Lejeune A, Shiomitsu K, Souza C, Hernandez O, Salute M. Novel application of single-cell next-generation sequencing for determination of intratumoral heterogeneity of canine osteosarcoma cell lines. J Vet Diagn Invest 2021; 33:261-278. [PMID: 33446089 PMCID: PMC7944434 DOI: 10.1177/1040638720985242] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OSA) is a highly aggressive and metastatic neoplasm of both the canine and human patient and is the leading form of osseous neoplasia in both species worldwide. To gain deeper insight into the heterogeneous and genetically chaotic nature of OSA, we applied single-cell transcriptome (scRNA-seq) analysis to 4 canine OSA cell lines. This novel application of scRNA-seq technology to the canine genome required uploading the CanFam3.1 reference genome into an analysis pipeline (10X Genomics Cell Ranger); this methodology has not been reported previously in the canine species, to our knowledge. The scRNA-seq outputs were validated by comparing them to cDNA expression from reverse-transcription PCR (RT-PCR) and Sanger sequencing bulk analysis of 4 canine OSA cell lines (COS31, DOUG, POS, and HMPOS) for 11 genes implicated in the pathogenesis of canine OSA. The scRNA-seq outputs revealed the significant heterogeneity of gene transcription expression patterns within the cell lines investigated (COS31 and DOUG). The scRNA-seq data showed 10 distinct clusters of similarly shared transcriptomic expression patterns in COS31; 12 clusters were identified in DOUG. In addition, cRNA-seq analysis provided data for integration into the Qiagen Ingenuity Pathway Analysis software for canonical pathway analysis. Of the 81 distinct pathways identified within the clusters, 33 had been implicated in the pathogenesis of OSA, of which 18 had not been reported previously in canine OSA.
Collapse
Affiliation(s)
- Jordan Ayers
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| | - Rowan J Milner
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| | | | - Alberto Riva
- ICBR Bioinformatics Core, University of Florida, Gainesville, FL
| | - Sandra Bechtel
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| | - Bikash Sahay
- Infectious Diseases and Immunology, College of Veterinary Medicine
| | - Matthew Cascio
- Pediatric Hematology-Oncology, Department of Pediatrics, College of Medicine
| | - Amandine Lejeune
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| | - Keijiro Shiomitsu
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| | - Carlos Souza
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| | - Oscar Hernandez
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| | - Marc Salute
- Departments of Small Animal Clinical Sciences, College of Veterinary Medicine
| |
Collapse
|
6
|
Labib PL, Yaghini E, Davidson BR, MacRobert AJ, Pereira SP. 5-Aminolevulinic acid for fluorescence-guided surgery in pancreatic cancer: Cellular transport and fluorescence quantification studies. Transl Oncol 2021; 14:100886. [PMID: 33059124 PMCID: PMC7566921 DOI: 10.1016/j.tranon.2020.100886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/30/2022] Open
Abstract
5-Aminolevulinic acid (ALA) is a potential contrast agent for fluorescence-guided surgery in pancreatic ductal adenocarcinoma (PDAC). However, factors influencing ALA uptake in PDAC have not been adequately assessed. We investigated ALA-induced porphyrin fluorescence in PDAC cell lines CFPAC-1 and PANC-1 and pancreatic ductal cell line H6c7 following incubation with 0.25-1.0 mM ALA for 4-48 h. Fluorescence was assessed qualitatively by microscopy and quantitatively by plate reader and flow cytometry. Haem biosynthesis enzymes and transporters were measured by quantitative polymerase chain reaction (qPCR). CFPAC-1 cells exhibited intense fluorescence under microscopy at low concentrations whereas PANC-1 cells and pancreatic ductal cell line H6c7 showed much lower fluorescence. Quantitative fluorescence studies demonstrated fluorescence saturation in the two PDAC cell lines at 0.5 mM ALA, whereas H6c7 cells showed increasing fluorescence with increasing ALA. Based on the PDAC:H6c7 fluorescence ratio studies, lower ALA concentrations provide better contrast between PDAC and benign pancreatic cells. Studies with qPCR showed upregulation of ALA influx transporter PEPT1 in CFPAC-1, whereas PANC-1 upregulated the efflux transporter ABCG2. We conclude that PEPT1 and ABCG2 expression may be key contributory factors for variability in ALA-induced fluorescence in PDAC.
Collapse
Affiliation(s)
- P L Labib
- UCL Institute for Liver & Digestive Health, University College London, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom of Great Britain and Northern Ireland.
| | - E Yaghini
- UCL Division of Surgery & Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom of Great Britain and Northern Ireland.
| | - B R Davidson
- UCL Division of Surgery & Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom of Great Britain and Northern Ireland.
| | - A J MacRobert
- UCL Division of Surgery & Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom of Great Britain and Northern Ireland.
| | - S P Pereira
- UCL Institute for Liver & Digestive Health, University College London, Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
7
|
|
8
|
Ren C, Liu J, Zheng B, Yan P, Sun Y, Yue B. The circular RNA circ-ITCH acts as a tumour suppressor in osteosarcoma via regulating miR-22. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3359-3367. [PMID: 31387405 DOI: 10.1080/21691401.2019.1649273] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Osteosarcoma (OS) is the most prevailing primary bone tumour and the third prevalent tumour in children and adolescents. Despite advanced treatments, the survival rate of OS has not been effectively improved. Here, we intended to investigate the functional impacts of circ-ITCH on OS. Methods: Circ-ITCH expression in OS tissues and cells was evaluated utilizing qRT-PCR. Viability and proliferation of MG63 and Saos-2 cells were determined by utilizing CCK-8 assay and BrdU assay. Transwell assay was utilized to investigate migration and invasion. Western blot was utilized to distinguish apoptosis and metastasis-related proteins expression. Sequentially, the above-mentioned parameters were reassessed when up-regulating miR-22. Results: Circ-ITCH was low expressed in OS tissues and cells. Overexpressing circ-ITCH facilitated apoptosis and repressed viability, proliferation, migration and invasion in MG63 and Saos-2 cells. MiR-22 expression was reduced by overexpressing circ-ITCH. The decline of viability, proliferation, migration and invasion made by overexpressing circ-ITCH was alleviated by up-regulating miR-22. Conclusively, circ-ITCH suppressed PTEN/PI3K/AKT and SP-1 pathways via down-regulating miR-22. Conclusion: Circ-ITCH took effects on apoptosis, viability, proliferation, migration and invasion through restraining PTEN/PI3K/AKT and SP-1 pathways via down-regulating miR-22 in MG63 and Saos-2 cells. Highlights Low expression of circ-ITCH is observed in osteosarcoma tissues and cell lines; Overexpression circ-ITCH suppresses miR-22 expression; Circ-ITCH promotes proliferation and represses apoptosis by up-regulating miR-22; Circ-ITCH promotes migration and invasion by up-regulating miR-22; Circ-ITCH activates PTEN/PI3K/AKT and SP-1 pathways by up-regulating miR-22.
Collapse
Affiliation(s)
- Chongmin Ren
- a Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Jia Liu
- b Department of Pediatrics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Bingxin Zheng
- a Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Peng Yan
- a Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Yuerong Sun
- a Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Bin Yue
- a Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| |
Collapse
|
9
|
Shen WC, Lai YC, Li LH, Liao K, Lai HC, Kao SY, Wang J, Chuong CM, Hung SC. Methylation and PTEN activation in dental pulp mesenchymal stem cells promotes osteogenesis and reduces oncogenesis. Nat Commun 2019; 10:2226. [PMID: 31110221 PMCID: PMC6527698 DOI: 10.1038/s41467-019-10197-x] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/24/2019] [Indexed: 12/13/2022] Open
Abstract
Lineage commitment and tumorigenesis, traits distinguishing stem cells, have not been well characterized and compared in mesenchymal stem cells derived from human dental pulp (DP-MSCs) and bone marrow (BM-MSCs). Here, we report DP-MSCs exhibit increased osteogenic potential, possess decreased adipogenic potential, form dentin pulp-like complexes, and are resistant to oncogenic transformation when compared to BM-MSCs. Genome-wide RNA-seq and differential expression analysis reveal differences in adipocyte and osteoblast differentiation pathways, bone marrow neoplasm pathway, and PTEN/PI3K/AKT pathway. Higher PTEN expression in DP-MSCs than in BM-MSCs is responsible for the lineage commitment and tumorigenesis differences in both cells. Additionally, the PTEN promoter in BM-MSCs exhibits higher DNA methylation levels and repressive mark H3K9Me2 enrichment when compared to DP-MSCs, which is mediated by increased DNMT3B and G9a expression, respectively. The study demonstrates how several epigenetic factors broadly affect lineage commitment and tumorigenesis, which should be considered when developing therapeutic uses of stem cells.
Collapse
Affiliation(s)
- Wen-Ching Shen
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Yung-Chih Lai
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung, 404, Taiwan
| | - Ling-Hui Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 105, Taiwan
| | - Kolin Liao
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Hung-Chang Lai
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Shou-Yen Kao
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, 112, Taiwan
| | - John Wang
- Department of Pathology, China Medical University Hospital, Taichung, 404, Taiwan
| | - Cheng-Ming Chuong
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung, 404, Taiwan
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Shih-Chieh Hung
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan.
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung, 404, Taiwan.
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 105, Taiwan.
| |
Collapse
|
10
|
Miyabe J, Ohgaki R, Saito K, Wei L, Quan L, Jin C, Liu X, Okuda S, Nagamori S, Ohki H, Yoshino K, Inohara H, Kanai Y. Boron delivery for boron neutron capture therapy targeting a cancer-upregulated oligopeptide transporter. J Pharmacol Sci 2019; 139:215-222. [PMID: 30833090 DOI: 10.1016/j.jphs.2019.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/16/2019] [Accepted: 01/24/2019] [Indexed: 12/21/2022] Open
Abstract
Boron neutron capture therapy (BNCT) is a radiotherapy utilizing the neutron capture and nuclear fission reaction of 10B taken up into tumor cells. The most commonly used boron agent in BNCT, p-borono-l-phenylalanine (BPA), is accumulated in tumors by amino acid transporters upregulated in tumor cells. Here, by using dipeptides of BPA and tyrosine (BPA-Tyr and Tyr-BPA), we propose a novel strategy of selective boron delivery into tumor cells via oligopeptide transporter PEPT1 upregulated in various cancers. Kinetic analyses indicated that BPA-Tyr and Tyr-BPA are transported by oligopeptide transporters, PEPT1 and PEPT2. The intrinsic oligopeptide transport activity in tumor cells clearly correlated with PEPT1 protein expression level but not with PEPT2, suggesting that PEPT1 is the predominant oligopeptide transporter at least in tumor cell lines. Furthermore, using BPA-Tyr and Tyr-BPA, boron was successfully delivered into PEPT1-expressing pancreatic cancer AsPC-1 cells via a PEPT1-mediated mechanism. Intravenous administration of BPA-Tyr into the mice bearing AsPC-1 xenograft tumors resulted in significant boron accumulation in the tumors. It is proposed that the oligopeptide transporters, especially PEPT1, are promising candidates for molecular targets of boron delivery in BNCT. The BPA-containing dipeptides would have a potential for the development of novel boron carriers targeting PEPT1.
Collapse
Affiliation(s)
- Junji Miyabe
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keijiro Saito
- Department of Chemistry, Faculty of Science, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Ling Wei
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Lili Quan
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chunhuan Jin
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Xingming Liu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Suguru Okuda
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shushi Nagamori
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroshi Ohki
- Department of Chemistry, Faculty of Science, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Kazuo Yoshino
- Department of Chemistry, Faculty of Science, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
11
|
Nagaoka S. Mystery of Cholesterol-Lowering Peptides, Lactostatin and Soystatin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:3993-3994. [PMID: 29582650 DOI: 10.1021/acs.jafc.8b01025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Affiliation(s)
- Satoshi Nagaoka
- Department of Applied Life Science, Faculty of Applied Biological Sciences , Gifu University , Gifu 501-1193 , Japan
| |
Collapse
|