651
|
Zhang H, Cheng J, Li Z, Xi Y. Identification of hub genes and molecular mechanisms in infant acute lymphoblastic leukemia with MLL gene rearrangement. PeerJ 2019; 7:e7628. [PMID: 31523525 PMCID: PMC6717502 DOI: 10.7717/peerj.7628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Infant acute lymphoblastic leukemia (ALL) with the mixed lineage leukemia (MLL) gene rearrangement (MLL-R) is considered a distinct leukemia from childhood or non-MLL-R infant ALL. To detect key genes and elucidate the molecular mechanisms of MLL-R infant ALL, microarray expression data were downloaded from the Gene Expression Omnibus (GEO) database, and differentially expressed genes (DEGs) between MLL-R and non-MLL-R infant ALL were identified. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were carried out. Then, we constructed a protein-protein interaction (PPI) network and identified the hub genes. Finally, drug-gene interactions were mined. A total of 139 cases of MLL-R infant ALL including 77 (55.4%) fusions with AF4, 38 (27.3%) with ENL, 14 (10.1%) with AF9, and 10 (7.2%) other gene fusions were characterized. A total of 236 up-regulated and 84 down-regulated DEGs were identified. The up-regulated DEGs were mainly involved in homophilic cell adhesion, negative regulation of apoptotic process and cellular response to drug GO terms, while down-regulated DEGs were mainly enriched in extracellular matrix organization, protein kinase C signaling and neuron projection extension GO terms. The up-regulated DEGs were enriched in seven KEGG pathways, mainly involving transcriptional regulation and signaling pathways, and down-regulated DEGs were involved in three main KEGG pathways including Alzheimer’s disease, TGF-beta signaling pathway, and hematopoietic cell lineage. The PPI network included 297 nodes and 410 edges, with MYC, ALB, CD44, PTPRC and TNF identified as hub genes. Twenty-three drug-gene interactions including four up-regulated hub genes and 24 drugs were constructed by Drug Gene Interaction database (DGIdb). In conclusion, MYC, ALB, CD44, PTPRC and TNF may be potential bio-markers for the diagnosis and therapy of MLL-R infant ALL.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Juan Cheng
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zijian Li
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yaming Xi
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
652
|
Zhou J, Du Y, Lu Y, Luan B, Xu C, Yu Y, Zhao H. CD44 Expression Predicts Prognosis of Ovarian Cancer Patients Through Promoting Epithelial-Mesenchymal Transition (EMT) by Regulating Snail, ZEB1, and Caveolin-1. Front Oncol 2019; 9:802. [PMID: 31497537 PMCID: PMC6712994 DOI: 10.3389/fonc.2019.00802] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/07/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives: CD44, a transmembrane glycoprotein, is involved in the generation of a stem cell niche and maintaining stem cell quiescence. The aim of this study was to evaluate its contribution to ovarian cancer prognosis and progression, as well as explore the possible mechanisms. Materials and Methods: The expression of CD44 in tissue microarray of 90 ovarian cancer patients was detected by immunohistochemistry. Kaplan-Meier method and Cox proportional hazard model were used to evaluate the factors associated with 5-year overall survival and disease-free survival. CD44 was knocked down by small interfering RNA, the expression of Snail, ZEB1, and Caveolin-1 in a stable Snail-expressing ovarian cancer cell line HO8910PM-Snail (HOPM-Snail) and its control cell line HO8910PM-vector (HOPM) was detected by western blotting analysis. Cell clone formation, migration, and invasion of HOPM-Snail and HOPM cells with CD44 silencing were examined by 3-D culture assay, wound healing assay, and transwell assay, respectively. Results: Over-expression of CD44 was associated with advanced histological grade (p = 0.014) and FIGO stage (p = 0.001). Multivariate analysis showed that CD44 expression was an independent prognostic factor to predict both overall survival (p = 0.004) and disease-free survival (p = 0.025) of ovarian cancer patients. Down-regulation of CD44 expression by small silencing RNA abrogated both basal Snail expression and TGF-β1-induced Snail expression in HOPM and HOPM-Snail cells. In addition, CD44 knockdown caused a decrease in ZEB1 expression. RPPA data indicated that Caveolin-1 may be another regulative target of CD44, and western blotting analysis confirmed that CD44 knockdown caused an increase in Caveolin-1 expression. However, there was no noticeable reciprocal regulation among ZEB1, Caveolin-1, and Snail. Moreover, CD44 knockdown caused a decrease in cell clone formation, migration, and invasion of HOPM and HOPM-Snail cells. Conclusions: As both Snail and ZEB1 are crucial inducers of epithelial-to-mesenchymal transition (EMT), our data suggested that CD44 may be crucial for the EMT process of ovarian cancer. Therefore, CD44 may be a potential prognostic marker as well as treatment target for ovarian cancer.
Collapse
Affiliation(s)
- Jiayi Zhou
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yan Du
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yiling Lu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Baoxin Luan
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Congjian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yinhua Yu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Hongbo Zhao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
653
|
Lv Y, Huang S. Role of non-coding RNA in pancreatic cancer. Oncol Lett 2019; 18:3963-3973. [PMID: 31579086 PMCID: PMC6757267 DOI: 10.3892/ol.2019.10758] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/05/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a malignant disease that develops rapidly and carries a poor prognosis. Currently, surgery is the only radical treatment. Non-coding RNAs (ncRNAs) are protein-free RNAs produced by genome transcription; they play important roles in regulating gene expression, participating in epigenetic modification, cell proliferation, differentiation and reproduction. ncRNAs also play key roles in the development of cancer; microRNA (miRNA) and long non-coding RNA (lncRNA) may lead the way to new treatments for pancreatic cancer. miRNAs are short-chain ncRNAs (19–24 nt) that inhibit the degradation of protein translation or their target gene mRNAs to regulate gene expression. lncRNAs contain >200 nt of ncRNA and play important regulatory roles in a number of malignant tumors, in terms of tumor cell proliferation, apoptosis, invasion and distant metastasis. lncRNAs can be exploited for the diagnosis and treatment of pancreatic cancer and have substantial prospects for clinical application. Nevertheless, the molecular mechanism of their regulation and function, as well as the significance of other ncRNAs, such as piwi-interacting RNA, in the pathogenesis of pancreatic cancer, are largely unknown. In this review, the structures of ncRNAs with various classifications, as well as the functions and important roles of ncRNAs in the diagnosis and treatment of pancreatic cancer are reviewed.
Collapse
Affiliation(s)
- Yinghao Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shuai Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
654
|
Prooxidative Activity of Celastrol Induces Apoptosis, DNA Damage, and Cell Cycle Arrest in Drug-Resistant Human Colon Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6793957. [PMID: 31485297 PMCID: PMC6710751 DOI: 10.1155/2019/6793957] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/12/2019] [Accepted: 07/18/2019] [Indexed: 01/02/2023]
Abstract
Cancer resistance to chemotherapy is closely related to tumor heterogeneity, i.e., the existence of distinct subpopulations of cancer cells in a tumor mass. An important role is assigned to cancer stem cells (CSCs), a small subset of cancer cells with high tumorigenic potential and capacity of self-renewal and differentiation. These properties of CSCs are sustained by the ability of those cells to maintain a low intracellular reactive oxygen species (ROS) levels, via upregulation of ROS scavenging systems. However, the accumulation of ROS over a critical threshold disturbs CSCs—redox homeostasis causing severe cytotoxic consequences. In the present study, we investigated the capacity of celastrol, a natural pentacyclic triterpenoid, to induce the formation of ROS and, consequently, cell death of the colon cancer cells with acquired resistant to cytotoxic drugs (LOVO/DX cell line). LOVO/DX cells express several important stem-like cell features, including a higher frequency of side population (SP) cells, higher expression of multidrug resistant proteins, overexpression of CSC-specific cell surface marker (CD44), increased expression of DNA repair gene (PARP1), and low intracellular ROS level. We found that celastrol, at higher concentrations (above 1 μM), significantly increased ROS amount in LOVO/DX cells at both cytoplasmic and mitochondrial levels. This prooxidant activity was associated with the induction of DNA double-strand breaks (DSBs) and apoptotic/necrotic cell death, as well as with inhibition of cell proliferation by S phase cell cycle arrest. Coincubation with NAC, a ROS scavenger, completely reversed the above effects. In summary, our results provide evidence that celastrol exhibits effective cytotoxic effects via ROS-dependent mechanisms on drug-resistant colon cancer cells. These findings strongly suggest the potential of celastrol to effectively kill cancer stem-like cells, and thus, it is a promising agent to treat severe, resistant to conventional therapy, colon cancers.
Collapse
|
655
|
Kim DM, Shim YH, Kwon H, Kim JP, Park JI, Kim DH, Kim DH, Kim JH, Jeong YI. CD44 Receptor-Specific and Redox-Sensitive Nanophotosensitizers of Hyaluronic Acid-Chlorin e6 Tetramer Having Diselenide Linkages for Photodynamic Treatment of Cancer Cells. J Pharm Sci 2019; 108:3713-3722. [PMID: 31394112 DOI: 10.1016/j.xphs.2019.07.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/27/2019] [Accepted: 07/19/2019] [Indexed: 01/10/2023]
Abstract
For reactive oxygen species (ROS)-sensitive and CD44 receptor-mediated delivery of photosensitizers, chlorin e6 (ce6) tetramer was synthesized using tetra acid (TA) via selenocystamine linkages and then conjugated with hyaluronic acid (HA) (abbreviated as HAseseCe6TA). HAseseCe6TA nanophotosensitizers were fabricated by dialysis procedure. HAseseCe6TA nanophotosensitizers showed spherical morphology with small particle sizes less than 100 nm and monomodal pattern. When H2O2 was added, size distribution was changed to multimodal pattern and morphological observation showed disintegration of nanophotosensitizers, indicating that HAseseCe6TA nanophotosensitizers have ROS sensitivity. Furthermore, H2O2 addition resulted in acceleration of Ce6 release from HAseseCe6TA nanophotosensitizers. In vitro cell culture study, HAseseCe6TA nanophotosensitizers increase Ce6 uptake ratio, ROS production efficiency, and photodynamic therapy efficacy in both B16F10 cells and CT26 cells. Especially, CD44-receptor blocking of cancer cells by pretreatment of HA showed that fluorescence intensity in B16F10 cells was significantly decreased while fluorescence intensity in CT26 cells was not significantly changed, indicating that HAseseCe6TA nanophotosensitizers can be delivered by CD44 receptor-mediated pathway. In vivo animal tumor xenograft study, HAseseCe6TA nanophotosensitizers was selectively delivered to B16F10 tumor rather than CT26 tumor. These results indicated that HAseseCe6TA nanophotosensitizers have ROS sensitivity and have CD44 receptor-recognition properties.
Collapse
Affiliation(s)
- Doo-Man Kim
- Department of Materials Science and Engineering, Chonnam National University, Gwangju 500757, Republic of Korea
| | - Yong Ho Shim
- R & D Center, UltraV Co. Ltd., Seoul 04779, Republic of Korea
| | - Hanjin Kwon
- R & D Center, UltraV Co. Ltd., Seoul 04779, Republic of Korea
| | - Jong-Pil Kim
- Busan Center, Korea Basic Science Institute, Busan 46241, Republic of Korea
| | - Ji-In Park
- Busan Center, Korea Basic Science Institute, Busan 46241, Republic of Korea
| | - Do Hoon Kim
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Douk-Hoon Kim
- Department of Optometry, Masan University, Gyeongnam 51217, Republic of Korea
| | - Jin Hyeok Kim
- Department of Materials Science and Engineering, Chonnam National University, Gwangju 500757, Republic of Korea.
| | - Young-Il Jeong
- Research Institute of Convergence of Biomedical Sciences, Pusan National University Yangsan Hospital, Gyeongnam 50612, Republic of Korea.
| |
Collapse
|
656
|
KLF4 expression in the surgical cut margin is associated with disease relapse of oral squamous cell carcinoma. Oral Surg Oral Med Oral Pathol Oral Radiol 2019; 128:154-165. [DOI: 10.1016/j.oooo.2019.02.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/20/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023]
|
657
|
Tseng CC, Stanciauskas R, Zhang P, Woo D, Wu K, Kelly K, Gill PS, Yu M, Pinaud F, Lee AS. GRP78 regulates CD44v membrane homeostasis and cell spreading in tamoxifen-resistant breast cancer. Life Sci Alliance 2019; 2:e201900377. [PMID: 31416894 PMCID: PMC6696983 DOI: 10.26508/lsa.201900377] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 12/26/2022] Open
Abstract
GRP78 conducts protein folding and quality control in the ER and shows elevated expression and cell surface translocation in advanced tumors. However, the underlying mechanisms enabling GRP78 to exert novel signaling functions at cell surface are just emerging. CD44 is a transmembrane protein and an important regulator of cancer metastasis, and isoform switch of CD44 through incorporating additional variable exons to the extracellular juxtamembrane region is frequently observed during cancer progression. Using super-resolution dual-color single-particle tracking, we report that GRP78 interacts with CD44v in plasma membrane nanodomains of breast cancer cells. We further show that targeting cell surface GRP78 by the antibodies can effectively reduce cell surface expression of CD44v and cell spreading of tamoxifen-resistant breast cancer cells. Our results uncover new functions of GRP78 as an interacting partner of CD44v and as a regulator of CD44v membrane homeostasis and cell spreading. This study also provides new insights into anti-CD44 therapy in tamoxifen-resistant breast cancer.
Collapse
Affiliation(s)
- Chun-Chih Tseng
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Ramunas Stanciauskas
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Pu Zhang
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA, USA
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Dennis Woo
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kaijin Wu
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, USA
| | - Kevin Kelly
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, USA
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Parkash S Gill
- Department of Pathology, University of Southern California, Los Angeles, CA, USA
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Min Yu
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Fabien Pinaud
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
- Department of Physics and Astronomy, University of Southern California, Los Angeles, CA, USA
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA
- University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
658
|
Hyaluronan-CD44 axis orchestrates cancer stem cell functions. Cell Signal 2019; 63:109377. [PMID: 31362044 DOI: 10.1016/j.cellsig.2019.109377] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023]
Abstract
The prominent role of CD44 in tumor cell signaling together with its establishment as a cancer stem cell (CSC) marker for various tumor entities imply a key role for CD44 in CSC functional properties. Hyaluronan, the main ligand of CD44, is a major constituent of CSC niche and, therefore, the hyaluronan-CD44 signaling axis is of functional importance in this special microenvironment. This review aims to provide recent advances in the importance of hyaluronan-CD44 interactions in the acquisition and maintenance of a CSC phenotype. Hyaluronan-CD44 axis has a substantial impact on stemness properties of CSCs and drug resistance through induction of EMT program, oxidative stress resistance, secretion of extracellular vesicles/exosomes and epigenetic control. Potential therapeutic approaches targeting CSCs based on the hyaluronan-CD44 axis are also presented.
Collapse
|
659
|
Martin ME, Reaves DK, Jeffcoat B, Enders JR, Costantini LM, Yeyeodu ST, Botta D, Kavanagh TJ, Fleming JM. Silver nanoparticles alter epithelial basement membrane integrity, cell adhesion molecule expression, and TGF-β1 secretion. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 21:102070. [PMID: 31351238 DOI: 10.1016/j.nano.2019.102070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/21/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
Silver nanoparticles (AgNPs) are widely used in consumer and pharmaceutical products due to their antipathogenic properties. However, safety concerns have been raised due to their bioactive properties. While reports have demonstrated AgNPs can embed within the extracellular matrix, their effects on basement membrane (BM) production, integrin engagement, and tissue-integrity are not well-defined. This study analyzed the effects of AgNPs on BM production, composition and integrin/focal adhesion interactions in representative lung, esophageal, breast and colorectal epithelia models. A multidisciplinary approach including focused proteomics, QPCR arrays, pathway analyses, and immune-based, structural and functional assays was used to identify molecular and physiological changes in cell adhesions and the BM induced by acute and chronic AgNP exposure. Dysregulated targets included CD44 and transforming growth factor-beta, two proteins frequently altered during pathogenesis. Results indicate AgNP exposure interferes with BM and cell adhesion dynamics, and provide insight into the mechanisms of AgNP-induced disruption of epithelial physiology.
Collapse
Affiliation(s)
- Megan E Martin
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Denise K Reaves
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Breanna Jeffcoat
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Jeffrey R Enders
- Molecular Education, Technology and Research Innovation Center, North Carolina State University, Raleigh, NC, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Lindsey M Costantini
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | | | - Diane Botta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Jodie M Fleming
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA; Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
660
|
Xu K, Sun G, Li M, Chen H, Zhang Z, Qian X, Li P, Xu L, Huang W, Wang X. Glibenclamide Targets Sulfonylurea Receptor 1 to Inhibit p70S6K Activity and Upregulate KLF4 Expression to Suppress Non-Small Cell Lung Carcinoma. Mol Cancer Ther 2019; 18:2085-2096. [PMID: 31341030 DOI: 10.1158/1535-7163.mct-18-1181] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/19/2019] [Accepted: 07/17/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Kexin Xu
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Geng Sun
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Min Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hongling Chen
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zuhao Zhang
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xixi Qian
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ping Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu Province, China
| | - Wenbin Huang
- Department of Pathology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xuerong Wang
- Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
661
|
Zhao L, Zheng X, Liu J, Zheng R, Yang R, Wang Y, Sun L. PPAR signaling pathway in the first trimester placenta from in vitro fertilization and embryo transfer. Biomed Pharmacother 2019; 118:109251. [PMID: 31351426 DOI: 10.1016/j.biopha.2019.109251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 01/27/2023] Open
Abstract
Placenta is a temporary critical organ related to fetal development and pregnancy outcomes. And epidemiologic data demonstrate an increased risk of placental abnormality after in vitro fertilization and embryo transfer (IVF-ET). This study aims to explore the molecular mechanism for PPAR signaling pathway in placenta subjected to IVF-ET in the first trimester. Four first trimester placenta samples from double chorionic twins to single reduction in IVF-ET only because of oviducts factors. The other four control placenta samples from double chorionic twin were derived from those unplanned spontaneously conceived pregnancy after the legal termination. Affymetrix HG-U133 Plus 2.0 Array was performed to evaluate the global gene expressions. We confirmed microarray results from 10 significant differential genes using RT-qPCR. And 10 deregulated gene products were stained in the first trimester placenta by immunohistochemistry. These differentially expressed genes in IVF-ET placentas were submitted to functional annotation of clustering tools of bioinformatics resources and gene ontology enrichment analysis. Schematic representation of placental PPAR signaling pathway was labelled by Kyoto Encyclopedia of Genes and Genomes (KEGG). Analysis results of early placental PPAR signaling pathway gene expression from 8 women demonstrated 34 genes with a significant change in expression between IVF-ET and control group, 25 up-regulated; 9 down-regulated. KEGG pathway analysis indicated that IVF-ET manipulation extensively over-activated PPAR signaling pathway. Immune tolerance, trophoblast invasion, syncytia formation, lipid and glucose metabolism, inflammatory response and other complex biological functions were disturbed. RT-qPCR results and proteins staining intensity were consisted with microarray. Placental gene expressions and functions in PPAR signaling pathway were affected by IVF-ET treatment in the first trimester, which may offer a potential mechanism for the pathogenesis of various adverse outcomes during the perinatal period.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Xiuli Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Jingfang Liu
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Rong Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Rui Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Lifang Sun
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
| |
Collapse
|
662
|
Role of cell surface proteoglycans in cancer immunotherapy. Semin Cancer Biol 2019; 62:48-67. [PMID: 31336150 DOI: 10.1016/j.semcancer.2019.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/23/2022]
Abstract
Over the past few decades, understanding how tumor cells evade the immune system and their communication with their tumor microenvironment, has been the subject of intense investigation, with the aim of developing new cancer immunotherapies. The current therapies against cancer such as monoclonal antibodies against checkpoint inhibitors, adoptive T-cell transfer, cytokines, vaccines, and oncolytic viruses have managed to improve the clinical outcome of the patients. However, in some tumor entities, the response is limited and could benefit from the identification of novel therapeutic targets. It is known that tumor-extracellular matrix interplay and matrix remodeling are necessary for anti-tumor and pro-tumoral immune responses. Proteoglycans are dominant components of the extracellular matrix and are a highly heterogeneous group of proteins characterized by the covalent attachment of a specific linear carbohydrate chain of the glycosaminoglycan type. At cell surfaces, these molecules modulate the expression and activity of cytokines, chemokines, growth factors, adhesion molecules, and function as signaling co-receptors. By these mechanisms, proteoglycans influence the behavior of cancer cells and their microenvironment during the progression of solid tumors and hematopoietic malignancies. In this review, we discuss why cell surface proteoglycans are attractive pharmacological targets in cancer, and we present current and recent developments in cancer immunology and immunotherapy utilizing proteoglycan-targeted strategies.
Collapse
|
663
|
Hong F, Pan S, Guo Y, Xu P, Zhai Y. PPARs as Nuclear Receptors for Nutrient and Energy Metabolism. Molecules 2019; 24:molecules24142545. [PMID: 31336903 PMCID: PMC6680900 DOI: 10.3390/molecules24142545] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
It has been more than 36 years since peroxisome proliferator-activated receptors (PPARs) were first recognized as enhancers of peroxisome proliferation. Consequently, many studies in different fields have illustrated that PPARs are nuclear receptors that participate in nutrient and energy metabolism and regulate cellular and whole-body energy homeostasis during lipid and carbohydrate metabolism, cell growth, cancer development, and so on. With increasing challenges to human health, PPARs have attracted much attention for their ability to ameliorate metabolic syndromes. In our previous studies, we found that the complex functions of PPARs may be used as future targets in obesity and atherosclerosis treatments. Here, we review three types of PPARs that play overlapping but distinct roles in nutrient and energy metabolism during different metabolic states and in different organs. Furthermore, research has emerged showing that PPARs also play many other roles in inflammation, central nervous system-related diseases, and cancer. Increasingly, drug development has been based on the use of several selective PPARs as modulators to diminish the adverse effects of the PPAR agonists previously used in clinical practice. In conclusion, the complex roles of PPARs in metabolic networks keep these factors in the forefront of research because it is hoped that they will have potential therapeutic effects in future applications.
Collapse
Affiliation(s)
- Fan Hong
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Shijia Pan
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yuan Guo
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | - Yonggong Zhai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
- Key Laboratory for Cell Proliferation and Regulation Biology of State Education Ministry, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
664
|
Shin KH, Kim RH. An Updated Review of Oral Cancer Stem Cells and Their Stemness Regulation. Crit Rev Oncog 2019; 23:189-200. [PMID: 30311574 DOI: 10.1615/critrevoncog.2018027501] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer stem cells (CSCs; also known as tumor-initiating cells) are a small population of cancer cells that retain characteristics similar to those of normal stem cells. CSCs are known to be responsible for metastasis, drug resistance, and cancer recurrence. Thus, controlling CSCs may provide an effective therapeutic intervention that inhibits tumor growth and aggressiveness. Despite the importance of targeting CSCs in cancer therapy, the detailed nature of oral CSCs remains underexplored. This article reviews the current understanding of oral CSCs, with emphasis on recent advances in novel signaling pathways involved in their stemness regulation.
Collapse
Affiliation(s)
- Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095
| |
Collapse
|
665
|
BRCA1 regulates the cancer stem cell fate of breast cancer cells in the context of hypoxia and histone deacetylase inhibitors. Sci Rep 2019; 9:9702. [PMID: 31273285 PMCID: PMC6609720 DOI: 10.1038/s41598-019-46210-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 06/21/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer cell stemness is essential for enabling malignant progression and clonal evolution. Cancer cell fate is likely determined by complex mechanisms involving both cell-intrinsic pathways and stress signals from tumor microenvironment. In this study, we examined the role of the tumor suppressor BRCA1 and hypoxia in the regulation of cancer cell stemness using genetically matched breast cancer cell lines. We have found that BRCA1, a multifunctional protein involved in DNA repair and epigenetic regulation, plays a critical role in the regulation of cancer stem cell (CSC)-like characteristics. Reconstitution of BRCA1 resulted in significant decrease of the CSC-like populations in breast cancer cells whereas down-regulation of BRCA1 resulted in significant increase of the CSC-like populations. Furthermore, the BRCA1-reconstituted tumor cells are more sensitive to the histone deacetylase (HDAC) inhibitor-induced loss of stemness than the BRCA1-deficient cells are. Surprisingly, hypoxia preferentially blocks HDAC inhibitor-induced differentiation of the BRCA1-reconstituted breast cancer cells. In light of the increasing numbers of clinical trials involving HDAC inhibitors in human cancers, our observations strongly suggest that the BRCA1 status and tumor hypoxia should be considered as potentially important clinical parameters that may affect the therapeutic efficacy of HDAC inhibitors.
Collapse
|
666
|
Ovarian Cancer Stem Cells: Role in Metastasis and Opportunity for Therapeutic Targeting. Cancers (Basel) 2019; 11:cancers11070934. [PMID: 31277278 PMCID: PMC6678643 DOI: 10.3390/cancers11070934] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 06/29/2019] [Accepted: 06/30/2019] [Indexed: 02/08/2023] Open
Abstract
Ovarian cancer (OC) is a heterogeneous disease usually diagnosed at a late stage. Cancer stem cells (CSCs) that exist within the bulk tumor survive first-line chemotherapy and contribute to resistant disease with metastasis. Understanding the key features of CSC biology provides valuable opportunities to develop OCSC-directed therapeutics, which will eventually improve the clinical outcomes of patients. Although significant developments have occurred since OCSCs were first described, the involvement of CSCs in ovarian tumor metastasis is not fully understood. Here, we discuss putative CSC markers and the fundamental role of CSCs in facilitating tumor dissemination in OC. Additionally, we focus on promising CSC-targeting strategies in preclinical and clinical studies of OC and discuss potential challenges in CSC research.
Collapse
|
667
|
Zuo X, Deguchi Y, Xu W, Liu Y, Li HS, Wei D, Tian R, Chen W, Xu M, Yang Y, Gao S, Jaoude JC, Liu F, Chrieki SP, Moussalli MJ, Gagea M, Sebastian MM, Zheng X, Tan D, Broaddus R, Wang J, Ajami NJ, Swennes AG, Watowich SS, Shureiqi I. PPARD and Interferon Gamma Promote Transformation of Gastric Progenitor Cells and Tumorigenesis in Mice. Gastroenterology 2019; 157:163-178. [PMID: 30885780 PMCID: PMC6581611 DOI: 10.1053/j.gastro.2019.03.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 02/20/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS The peroxisome proliferator-activated receptor delta (PPARD) regulates cell metabolism, proliferation, and inflammation and has been associated with gastric and other cancers. Villin-positive epithelial cells are a small population of quiescent gastric progenitor cells. We expressed PPARD from a villin promoter to investigate the role of these cells and PPARD in development of gastric cancer. METHODS We analyzed gastric tissues from mice that express the Ppard (PPARD1 and PPARD2 mice) from a villin promoter, and mice that did not carry this transgene (controls), by histology and immunohistochemistry. We performed cell lineage-tracing experiments and analyzed the microbiomes, chemokine and cytokine production, and immune cells and transcriptomes of stomachs of these mice. We also performed immunohistochemical analysis of PPARD levels in 2 sets of human gastric tissue microarrays. RESULTS Thirty-eight percent of PPARD mice developed spontaneous, invasive gastric adenocarcinomas, with severe chronic inflammation. Levels of PPARD were increased in human gastric cancer tissues, compared with nontumor tissues, and associated with gastric cancer stage and grade. We found an inverse correlation between level of PPARD in tumor tissue and patient survival time. Gastric microbiomes from PPARD and control mice did not differ significantly. Lineage-tracing experiments identified villin-expressing gastric progenitor cells (VGPCs) as the origin of gastric tumors in PPARD mice. In these mice, PPARD up-regulated CCL20 and CXCL1, which increased infiltration of the gastric mucosa by immune cells. Immune cell production of inflammatory cytokines promoted chronic gastric inflammation and expansion and transformation of VGPCs, leading to tumorigenesis. We identified a positive-feedback loop between PPARD and interferon gamma signaling that sustained gastric inflammation to induce VGPC transformation and gastric carcinogenesis. CONCLUSIONS We found PPARD overexpression in VPGCs to result in inflammation, dysplasia, and tumor formation. PPARD and VGPCs might be therapeutic targets for stomach cancer.
Collapse
Affiliation(s)
- Xiangsheng Zuo
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Yasunori Deguchi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiguo Xu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yi Liu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Haiyan S. Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rui Tian
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weidong Chen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Min Xu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yaying Yang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shen Gao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan C. Jaoude
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Fuyao Liu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sarah P. Chrieki
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Micheline J. Moussalli
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Manu M. Sebastian
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dongfeng Tan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Russell Broaddus
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nadim J. Ajami
- Alkek Center for Metagenomics and Microbiome Research and Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alton G. Swennes
- Center for Comparative Medicine and Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephanie S. Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Imad Shureiqi
- Departments of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
668
|
Feng Z, Meng S, Zhou H, Xu Z, Tang Y, Li P, Liu C, Huang Y, Wu M. Functions and Potential Applications of Circular RNAs in Cancer Stem Cells. Front Oncol 2019; 9:500. [PMID: 31263676 PMCID: PMC6584801 DOI: 10.3389/fonc.2019.00500] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022] Open
Abstract
Circular RNAs (circRNAs) were discovered in the 1970s, but they have drawn increasing attention in recent years. Currently, we know that circRNAs are not "wrongly spliced" during transcription but play important roles in the initiation and development of various diseases, including cancers. Recently, a growing number of studies have suggested that cancer stem cells (CSCs) may contribute to the origination and maintenance of cancers. This review briefly introduces the major functions of circRNAs, including interacting with other noncoding RNAs, competing with pre-mRNA splicing, binding with proteins to form a scaffold, promoting protein nuclear translocation and even translating proteins in a cap-independent manner. Furthermore, we describe the regulatory mechanism of circRNAs in CSC phenotypes and discuss the potential clinical applications of circRNAs in CSC-targeted therapy, including functioning as new biomarkers, acting as vaccines and breaking the therapeutic resistance of CSCs. Finally, we discuss the major limitations and challenges in the field, which will be beneficial for the future clinical use of circRNAs.
Collapse
Affiliation(s)
- Ziyang Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Shujuan Meng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Hecheng Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Zihao Xu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Ying Tang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Peiyao Li
- The Xiangya Hospital, Central South University, Changsha, China
| | - Changhong Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Yongkai Huang
- The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| |
Collapse
|
669
|
Sahin Inan ZD, Unver Saraydin S. Immunohistochemical profile of CD markers in experimental neural tube defect. Biotech Histochem 2019; 94:617-627. [PMID: 31184499 DOI: 10.1080/10520295.2019.1622783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Neural tube defects (NTDs) are the second most common birth defects worldwide. Stem cells play a critical role in the mechanisms underlying NTDs. We established an experimental NTD model in rats using retinoic acid (RA). We used mesenchymal and hemopoietic stem cell markers to determine their distribution in the mesenchyme in and around the neuroepithelium during the embryonic and fetal periods in both cranial and caudal regions. Adult female rats were given RA on days 5 and 10 of gestation and olive oil was administered to the control group. On days 10.5 and 15.5, embryos in the experimental and control groups were removed from the uterus. Embryos were embedded in paraffin and serial sections of the cranial and caudal neural tube were examined. We found severe cranial and caudal defects including axial rotation in the experimental groups using histochemistry. We used CD44, CD56, CD73, CD90, CD105, CD271 antibodies as mesenchymal stem cell markers and CD14, CD45 as hemopoietic stem cell markers. More CD44, CD56, CD90, CD105 and CD14 were detected during the embryonic period than the fetal period. CD73 was more frequent during the fetal period, whereas CD271 and CD45 were not significantly different. When CD44, CD56, CD73, CD90, CD105, CD271 immunostaining was found, NTDs were decreased early and increased later. We found no significant difference between CD14 and CD45. Formation of NTDs was due to deterioration of the of the neuroepithelial and surrounding stem cells. One reason for the formation of NTDs is that stem cells may develop defective cell-cell or cell-matrix interactions.
Collapse
Affiliation(s)
- Z D Sahin Inan
- Department of Histology-Embryology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| | - S Unver Saraydin
- Department of Histology-Embryology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
670
|
Muhlebach MS, Sha W, MacIntosh B, Kelley TJ, Muenzer J. Metabonomics reveals altered metabolites related to inflammation and energy utilization at recovery of cystic fibrosis lung exacerbation. Metabol Open 2019; 3:100010. [PMID: 32812947 PMCID: PMC7424819 DOI: 10.1016/j.metop.2019.100010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/31/2019] [Accepted: 06/01/2019] [Indexed: 02/07/2023] Open
Abstract
Background Cystic fibrosis lung disease is characterized by chronic bacterial infections in the setting of mucus abnormalities. Patients experience periodic exacerbations that manifest with increased respiratory symptoms that require intensification of therapy with enhanced airway clearance and intravenous (IV) antibiotics. Objectives In an observational study we tested if the profile of metabolites in serum distinguished the pre-from post-exacerbation state and which systemically measurable pathways were affected during the process to recovery. Methods Serum collected within 48 h of start and completion, respectively of IV antibiotics was collected from people with CF ages 6–30 years. Three day food records were collected prior to each sample. To reduce variation between subjects only subjects who had pancreatic insufficiency, had similar CF mutations, and did not have CF liver disease or diabetes were included. Metabolomic profiling was conducted by Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy with metabolites being identified based on retention time/index, mass to charge ratio and comparison to known compounds. Biostatistical analyses used paired t-test with correction for multiple comparisons and orthogonal partial least square discriminant analysis. Results Thirty subjects (20 male) with a mean ± SEM age of 15.3 ± 1.2 years participated, 17 of whom had matched food-records. Lung function was significantly improved post-therapy compared to pre-therapy, (mean ± SEM) 75 ± 4% vs. 68 ± 4% predicted (n = 26). Serum metabonomics showed distinction of the pre-vs. post-therapy groups with 123 compounds contributing to the differentiation pre-versus post-antibiotics by multiple biostatistical analyses. Compounds and pathways affected included bile acids and microbial derived amino acid metabolites, increases in lipid classes of the glycerophospholipid, glycerolipids, cholesterol, phopsholipids, and most pronounced, the class of sphingolipids. Changes in n6/n3 fatty acids, decreased polyamines but increased metabolites in the nitric oxide pathway, and changes in the tryptophan-kynurenine pathway indicated decreased inflammation at resolution of exacerbation. Conclusions Changes in serum metabolites that distinguished CF pulmonary exacerbation vs. resolution of symptoms showed evidence of decreased inflammation and improvement from a catabolic state. Serum metabonomics distinguishes cystic fibrosis exacerbation to resolution state. Distinguishing pathways indicate alterations in (gut) microbiome and bile acids. Serum metabolites at resolution show decreased inflammation and oxidative stress. Lipid markers indicate improvement from a catabolic state at exacerbation.
Collapse
Key Words
- AA, arachidonate
- ARG, arginase
- BA, bile acids
- BHBA, 3-hydroxybutyrate
- CF, cystic fibrosis
- CFTR, Cystic Fibrosis Transmembrane Regulator
- CRP, C-reactive protein
- DHA, docosahexaenoate
- ESI, electrospray ionization
- FDR, false discovery rate
- FEV1, forced expiratory volume in 1st second
- IDO, indoleamine-2-3-dioxygenase
- IV, intravenous
- NOS, nitric oxide synthase
- ODC, ornithine decarboxylase
- OPLS-DA, orthogonal partial least square discriminant analysis
- QC, quality control
- RI, retention time/index
- UNC, University of North Carolina at Chapel Hill
- UPLC, ultrahigh performance liquid chromatography-tandem mass spectroscopy
- VIP, variable influence on projection score
- n3-DPA, docosapentaenoate
- q, significance at a 5% FDR cut-off
Collapse
Affiliation(s)
- Marianne S Muhlebach
- Dept. Pediatrics, Division Pulmonary Medicine, University of North Carolina at Chapel Hill, 450 MacNider, 330 S. Columbia Road, Chapel Hill, NC, 27599-7217, USA.,Marsico Lung Institute, Chapel Hill, NC, USA
| | - Wei Sha
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, 150 Research Campus Dr, Kannapolis, NC 28081, NC, USA
| | - Beth MacIntosh
- Metabolic and Nutrition Research Core, UNC Healthcare, Nutrition and Food Services Department, 102 Mason Farm Rd, CB#7777, Chapel Hill, NC, USA
| | - Thomas J Kelley
- Departments of Pediatrics/Pharmacology, Case Western Reserve University, 833 BRB, 10900, Euclid Ave, Cleveland, OH, USA
| | - Joseph Muenzer
- Dept. Pediatrics, Division Metabolism and Genetics, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
671
|
Alinezhadbalalami N, Douglas TA, Balani N, Verbridge SS, Davalos RV. The feasibility of using dielectrophoresis for isolation of glioblastoma subpopulations with increased stemness. Electrophoresis 2019; 40:2592-2600. [PMID: 31127957 DOI: 10.1002/elps.201900026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 01/17/2023]
Abstract
Cancer stem cells (CSCs) are aggressive subpopulations with increased stem-like properties. CSCs are usually resistant to most standard therapies and are responsible for tumor repropagation. Similar to normal stem cells, isolation of CSCs is challenging due to the lack of reliable markers. Antigen-based sorting of CSCs usually requires staining with multiple markers, making the experiments complicated, expensive, and sometimes unreliable. Here, we study the feasibility of using dielectrophoresis (DEP) for isolation of glioblastoma cells with increased stemness. We culture a glioblastoma cell line in the form of neurospheres as an in vitro model for glioblastoma stem cells. We demonstrate that spheroid forming cells have higher expression of stem cell marker, nestin. Next, we show that dielectric properties of neurospheres change as a result of changing culture conditions. Our results indicate that spheroid forming cells need higher voltages to experience the same DEP force magnitude compared to normal monolayer cultures of glioblastoma cell line. This study confirms the possibility of using DEP to isolate glioblastoma stem cells.
Collapse
Affiliation(s)
- Nastaran Alinezhadbalalami
- Bioelectromechanical Systems Lab, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, USA.,Laboratory of Integrative Tumor Ecology, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| | - Temple A Douglas
- Bioelectromechanical Systems Lab, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| | - Nikita Balani
- Bioelectromechanical Systems Lab, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| | - Scott S Verbridge
- Laboratory of Integrative Tumor Ecology, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| | - Rafael V Davalos
- Bioelectromechanical Systems Lab, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, USA
| |
Collapse
|
672
|
Weng J, Han X, Liu K, Yang J, Wei S, Zhang Y, Zeng F, Li Y, Shen L, Gao Y. CD44 3'-Untranslated Region Functions as a Competing Endogenous RNA to Enhance NK Sensitivity of Liver Cancer Stem Cell by Regulating ULBP2 Expression. Int J Biol Sci 2019; 15:1664-1675. [PMID: 31360109 PMCID: PMC6643214 DOI: 10.7150/ijbs.35216] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/15/2019] [Indexed: 12/18/2022] Open
Abstract
Liver CSCs are a rare subpopulation of heterogenous liver cancer cells with self-renewal and differentiation properties, which has emerged as a promising therapeutic target. Compelling data shows that NK cells selectively eliminate human cancer derived CSCs like colorectal carcinoma, melanoma, and glioblastoma. But the effect of NK cells on liver CSCs still remains unknown. To study the cytotoxic effect of NK cells on liver CSCs and the mechanism, we performed cytotoxicity assay, ELISA assays, CRISPRi, qRT-PCR, immunoblotting, RNA immunoprecipitation, and luciferase reporter using two types of CSCs reprogrammed from HCC. CSCs derived from liver cancer were susceptible to NK cell mediated cytotoxicity. The susceptibility of liver CSCs to NK cell-mediated cytotoxicity declined significantly after silencing CD44 by CRISPRi-mediated gene knockdown. CD44 3ʹ UTR functioned as a ceRNA to regulate the expression of ULBP2 mainly by competing miR-34a. CD44 3ʹ UTR functioned as a ceRNA to enhance NK sensitivity of liver cancer stem cell by regulating ULBP2 expression.
Collapse
Affiliation(s)
- Jun Weng
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Xu Han
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Kaiyu Liu
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Jiong Yang
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiruo Wei
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yue Zhang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Fanhong Zeng
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yang Li
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Li Shen
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yi Gao
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| |
Collapse
|
673
|
Abstract
Cancer-initiating cells (CIC) are the driving force in tumor progression. There is strong evidence that CIC fulfill this task via exosomes (TEX), which modulate and reprogram stroma, nontransformed cells, and non-CIC. Characterization of CIC, besides others, builds on expression of CIC markers, many of which are known as metastasis-associated molecules. We here discuss that the linkage between CIC/CIC-TEX and metastasis-associated molecules is not fortuitously, but relies on the contribution of these markers to TEX biogenesis including loading and TEX target interactions. In addition, CIC markers contribute to TEX binding- and uptake-promoted activation of signaling cascades, transcription initiation, and translational control. Our point of view will be outlined for pancreas and colon CIC highly expressing CD44v6, Tspan8, EPCAM, claudin7, and LGR5, which distinctly but coordinately contribute to tumor progression. Despite overwhelming progress in unraveling the metastatic cascade and the multiple tasks taken over by CIC-TEX, there remains a considerable gap in linking CIC biomarkers, TEX, and TEX-initiated target modulation with metastasis. We will try to outline possible bridges, which could allow depicting pathways for new and expectedly powerful therapeutic interference with tumor progression.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - Margot Zöller
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany.
| |
Collapse
|
674
|
Mai J, Zhong ZY, Guo GF, Chen XX, Xiang YQ, Li X, Zhang HL, Chen YH, Xu XL, Wu RY, Yu Y, Li ZL, Peng XD, Huang Y, Zhou LH, Feng GK, Guo X, Deng R, Zhu XF. Polo-Like Kinase 1 phosphorylates and stabilizes KLF4 to promote tumorigenesis in nasopharyngeal carcinoma. Theranostics 2019; 9:3541-3554. [PMID: 31281496 PMCID: PMC6587166 DOI: 10.7150/thno.32908] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: Advanced nasopharyngeal carcinoma (NPC) is an aggressive disease with no targeted therapies and poor outcomes. New innovative targets are urgently needed. KLF4 has been extensively studied in the context of tumors, and current data suggest that it can act as either a tissue-specific tumor-inhibiting or a tumor-promoting gene. Here, we found that KLF4 played as a tumor-promoting gene in NPC, and could be mediated by PLK1. Methods: Tissue immunohistochemistry (IHC) assay was performed to identify the role of KLF4 in NPC. Global gene expression experiments were performed to explore the molecular mechanisms underlying KLF4-dependent tumorigenesis. Small-molecule kinase inhibitor screening was performed to identify potential upstream kinases of KLF4. The pharmacologic activity of polo-like kinase inhibitor volasertib (BI6727) in vitro and in vivo was determined. Result: Our investigation showed that high expression of KLF4 was correlated with poor prognosis in NPC. Moreover, genome-wide profiling revealed that KLF4 directly activated oncogenic programmes, including gene sets associated with KRAS, VEGF, and MYC signalling. We further found that inhibition of polo-like kinase 1 could downregulate the expression of KLF4 and that PLK1 directly phosphorylated KLF4 at Ser234. Notably, phosphorylation of KLF4 by PLK1 caused the recruitment and binding of the E3 ligase TRAF6, which resulted in KLF4 K32 K63-linked ubiquitination and stabilization. Moreover, KLF4 could enhance TRAF6 expression at the transcriptional level, thus initiating a KLF4-TRAF6 feed-forward loop. Treatment with the PLK1 inhibitor volasertib (BI6727) significantly inhibited tumor growth in nude mice. Conclusion: Our study unveiled a new PLK1-TRAF6-KLF4 feed-forward loop. The resulting increase in KLF4 ubiquitination leads to stabilization and upregulation of KLF4, which leads to tumorigenesis in NPC. These results expand our understanding of the role of KLF4 in NPC and validate PLK1 inhibitors as potential therapeutic agents for NPC, especially cancer patients with KLF4 overexpression.
Collapse
Affiliation(s)
- Jia Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhuo-Yan Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gui-Fang Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of VIP Inpatient, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiu-Xing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of VIP Inpatient, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan-Qun Xiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xuan Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hai-Liang Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu-Hong Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xue-Lian Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui-Yan Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi-Ling Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Dan Peng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li-Huan Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gong-Kan Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiang Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rong Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Feng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
675
|
Zhang Z, Zhao W, Lin X, Gao J, Zhang Z, Shen L. Voltage-dependent calcium channel α2δ1 subunit is a specific candidate marker for identifying gastric cancer stem cells. Cancer Manag Res 2019; 11:4707-4718. [PMID: 31213895 PMCID: PMC6536713 DOI: 10.2147/cmar.s199329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 04/03/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose: Cancer stem cells (CSCs) are a subpopulation of cancer cells with self-renewal property and responsible for tumor malignancy, progression and drug resistance. Researches on CSC-specific markers in gastric cancer remain limited. Our current study explored the expression of voltage-dependent calcium channel α2δ1 subunit and the potential of using α2δ1 as a CSC marker in gastric cancer. We also compared the specificity of α2δ1 and CD44 in identifying gastric cancer stem cells (GCSCs). Materials and methods: Expression of α2δ1 was analyzed in gastric cancer cell lines, patient-derived xenograft (PDX) models and clinical samples of malignant ascites of gastric cancer patients. α2δ1+ gastric cancer cells were isolated from gastric cancer cell lines. CSC properties of α2δ1+ gastric cancer cells were then verified by subsequent tests both in vitro and in vivo. Results: The expression level of α2δ1 was found to differ drastically among gastric cancer cell lines, PDX models and clinical samples of malignant ascites. α2δ1+ gastric cancer cells sorted from HGC-27 and SGC-7901 cell lines demonstrated significant self-renewal properties, including tumorigenic capacity, sphere-formation capacity and asymmetric differentiation potential. Knockdown of α2δ1 in α2δ1+ HGC-27 significantly inhibited CSC properties and rendered HGC-27 more sensitive to chemotherapy. Flow cytometry showed that α2δ1+ gastric cancer cells accounted for a small fraction of CD44+ gastric cancer cells. Isolated CD44+α2δ1+ HGC-27 cells displayed more significant tumorigenic capacity and sphere-forming capacity compared with their CD44+α2δ1− counterparts. Conclusion: α2δ1+ gastric cancer cells possessed CSC properties. α2δ1 could be a proper marker in identifying GCSCs with superior specificity than CD44. The combination of α2δ1 and CD44 could be used to identify GCSCs with improved accuracy. Knockdown of α2δ1 combined with chemotherapy displayed higher therapeutic efficacy on gastric cancer cells, suggesting that α2δ1 could be a potential target for anticancer treatment.
Collapse
Affiliation(s)
- Ziran Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Wei Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Xiaoting Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Jing Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Zhiqian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| |
Collapse
|
676
|
Zhang L, Song Y, Ling Z, Li Y, Ren X, Yang J, Wang Z, Xia J, Zhang W, Cheng B. R-spondin 2-LGR4 system regulates growth, migration and invasion, epithelial-mesenchymal transition and stem-like properties of tongue squamous cell carcinoma via Wnt/β-catenin signaling. EBioMedicine 2019; 44:275-288. [PMID: 31097406 PMCID: PMC6603804 DOI: 10.1016/j.ebiom.2019.03.076] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/13/2019] [Accepted: 03/26/2019] [Indexed: 01/04/2023] Open
Abstract
Background R-spondins (Rspo) and leucine-rich repeat-containing G-protein-coupled receptors (LGR) play important roles in development, stem cells survival, and tumorigenicity by activating Wnt signaling pathway. Whether R-spondins-LGR signaling affects the progression of squamous cell carcinoma (SCC) remain unknown. This study aims to uncover the role of R-spodin2/LGR4 in tongue SCC (TSCC). Methods The expression of Rspo2 in TSCC specimens and its correlation with TSCC clinical outcome were evaluated. Levels of Rspo2 or LGR4 were altered by pharmacological and genetic approaches, and the effects on TSCC progression were assessed. Findings Aberrantly high levels of Rspo2 were detected in TSCC specimens. Its levels were closely related with lymph node metastasis, clinical stage and survival rate in patients with tongue SCC. Exogenous Rspo2 or overexpression of Rspo2 promoted growth, migration and invasion, epithelial-mesenchymal transition (EMT) and stem-like properties in SCC both in vivo and in vitro. Silence of Rspo2 abolished these phenotypes. LGR4 was functionally upregulated by Rspo2 in TSCC. Overexpression of Rspo2 increased, whereas Rspo2 silencing decreased the expression of LGR4, leading to subsequent phosphorylation of LRP6 and nuclear translocation of β-catenin in TSCC cell lines. This nuclear translocation of β-catenin was associated with a significant alteration in TCF-1, a downstream nuclear transcription factor of β-catenin, as well as its target genes: CD44, CyclinD1 and c-Myc. Interpretation Rspo2-LGR4 system regulates growth, migration and invasion, EMT and stem-like properties of TSCC via Wnt/β-catenin signaling pathway. Rspo2 and LGR4 are aberrantly expressed in TSCC. Rspo2-LGR4 up-regulates growth, migration and invasion, EMT and stem-like properties of TSCC. Rspo2-LGR4 regulates TSCC progression via Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Liping Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yan Song
- First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zihang Ling
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuanyuan Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Xianyue Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing Yang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhi Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Weizhen Zhang
- School of Basic Medical Science, Peking University, Beijing 100191, China.
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510060, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
677
|
Tang Y, Dai Y, Huang X, Li L, Han B, Cao Y, Zhao J. Self-Assembling Peptide-Based Multifunctional Nanofibers for Electrochemical Identification of Breast Cancer Stem-like Cells. Anal Chem 2019; 91:7531-7537. [PMID: 31018636 DOI: 10.1021/acs.analchem.8b05359] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer stem-like cells are closely related with the development and metastasis of tumors. Herein, an electrochemical method is proposed to identify stem-like cells in breast tumor. The core concept of the method is the use of multifunctional nanofibers (MNFs), which are synthesized through facile self-assembly of peptide probes. MNFs can perform three functions, specifically targeting surface biomarker to identify stem-like cells, recruiting silver nanoparticles (AgNPs) to generate electrochemical signals, and providing large amounts of reaction sites to amplify signals. Specially, breast cancer stem cells (BCSCs) are first captured by nucleolin aptamer immobilized on the electrode surface and then selectively recognized by MNFs through the binding with CD44, thereby offering a large number of azide groups for signal labeling. By tracing electrochemical signals from MNF-recruited AgNPs, the method demonstrates to detect target cells as low as 6 cells/mL within a wide linear range from 10 to 5 × 105 cells/mL. Moreover, the method can not only recognize BCSCs with high selectivity in complex environment but also monitor drug-induced stemness changes with high sensitivity, providing promising prospective clinic applications in the future.
Collapse
Affiliation(s)
- Yingying Tang
- Center for Molecular Recognition and Biosensing, School of Life Sciences , Shanghai University , Shanghai 200444 , P. R. China
| | - Yuhao Dai
- Center for Molecular Recognition and Biosensing, School of Life Sciences , Shanghai University , Shanghai 200444 , P. R. China
| | - Xiang Huang
- Department of Oncology , The First Affiliated Hospital of Nanjing Medical University , Nanjing 210029 , P. R. China
| | - Lingling Li
- Center for Molecular Recognition and Biosensing, School of Life Sciences , Shanghai University , Shanghai 200444 , P. R. China
| | - Bing Han
- Center for Molecular Recognition and Biosensing, School of Life Sciences , Shanghai University , Shanghai 200444 , P. R. China
| | - Ya Cao
- Center for Molecular Recognition and Biosensing, School of Life Sciences , Shanghai University , Shanghai 200444 , P. R. China
| | - Jing Zhao
- Center for Molecular Recognition and Biosensing, School of Life Sciences , Shanghai University , Shanghai 200444 , P. R. China
| |
Collapse
|
678
|
Whittle MC, Hingorani SR. Fibroblasts in Pancreatic Ductal Adenocarcinoma: Biological Mechanisms and Therapeutic Targets. Gastroenterology 2019; 156:2085-2096. [PMID: 30721663 PMCID: PMC6486863 DOI: 10.1053/j.gastro.2018.12.044] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 12/13/2022]
Abstract
The desmoplastic reaction of pancreas cancer may begin as a wound healing response to the nascent neoplasm, but it soon creates an insidious shelter that can sustain the growing tumor and rebuff therapy. Among the many cell types subverted by transformed epithelial cells, fibroblasts are recruited and activated to lay a foundation of extracellular matrix proteins and glycosaminoglycans that alter tumor biophysics and signaling. Their near-universal presence in pancreas cancer and ostensible support of disease progression make fibroblasts attractive therapeutic targets. More recently, however, it has also become apparent that diverse subpopulations of fibroblasts with distinct phenotypes and secretomes inhabit the stroma, and that targeted depletion of particular fibroblast subsets could either provide substantial therapeutic benefit or accelerate disease progression. An improved characterization of these fibroblast subtypes, along with their potential relationships to tumor subtypes and mutational repertoires, is needed in order to make anti-fibroblast therapies clinically viable.
Collapse
Affiliation(s)
- Martin C. Whittle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109,Correspondence: Martin C. Whittle, PhD, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, M5-C800, Seattle, WA 98109-1024, , Sunil R. Hingorani, MD, PhD, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, M5-C800, Seattle, WA 98109-1024,
| | - Sunil R. Hingorani
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109,Division of Medical Oncology, University of Washington School of Medicine, Seattle, WA, 98195,Correspondence: Martin C. Whittle, PhD, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, M5-C800, Seattle, WA 98109-1024, , Sunil R. Hingorani, MD, PhD, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, M5-C800, Seattle, WA 98109-1024,
| |
Collapse
|
679
|
Qin Y, Zhao W, Cheng L, Gao J, Bian Z, Wu S, Cheng Z, Tao Y, Ma L. Evaluation of the correlativity of gender determining region Y-box 4, N-cadherin, CD44 and E-cadherin expression in the prognosis of esophageal squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1745-1756. [PMID: 31933993 PMCID: PMC6947138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/26/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND SOX4 is highly expressed in many different tumor types, and SOX4 has been reported in the literature to participate in tumor proliferation, damaging and movement by leading Epithelial-Mesenchymal Transition. Cancer vital cells and Epithelial-Mesenchymal Transition have been repeatedly confirmed to participate during the proliferation, damaging and movement of cancer. This research examined the association of the Epithelial-Mesenchymal Transition-related molecules E-cadherin, N-cadherin, CD44, and SOX4 in the ESCC and aimed for providing inspiration for clinical treatment as well as to indicate a new direction for detecting invasion and forecasting the prospect of affected role using ESCC. METHODS Immunohistochemistry was utilized to observe the expression of the S0X4, N-cadherin, CD44 and E-cadherin proteins. Survival analysis of the positive and negative SOX4, E-cadherin, N-cadherin and CD44 protein expression groups was performed by the Kaplan-Meier approach. OUTCOMES A confirming relationship was observed among the expression of SOX4, N-cadherin or CD44 and tumor diameter, distant metastasis, deepness of damaging, lymph node metastasis, pTNM stage and histological grade (P<0.05). Spearman correlativity calculation displayed that the expression of the SOX4 protein was obviously responded with the expression of the N-cadherin and CD44 proteins. Moreover, the expression of the N-cadherin and CD44 proteins was also positively correlated. The E-cadherin protein was negatively correlated with SOX4, N-cadherin and CD44 protein expression in ESCC. SOX4, N-cadherin, CD44, E-cadherin, age and distant metastasis were determined to be separate elements that influenced the prognosis of patients with ESCC. CONCLUSIONS We found that suppression of ESCC providers can suppress the growth of bad tumors and change therapeutic results for ESCC patient since CD44 supports the induction of Epithelial-Mesenchymal Transition in ESCC.
Collapse
Affiliation(s)
- Yanzi Qin
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu 233003, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Wenjun Zhao
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu 233003, Anhui Province, China
| | - Lili Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu 233003, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Jin Gao
- Department of Clinical Medicine, Bengbu Medical CollegeAnhui Province, China
| | - Zhaonan Bian
- Department of Clinical Medicine, Bengbu Medical CollegeAnhui Province, China
| | - Shiwu Wu
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu 233003, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Zenong Cheng
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu 233003, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Yisheng Tao
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu 233003, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| | - Li Ma
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Medical CollegeBengbu 233003, Anhui Province, China
- Department of Pathology, Bengbu Medical CollegeAnhui Province, China
| |
Collapse
|
680
|
Torres A, Pac-Sosińska M, Wiktor K, Paszkowski T, Maciejewski R, Torres K. CD44, TGM2 and EpCAM as novel plasma markers in endometrial cancer diagnosis. BMC Cancer 2019; 19:401. [PMID: 31035965 PMCID: PMC6489287 DOI: 10.1186/s12885-019-5556-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 03/31/2019] [Indexed: 12/19/2022] Open
Abstract
Background Endometrial cancer (EC) is the most common malignancy of the female reproductive tract. Despite years of research, the accurate screening strategy is still not available in this disease and it is usually diagnosed only after the clinical signs are present. The recent technological advances in analytical methodologies enabled detection of multiple molecules in one, small sample of biological materials. Such approach was undertaken in the presented study. Methods Concentrations of aldehyde dehydrogenase 1 family, member A1 (ALDH1A1), carbonic anhydrase IX (CA9), CD44, epithelial cell adhesion molecule (EpCAM), hepsin, kallikrein-6, mesothelin, midkine, neural cell adhesion molecule L1 (L1CAM), and transglutaminase 2 (TGM2) were measured using MAGPIX®System in plasma samples of 45 EC, 20 healthy controls and 11 patients with endometriosis. Results Significantly increased concentration in EC as compared to healthy controls were found in case of CD44 (p < 0.001), EpCAM (p = 0.033) and TGM2 (p < 0.001). EpCAM and mesothelin concentrations differed based on FIGO stages. Regression analysis revealed marker panels with high accuracy in detection of EC. The highest AUC 0.937 was attributed to the 3-marker panel of CD44/TGM2/EpCAM (84% sensitivity, 100% specificity), FIGO IA samples were discriminated from more advanced stages of EC with the mesothelin/grade 1 model featuring AUC of 0.911 (95.24% sensitivity, 78.26% specificity). Conclusions Novel plasma biomarkers presenting good accuracy in diagnosing EC were found with TGM2 reported for the first time as plasma marker. It was also revealed that endometriosis may share similarities in the pattern of markers alterations characteristic for EC. Electronic supplementary material The online version of this article (10.1186/s12885-019-5556-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Torres
- Laboratory of Biostructure, Chair of Human Anatomy, Medical University of Lublin, Lublin, Poland. .,III Chair and Department of Gynaecology, Medical University of Lublin, Lublin, Poland. .,Pediatric and Adolescent Gynecology Unit, University Children's Hospital, Medical University of Lublin, Lublin, Poland. .,Collegium Anatomicum, Jaczewskiego 4, 20-090, Lublin, Poland.
| | - Małgorzata Pac-Sosińska
- Laboratory of Biostructure, Chair of Human Anatomy, Medical University of Lublin, Lublin, Poland
| | - Krzysztof Wiktor
- Laboratory of Diagnostic Procedures, Medical University of Lublin, Lublin, Poland
| | - Tomasz Paszkowski
- III Chair and Department of Gynaecology, Medical University of Lublin, Lublin, Poland
| | - Ryszard Maciejewski
- Laboratory of Biostructure, Chair of Human Anatomy, Medical University of Lublin, Lublin, Poland
| | - Kamil Torres
- Laboratory of Biostructure, Chair of Human Anatomy, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
681
|
Godavarthy PS, Kumar R, Herkt SC, Pereira RS, Hayduk N, Weissenberger ES, Aggoune D, Manavski Y, Lucas T, Pan KT, Voutsinas JM, Wu Q, Müller MC, Saussele S, Oellerich T, Oehler VG, Lausen J, Krause DS. The vascular bone marrow niche influences outcome in chronic myeloid leukemia via the E-selectin - SCL/TAL1 - CD44 axis. Haematologica 2019; 105:136-147. [PMID: 31018977 PMCID: PMC6939533 DOI: 10.3324/haematol.2018.212365] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
The endosteal bone marrow niche and vascular endothelial cells provide sanctuaries for leukemic cells. In murine chronic myeloid leukemia (CML) CD44 on leukemia cells and E-selectin on bone marrow endothelium are essential mediators for the engraftment of leukemic stem cells. We hypothesized that non-adhesion of CML-initiating cells to E-selectin on the bone marrow endothelium may lead to superior eradication of leukemic stem cells in CML after treatment with imatinib than imatinib alone. Indeed, here we show that treatment with the E-selectin inhibitor GMI-1271 in combination with imatinib prolongs survival of mice with CML via decreased contact time of leukemia cells with bone marrow endothelium. Non-adhesion of BCR-ABL1+ cells leads to an increase of cell cycle progression and an increase of expression of the hematopoietic transcription factor and proto-oncogene Scl/Tal1 in leukemia-initiating cells. We implicate SCL/TAL1 as an indirect phosphorylation target of BCR-ABL1 and as a negative transcriptional regulator of CD44 expression. We show that increased SCL/TAL1 expression is associated with improved outcome in human CML. These data demonstrate the BCR-ABL1-specific, cell-intrinsic pathways leading to altered interactions with the vascular niche via the modulation of adhesion molecules - which could be exploited therapeutically in the future.
Collapse
Affiliation(s)
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Stefanie C Herkt
- Institute for Transfusion Medicine DRK- Blutspendedienst Baden-Württemberg - Hessen, Frankfurt am Main, Germany
| | - Raquel S Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Nina Hayduk
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Eva S Weissenberger
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Djamel Aggoune
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Yosif Manavski
- Institute of Cardiovascular Regeneration, Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Tina Lucas
- Institute of Cardiovascular Regeneration, Center for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kuan-Ting Pan
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Jenna M Voutsinas
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Biostatistics, Seattle, WA, USA
| | - Qian Wu
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Biostatistics, Seattle, WA, USA
| | | | - Susanne Saussele
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Oellerich
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany.,German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany
| | - Vivian G Oehler
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Division of Hematology, University of Washington Medical Center, Seattle, WA, USA
| | - Joern Lausen
- Institute for Transfusion Medicine DRK- Blutspendedienst Baden-Württemberg - Hessen, Frankfurt am Main, Germany
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany .,German Cancer Research Center and German Cancer Consortium, Heidelberg, Germany.,Faculty of Medicine, Johann Wolfgang Goethe University, Frankfurt.,Frankfurt Cancer Institute, Frankfurt, Germany
| |
Collapse
|
682
|
Esler WP, Bence KK. Metabolic Targets in Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2019; 8:247-267. [PMID: 31004828 PMCID: PMC6698700 DOI: 10.1016/j.jcmgh.2019.04.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/18/2022]
Abstract
The prevalence and diagnosis of nonalcoholic fatty liver disease (NAFLD) is on the rise worldwide and currently has no FDA-approved pharmacotherapy. The increase in disease burden of NAFLD and a more severe form of this progressive liver disease, nonalcoholic steatohepatitis (NASH), largely mirrors the increase in obesity and type 2 diabetes (T2D) and reflects the hepatic manifestation of an altered metabolic state. Indeed, metabolic syndrome, defined as a constellation of obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension, is the major risk factor predisposing the NAFLD and NASH. There are multiple potential pharmacologic strategies to rebalance aspects of disordered metabolism in NAFLD. These include therapies aimed at reducing hepatic steatosis by directly modulating lipid metabolism within the liver, inhibiting fructose metabolism, altering delivery of free fatty acids from the adipose to the liver by targeting insulin resistance and/or adipose metabolism, modulating glycemia, and altering pleiotropic metabolic pathways simultaneously. Emerging data from human genetics also supports a role for metabolic drivers in NAFLD and risk for progression to NASH. In this review, we highlight the prominent metabolic drivers of NAFLD pathogenesis and discuss the major metabolic targets of NASH pharmacotherapy.
Collapse
Key Words
- acc, acetyl-coa carboxylase
- alt, alanine aminotransferase
- aso, anti-sense oligonucleotide
- ast, aspartate aminotransferase
- chrebp, carbohydrate response element binding protein
- ci, confidence interval
- dgat, diacylglycerol o-acyltransferase
- dnl, de novo lipogenesis
- fas, fatty acid synthase
- ffa, free fatty acid
- fgf, fibroblast growth factor
- fxr, farnesoid x receptor
- glp-1, glucagon-like peptide-1
- hdl, high-density lipoprotein
- homa-ir, homeostatic model assessment of insulin resistance
- ldl, low-density lipoprotein
- nafld, nonalcoholic fatty liver disease
- nas, nonalcoholic fatty liver disease activity score
- nash, nonalcoholic steatohepatitis
- or, odds ratio
- pdff, proton density fat fraction
- ppar, peroxisome proliferator-activated receptor
- sglt2, sodium glucose co-transporter 2
- srebp-1c, sterol regulatory element binding protein-1c
- t2d, type 2 diabetes
- t2dm, type 2 diabetes mellitus
- tg, triglyceride
- th, thyroid hormone
- thr, thyroid hormone receptor
- treg, regulatory t cells
- tzd, thiazolidinedione
- vldl, very low-density lipoprotein
Collapse
Affiliation(s)
- William P Esler
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts
| | - Kendra K Bence
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts.
| |
Collapse
|
683
|
Ruiu R, Tarone L, Rolih V, Barutello G, Bolli E, Riccardo F, Cavallo F, Conti L. Cancer stem cell immunology and immunotherapy: Harnessing the immune system against cancer's source. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:119-188. [PMID: 31383404 DOI: 10.1016/bs.pmbts.2019.03.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite recent advances in diagnosis and therapy having improved cancer outcome, many patients still do not respond to treatments, resulting in the progression or relapse of the disease, eventually impairing survival expectations. The limited efficacy of therapy is often attributable to its inability to affect cancer stem cells (CSCs), a small population of cells resistant to current radio- and chemo-therapies. CSCs are characterized by self-renewal and tumor-initiating capabilities, and function as a reservoir for the local and distant recurrence of the disease. Therefore, new therapeutic approaches able to effectively target and deplete CSCs are urgently needed. Immunotherapy is facing a renewed interest for its potential in cancer treatment, and the possibility of harnessing the immune system to target CSCs is being addressed by a new exciting research field. In this chapter, we discuss the cancer stem cell model and illustrate CSC biological and molecular properties, critically addressing theoretical and practical issues linked with their definition and study. We then review the existing literature regarding the immunological properties of CSCs and the complex interplay occurring between CSCs and immune cells. Finally, we present up-to-date studies on CSC immunotargeting and its potential future perspective. In conclusion, understanding the interplay between CSC biology and tumor immunology will provide a deeper understanding of the mechanisms that regulate CSC immunological properties. This will contribute to the design of new CSC-directed immunotherapeutic strategies with the potential of strongly improving cancer outcomes.
Collapse
Affiliation(s)
- Roberto Ruiu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Valeria Rolih
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| |
Collapse
|
684
|
Abstract
Cancer stem cells (CSCs) are crucial for tumor recurrence and distant metastasis. Immunologically targeting CSCs represents a promising strategy to improve efficacy of multimodal cancer therapy. Modulating the innate immune response involving Toll-like receptors, macrophages, natural killer cells, and γδT cells has therapeutic effects on CSCs. Antigens expressed by CSCs provide specific targets for immunotherapy. CSC-primed dendritic cell-based vaccines have induced significant antitumor immunity as an adjuvant therapy in experimental models of established tumors. Targeting the tumor microenvironment CSC niche with cytokines or checkpoint blockade provides additional strategies to eliminate CSCs.
Collapse
Affiliation(s)
- Jing Zhang
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, Room 3410, 1150 East Medical Center Drive, Ann Arbor, MI 48109, USA; Department of the 2nd Thoracic Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Road, Hongshan District, Wuhan, Hubei Province 430070, China
| | - Qiao Li
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, 3520B MSRB-1, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Alfred E Chang
- Division of Surgical Oncology, University of Michigan Rogel Cancer Center, Room 3304, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
685
|
Zhang J, Yuan B, Zhang H, Li H. Human epithelial ovarian cancer cells expressing CD105, CD44 and CD106 surface markers exhibit increased invasive capacity and drug resistance. Oncol Lett 2019; 17:5351-5360. [PMID: 31186752 PMCID: PMC6507388 DOI: 10.3892/ol.2019.10221] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/14/2019] [Indexed: 02/06/2023] Open
Abstract
The high rate of mortality associated with ovarian cancer (OC) is due in part to the development of resistance to chemotherapy, which allows the resistant tumour cells to invade and metastasise. Clarifying the mechanistic basis for drug resistance may reveal novel avenues for treatment. The present study investigated the mechanism of paclitaxel (PTX) resistance in human epithelial OC by evaluating the expression of stem cell-associated cell surface markers endoglin (CD105), CD44 antigen and vascular cell adhesion molecule 1 (CD106), in association with the malignant potential of the human OC OVCAR3 cell line and its PTX-resistant derivative OC3/TAX300. The expression of CD105, CD44 and CD106 was detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and flow cytometry, and cell invasion was evaluated using a Transwell invasion assay. CD105, CD44 and CD106 levels were increased in OC3/TAX300 cells compared with the OVCAR3 cells, as determined by flow cytometry (P<0.01) and RT-qPCR (P<0.05). Additionally, the number of invading cells was increased in the OC3/TAX300 group compared with the OVCAR3 group (54.7±6.65 vs. 31.8±6.55; P<0.01). A western blot analysis of cell surface marker expression in 80 clinical epithelial OC tissue samples, differing in terms of sensitivity to drug treatments, disease stage and degree of differentiation, revealed that high CD105, CD44 or CD106 expression was associated with drug resistance, advanced disease stage, poor differentiation and high rate of recurrence. These data indicated that exposure to high doses of PTX enhanced the stem-like properties of OC cells, which are associated with drug resistance and invasion and lead to poor prognosis due to induced chemoresistance and/or metastasis. Therefore, CD105, CD44 and CD106 may serve as potential stem cell-associated cell surface and prognostic markers, and therapeutic targets, in OC.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Baozhu Yuan
- Cell Collection and Research Centre, National Institutes for Food and Drug Control, Beijing 100050, P.R. China
| | - Huidan Zhang
- Department of Gynaecology, Beijing Maternal and Child Health Hospital of Haidian District, Beijing 100080, P.R. China
| | - Hongxia Li
- Department of Obstetrics and Gynaecology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| |
Collapse
|
686
|
Chen X, Ding X, Wu Q, Qi J, Zhu M, Miao C. Monomethyltransferase SET8 facilitates hepatocellular carcinoma growth by enhancing aerobic glycolysis. Cell Death Dis 2019; 10:312. [PMID: 30952833 PMCID: PMC6450876 DOI: 10.1038/s41419-019-1541-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/17/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive cancers worldwide. Despite such a public health importance, efficient therapeutic agents are still lacking for this malignancy. Most tumor cells use aerobic glycolysis to sustain anabolic growth, including HCC, and the preference of glycolysis often leads to a close association with poorer clinical outcomes. The histone methyltransferase SET8 plays crucial roles in controlling cell-cycle progression, transcription regulation, and tumorigenesis. However, it remains largely undefined whether SET8 affects the glucose metabolism in HCC. Here, we report that upregulation of SET8 is positively correlated with a poor survival rate in HCC patients. Both in vitro and in vivo studies revealed that SET8 deficiency conferred an impaired glucose metabolism phenotype and thus inhibited the progression of HCC tumors. By contrast, SET8 overexpression aggravated the glycolytic alterations and tumor progression. Mechanistically, SET8 directly binds to and inactivates KLF4, resulting in suppression of its downstream SIRT4. We also provided further evidence that mutations in SET8 failed to restrain the transactivation of SIRT4 by KLF4. Our data collectively uncover a novel mechanism of SET8 in mediating glycolytic metabolism in HCC cells and may provide a basis for targeting SET8 as a therapeutic strategy in HCC.
Collapse
Affiliation(s)
- Xiangyuan Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China
| | - Xiaowei Ding
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China
| | - Qichao Wu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China
| | - Jie Qi
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China
| | - Minmin Zhu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China.
| | - Changhong Miao
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China.
| |
Collapse
|
687
|
Sadeghi A, Roudi R, Mirzaei A, Zare Mirzaei A, Madjd Z, Abolhasani M. CD44 epithelial isoform inversely associates with invasive characteristics of colorectal cancer. Biomark Med 2019; 13:419-426. [PMID: 30942083 DOI: 10.2217/bmm-2018-0337] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: There is no consensus regarding the clinical significance of CD44 and CD24 as cancer stem cell (CSC) marker in colorectal cancer (CRC). Methodology: A total of 494 CRC samples (2008-2017) were assessed for CD44 (epithelial isoform) and CD24 expression using tissue microarray. Results: CD24 individually or in combination with CD44 was not associated with any of the clinicopathologic characteristics of the tumor. CD44 expression was inversely associated with pathological Tumor, Node, Metastasis (pTNM) lower stages (p = 0.038) and lymphatic invasion (p = 0.05). Conclusion: In summary, the epithelial isoform of CD44 is inversely associated with invasive characteristics of CRC.
Collapse
Affiliation(s)
- Asieh Sadeghi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Mirzaei
- Bone & Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zare Mirzaei
- Department of Pathology, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Hasheminejad Kidney Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
688
|
Roy Choudhury A, Gupta S, Chaturvedi PK, Kumar N, Pandey D. Mechanobiology of Cancer Stem Cells and Their Niche. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2019; 12:17-27. [PMID: 31004332 PMCID: PMC6529500 DOI: 10.1007/s12307-019-00222-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/10/2019] [Indexed: 01/25/2023]
Abstract
Though the existence of cancer stem cells remained enigmatic initially, over the time their participation in tumorigenesis and tumor progression has become highly evident. Today, they are also appreciated as the causal element for tumor heterogeneity and drug-resistance. Cancer stem cells activate a set of molecular pathways some of which are triggered by the unique mechanical properties of the tumor tissue stroma. A relatively new field called mechanobiology has emerged, which aims to critically evaluate the mechanical properties associated with biological events like tissue morphogenesis, cell-cell or cell-matrix interactions, cellular migration and also the development and progression of cancer. Development of more realistic model systems and biophysical instrumentation for observation and manipulation of cell-dynamics in real-time has invoked a hope for some novel therapeutic modalities against cancer in the future. This review discusses the fundamental concepts of cancer stem cells from an intriguing viewpoint of mechanobiology and some important breakthroughs to date.
Collapse
Affiliation(s)
- Ankit Roy Choudhury
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Surabhi Gupta
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Pradeep Kumar Chaturvedi
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Neeraj Kumar
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Deepak Pandey
- Department of Reproductive Biology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
689
|
Qi XT, Li YL, Zhang YQ, Xu T, Lu B, Fang L, Gao JQ, Yu LS, Zhu DF, Yang B, He QJ, Ying MD. KLF4 functions as an oncogene in promoting cancer stem cell-like characteristics in osteosarcoma cells. Acta Pharmacol Sin 2019; 40:546-555. [PMID: 29930276 DOI: 10.1038/s41401-018-0050-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/20/2018] [Indexed: 12/20/2022]
Abstract
Despite more effective chemotherapy combined with limb-salvage surgery for the osteosarcoma treatment, survival rates for osteosarcoma patients have stagnated over the past three decades due to the poor prognosis. Osteosarcoma cancer stem cells (OSCs) are responsible for the growth and metastasis of osteosarcoma. The existence of OSCs offers a theoretical explanation for therapeutic failures including tumor recurrence, metastasis, and drug resistance. Understanding the pathways that regulate properties of OSCs may shed light on mechanisms that lead to osteosarcoma and suggest better modes of treatment. In this study, we showed that the expression level of Kruppel-like factor 4 (KLF4) is highly associated with human osteosarcoma cancer stemness. KLF4-overexpressed osteosarcoma cells displayed characteristics of OSCs: increased sphere-forming potential, enhanced levels of stemness-associated genes, great chemoresistance to adriamycin and CDDP, as well as more metastasis potential. Inversely, KLF4 knockdown could reduce colony formation in vitro and inhibit tumorigenesis in vivo, supporting an oncogenic role for KLF4 in osteosarcoma pathogenesis. Furthermore, KLF4 was shown to activate the p38 MAPK signaling pathway to promote cancer stemness. Altogether, our studies uncover an essential role for KLF4 in regulation of OSCs and identify KLF4-p38 MAPK axis as a potential therapeutic target for osteosarcoma treatment.
Collapse
|
690
|
Shelygin YA, Obraztsov IV, Sukhina MA, Achkasov SI, Kashnikov VN, Sushkov OI, Sayfutdinova KR. IMMUNE PHENOTYPING OF FREE TUMOUR CELLS FOR EARLY DIAGNOSIS OF PERITONEAL CARCINOMATOSIS IN COLORECTAL CANCER. ACTA ACUST UNITED AC 2019. [DOI: 10.33878/2073-7556-2019-18-1-39-45] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
691
|
Temporal Splicing Switches in Elements of the TNF-Pathway Identified by Computational Analysis of Transcriptome Data for Human Cell Lines. Int J Mol Sci 2019; 20:ijms20051182. [PMID: 30857150 PMCID: PMC6429354 DOI: 10.3390/ijms20051182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 12/22/2022] Open
Abstract
Alternative splicing plays an important role in numerous cellular processes and aberrant splice decisions are associated with cancer. Although some studies point to a regulation of alternative splicing and its effector mechanisms in a time-dependent manner, the extent and consequences of such a regulation remains poorly understood. In the present work, we investigated the time-dependent production of isoforms in two Hodgkin lymphoma cell lines of different progression stages (HD-MY-Z, stage IIIb and L-1236, stage IV) compared to a B lymphoblastoid cell line (LCL-HO) with a focus on tumour necrosis factor (TNF) pathway-related elements. For this, we used newly generated time-course RNA-sequencing data from the mentioned cell lines and applied a computational pipeline to identify genes with isoform-switching behaviour in time. We analysed the temporal profiles of the identified events and evaluated in detail the potential functional implications of alterations in isoform expression for the selected top-switching genes. Our data indicate that elements within the TNF pathway undergo a time-dependent variation in isoform production with a putative impact on cell migration, proliferation and apoptosis. These include the genes TRAF1, TNFRSF12A and NFKB2. Our results point to a role of temporal alternative splicing in isoform production, which may alter the outcome of the TNF pathway and impact on tumorigenesis.
Collapse
|
692
|
Manzo G. Similarities Between Embryo Development and Cancer Process Suggest New Strategies for Research and Therapy of Tumors: A New Point of View. Front Cell Dev Biol 2019; 7:20. [PMID: 30899759 PMCID: PMC6416183 DOI: 10.3389/fcell.2019.00020] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/05/2019] [Indexed: 12/25/2022] Open
Abstract
Here, I propose that cancer stem cells (CSCs) would be equivalent to para-embryonic stem cells (p-ESCs), derived from adult cells de-re-programmed to a ground state. p-ESCs would differ from ESCs by the absence of genomic homeostasis. A p-ESC would constitute the cancer cell of origin (i-CSC or CSC0), capable of generating an initial tumor, corresponding to a pre-implantation blastocyst. In a niche with proper signals, it would engraft as a primary tumor, corresponding to a post-implantation blastocyst. i-CSC progeny would form primary pluripotent and slow self-renewing CSCs (CSC1s), blocked in an undifferentiated state, corresponding to epiblast cells; CSC1s would be tumor-initiating cells (TICs). CSC1s would generate secondary CSCs (CSC2s), corresponding to hypoblast cells; CSC2s would be tumor growth cells (TGCs). CSC1s/CSC2s would generate tertiary CSCs (CSC3s), with a mesenchymal phenotype; CSC3s would be tumor migrating cells (TMCs), corresponding to mesodermal precursors at primitive streak. CSC3s with more favorable conditions (normoxia), by asymmetrical division, would differentiate into cancer progenitor cells (CPCs), and these into cancer differentiated cells (CDCs), thus generating a defined cell hierarchy and tumor progression, mimicking somito-histo-organogenesis. CSC3s with less favorable conditions (hypoxia) would delaminate and migrate as quiescent circulating micro-metastases, mimicking mesenchymal cells in gastrula morphogenetic movements. In metastatic niches, these CSC3s would install and remain dormant in the presence of epithelial/mesenchymal transition (EMT) signals and hypoxia. But, in the presence of mesenchymal/epithelial transition (MET) signals and normoxia, they would revert to self-renewing CSC1s, reproducing the same cell hierarchy of the primary tumor as macro-metastases. Further similarities between ontogenesis and oncogenesis involving crucial factors, such as ID, HSP70, HLA-G, CD44, LIF, and STAT3, are strongly evident at molecular, physiological and immunological levels. Much experimental data about these factors led to considering the cancer process as ectopic rudimentary ontogenesis, where CSCs have privileged immunological conditions. These would consent to CSC development in an adverse environment, just like an embryo, which is tolerated, accepted and favored by the maternal organism in spite of its paternal semi-allogeneicity. From all these considerations, novel research directions, potential innovative tumor therapy and prophylaxis strategies might, theoretically, result.
Collapse
Affiliation(s)
- Giovanni Manzo
- General Pathology, “La Sapienza” University of Rome, Retired, Botrugno, Italy
| |
Collapse
|
693
|
Liskova A, Kubatka P, Samec M, Zubor P, Mlyncek M, Bielik T, Samuel SM, Zulli A, Kwon TK, Büsselberg D. Dietary Phytochemicals Targeting Cancer Stem Cells. Molecules 2019; 24:molecules24050899. [PMID: 30836718 PMCID: PMC6429493 DOI: 10.3390/molecules24050899] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
There is an increasing awareness of the importance of a diet rich in fruits and vegetables for human health. Cancer stem cells (CSCs) are characterized as a subpopulation of cancer cells with aberrant regulation of self-renewal, proliferation or apoptosis leading to cancer progression, invasiveness, metastasis formation, and therapy resistance. Anticancer effects of phytochemicals are also directed to target CSCs. Here we provide a comprehensive review of dietary phytochemicals targeting CSCs. Moreover, we evaluate and summarize studies dealing with effects of dietary phytochemicals on CSCs of various malignancies in preclinical and clinical research. Dietary phytochemicals have a significant impact on CSCs which may be applied in cancer prevention and treatment. However, anticancer effects of plant derived compounds have not yet been fully investigated in clinical research.
Collapse
Affiliation(s)
- Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Pavol Zubor
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Milos Mlyncek
- Department of Obstetrics and Gynecology Faculty Hospital Nitra Constantine the Philosopher University, 949 01 Nitra, Slovakia.
| | - Tibor Bielik
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, 03601 Bratislava, Slovakia.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha 24144, Qatar.
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011, Australia.
| | - Taeg Kyu Kwon
- Department of Immunology and School of Medicine, Keimyung University, Dalseo-Gu, Daegu 426 01, Korea.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha 24144, Qatar.
| |
Collapse
|
694
|
Wang CY, Huang CS, Yang YP, Liu CY, Liu YY, Wu WW, Lu KH, Chen KH, Chang YL, Lee SD, Lin HC. The subpopulation of CD44-positive cells promoted tumorigenicity and metastatic ability in lung adenocarcinoma. J Chin Med Assoc 2019; 82:196-201. [PMID: 30908413 DOI: 10.1097/jcma.0000000000000056] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Lung cancer is one of the major causes of carcinoma-related deaths in the world. Importantly, lung adenocarcinoma (LAC) is the most common type with poor outcome. However, the progressive clinical phenotype and biomolecular signature of lung cancer presenting the cancer stem-like and metastatic characteristics are still unclear. METHODS In this study, we identified CD44 marker in lung cancers. The capabilities, including tumorigenic and migration assays, were analyzed in CD44 expression and CD44 expression subgroups. Meanwhile, the potential bio-signature and properties of lung tumor stem-like cells were further studied. RESULTS The high expression of CD44 subpopulation (CD44-positive) in isolated lung cancer cells showed significantly higher abilities of tumorigenic colonies, tumor-sphere formation, and migratory properties when compared with the CD44 expression group. These subgroups of CD44-positive lung cancer cells further demonstrated the metastatic potential with epithelial-mesenchymal transition (EMT), as well as the high expression of Twist and Snail gene profile. Importantly, the overexpression of Snail with gene vector in CD44 expression cells further significantly promoted the properties of lung tumor stem-like cells. CONCLUSION The results of this study highlighted the role of CD44-posivite subpopulation in modulating tumor initiation and EMT-based metastatic ability of lung malignancy.
Collapse
Affiliation(s)
- Chien-Ying Wang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Trauma, Emergency Department Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chi-Shuan Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Colorectal Surgery, Department of Surgery, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Chao-Yu Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Thoracic Surgery, Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Yung-Yang Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Chest, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Wai-Wah Wu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Gastroenterology, Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Kai-Hsi Lu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Kuan-Hsuan Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yuh-Lih Chang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Shou-Dong Lee
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Gastroenterology, Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Chi Lin
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Gastroenterology, Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
695
|
Tang RZ, Zhu JJ, Yang FF, Zhang YP, Xie SA, Liu YF, Yao WJ, Pang W, Han LL, Kong W, Wang YX, Zhang T, Zhou J. DNA methyltransferase 1 and Krüppel-like factor 4 axis regulates macrophage inflammation and atherosclerosis. J Mol Cell Cardiol 2019; 128:11-24. [DOI: 10.1016/j.yjmcc.2019.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/20/2018] [Accepted: 01/13/2019] [Indexed: 12/17/2022]
|
696
|
Wu DM, Wen X, Han XR, Wang S, Wang YJ, Shen M, Fan SH, Zhang ZF, Shan Q, Li MQ, Hu B, Lu J, Chen GQ, Zheng YL. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal MicroRNA-126-3p Inhibits Pancreatic Cancer Development by Targeting ADAM9. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:229-245. [PMID: 30925451 PMCID: PMC6439275 DOI: 10.1016/j.omtn.2019.02.022] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/30/2019] [Accepted: 02/23/2019] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is a lethal malignancy with relatively few effective therapies. Recent investigations have highlighted the role of microRNAs (miRNAs) as crucial regulators in various tumor processes including tumor progression. Hence the current study aimed to investigate the role of bone marrow mesenchymal stem cell (BMSC)-derived exosomal microRNA-126-3p (miR-126-3p) in pancreatic cancer. Initially, miRNA candidates and related genes associated with pancreatic cancer were screened. PANC-1 cells were transfected with miR-126-3p or silenced a disintegrin and a metalloproteinase-9 (ADAM9) to examine their regulatory roles in pancreatic cancer cells. Additionally, exosomes derived from BMSCs were isolated and co-cultured with pancreatic cancer cells to elucidate the effects of exosomes in pancreatic cancer. Furthermore, the effects of overexpressed miR-126-3p derived from BMSCs exosomes on proliferation, migration, invasion, apoptosis, tumor growth, and metastasis of pancreatic cancer cells were analyzed in connection with lentiviral packaged miR-126-3p in vivo. Restored miR-126-3p was observed to suppress pancreatic cancer through downregulating ADAM9. Notably, overexpressed miR-126-3p derived from BMSCs exosomes inhibited the proliferation, invasion, and metastasis of pancreatic cancer cells, and promoted their apoptosis both in vitro and in vivo. Taken together, the key findings of the study indicated that overexpressed miR-126-3p derived from BMSCs exosomes inhibited the development of pancreatic cancer through the downregulation of ADAM9, highlighting the potential of miR-126-3p as a novel biomarker for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| | - Gui-Quan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing 210061, Jiangsu, China.
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, China; College of Health Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| |
Collapse
|
697
|
Park CS, Lewis A, Chen T, Lacorazza D. Concise Review: Regulation of Self-Renewal in Normal and Malignant Hematopoietic Stem Cells by Krüppel-Like Factor 4. Stem Cells Transl Med 2019; 8:568-574. [PMID: 30790473 PMCID: PMC6525558 DOI: 10.1002/sctm.18-0249] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
Pluripotent and tissue‐specific stem cells, such as blood‐forming stem cells, are maintained through a balance of quiescence, self‐renewal, and differentiation. Self‐renewal is a specialized cell division that generates daughter cells with the same features as the parental stem cell. Although many factors are involved in the regulation of self‐renewal, perhaps the most well‐known factors are members of the Krüppel‐like factor (KLF) family, especially KLF4, because of the landmark discovery that this protein is required to reprogram somatic cells into induced pluripotent stem cells. Because KLF4 regulates gene expression through transcriptional activation or repression via either DNA binding or protein‐to‐protein interactions, the outcome of KLF4‐mediated regulation largely depends on the cellular context, cell cycle regulation, chromatin structure, and the presence of oncogenic drivers. This study first summarizes the current understanding of the regulation of self‐renewal by KLF proteins in embryonic stem cells through a KLF circuitry and then delves into the potential function of KLF4 in normal hematopoietic stem cells and its emerging role in leukemia‐initiating cells from pediatric patients with T‐cell acute lymphoblastic leukemia via repression of the mitogen‐activated protein kinase 7 pathway. stem cells translational medicine2019;8:568–574
Collapse
Affiliation(s)
- Chun S Park
- Department Pathology & Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Andrew Lewis
- Department Pathology & Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Taylor Chen
- Department Pathology & Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Daniel Lacorazza
- Department Pathology & Immunology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
698
|
Han S, Huang T, Li W, Wang X, Wu X, Liu S, Yang W, Shi Q, Li H, Hou F. Prognostic Value of CD44 and Its Isoforms in Advanced Cancer: A Systematic Meta-Analysis With Trial Sequential Analysis. Front Oncol 2019; 9:39. [PMID: 30788285 PMCID: PMC6372530 DOI: 10.3389/fonc.2019.00039] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022] Open
Abstract
Objective: Cancer stem cell marker CD44 and its variant isoforms (CD44v) may be correlated with tumor growth, metastasis, and chemo-radiotherapy resistance. However, the prognostic power of CD44 and CD44v in advanced cancer remains controversial. Therefore, the purpose of our study was to generalize the prognostic significance of these cancer stem cell markers in advanced cancer patients. Methods: Hazard ratios (HRs) with 95% confidence intervals (95% CIs) were calculated from multivariable analysis to assess the associations among CD44, CD44v6, and CD44v9 positivity and overall survival (OS), disease-free survival (DFS), progression-free survival (PFS), cancer-specific survival (CSS), and recurrence-free survival (RFS). Trial sequential analysis (TSA) was also conducted. Results: We included 15 articles that reported on 1,201 patients with advanced cancer (CD44: nine studies with 796 cases, CD44v6: three studies with 143 cases, and CD44v9: three studies with 262 cases). CD44 expression was slightly linked to worse OS (HR = 2.03, P = 0.027), but there was no correlation between CD44 expression and DFS, RFS, or PFS. Stratified analysis showed that CD44 expression was not correlated with OS at ≥5 years or OS in patients receiving adjuvant therapy. CD44v6 expression was not associated with OS. CD44v9 expression was closely associated with poor 5-years CSS in patients treated with chemo/radiotherapy (HR = 3.62, P < 0.001). However, TSA suggested that additional trials were needed to confirm these conclusions. Conclusions: CD44 or CD44v9 might be novel therapeutic targets for improving the treatment of advanced cancer patients. Additional prospective clinical trials are strongly needed across different cancer types.
Collapse
Affiliation(s)
- Susu Han
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Huang
- The Affiliated Hospital of Ningbo University, Ningbo, China
| | - Wen Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiyu Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xing Wu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shanshan Liu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Yang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Shi
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongjia Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenggang Hou
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
699
|
Kim SH, Singh SV. Role of Krüppel-like Factor 4-p21 CIP1 Axis in Breast Cancer Stem-like Cell Inhibition by Benzyl Isothiocyanate. Cancer Prev Res (Phila) 2019; 12:125-134. [PMID: 30723175 DOI: 10.1158/1940-6207.capr-18-0393] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/11/2018] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
Abstract
Cancer chemoprevention by benzyl isothiocyanate (BITC), which is derived from cruciferous vegetables like garden cress, in a transgenic mouse model of breast cancer is associated with inhibition of breast cancer stem-like cells (bCSC), but the molecular regulators of this effect remain elusive. This study demonstrates a protective effect of Krüppel-like factor 4 (KLF4)-p21CIP1 axis in bCSC inhibition by BITC. Exposure of human breast cancer cells (MCF-7, MDA-MB-231, and SUM159) to plasma-achievable concentrations of BITC resulted in a robust induction of KLF4 mRNA and its protein expression as determined by qRT-PCR and Western blotting or confocal microscopy. BITC-mediated suppression of bCSC markers, including aldehyde dehydrogenase 1 activity and mammosphere frequency, was significantly augmented by transient or stable knockdown of KLF4. Western blotting and IHC revealed relatively higher levels of KLF4 protein in mammary tumor sections from BITC-treated mice in comparison with controls, but the difference was insignificant. Analysis of the breast cancer RNA-Seq data from The Cancer Genome Atlas indicated significant positive correlation between expression of KLF4 and that of p21CIP1 (CDKN1A) but not β-Catenin (CTNNB1). Knockdown of p21CIP1 protein also amplified BITC-mediated suppression of bCSC. Finally, KLF4 was recruited to the promoter of p21CIP1 as indicated by chromatin immunoprecipitation assay. These results indicate that induction of KLF4-p21CIP1 axis attenuates inhibitory effect of BITC on bCSC self-renewal. Translational implication of these findings is that breast cancer chemoprevention by BITC may be augmented with a combination regimen involving BITC and an inhibitor of KLF4.
Collapse
Affiliation(s)
- Su-Hyeong Kim
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shivendra V Singh
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
700
|
Abdel-Salam IM, Awadein NES, Ashour M. Cytotoxicity of Luffa cylindrica (L.) M.Roem. extract against circulating cancer stem cells in hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2019; 229:89-96. [PMID: 30287196 DOI: 10.1016/j.jep.2018.09.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/22/2018] [Accepted: 09/26/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Luffa cylindrica (L.) M.Roem. has been recognized as traditional medicine for the treatment of various diseases like inflammatory diseases, diarrhea and viral infections. The usual parts used include fruit, seeds and leaves. AIM OF THE STUDY To evaluate the anticancer activity of the hot water extract of the whole plant of Luffa cylindrica using circulating tumor cells and cancer stem cells isolated from the peripheral blood of hepatocellular carcinoma patients in vitro. MATERIALS AND METHODS Seventy five adult patients who reported as stage II and III Hepatocellular carcinoma were selected. Blood samples were withdrawn and the circulating tumor cells were isolated from the whole blood. Tumor cells and cancer stem cells were detected and isolated by flow cytometric techniques. The isolated cell types were cultured and propagated in the tissue culture facility, the extract was tested on the isolated cells. RESULTS Luffa cylindrica hot water extract has shown cytotoxic activity against circulating tumor cells of hepatocellular carcinoma especially the cells sub-population CD133+/CD44+ with little effect among CD133+/CD44- sub-population. CONCLUSION Hot water extract of Luffa cylindrica whole plant could decrease the ratio of cancer stem cells in blood of HCC patients and may be used to minimize recurrence and metastasis in hepatocellular carcinoma patients.
Collapse
Affiliation(s)
| | | | - Mohamed Ashour
- Medical Research Department, National Institute of Occupational Safety and Health, Egypt, 156 - EL Hegas Street, Cairo, Egypt.
| |
Collapse
|