1
|
Kzhyshkowska J, Shen J, Larionova I. Targeting of TAMs: can we be more clever than cancer cells? Cell Mol Immunol 2024:10.1038/s41423-024-01232-z. [PMID: 39516356 DOI: 10.1038/s41423-024-01232-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
АBSTRACT: With increasing incidence and geography, cancer is one of the leading causes of death, reduced quality of life and disability worldwide. Principal progress in the development of new anticancer therapies, in improving the efficiency of immunotherapeutic tools, and in the personification of conventional therapies needs to consider cancer-specific and patient-specific programming of innate immunity. Intratumoral TAMs and their precursors, resident macrophages and monocytes, are principal regulators of tumor progression and therapy resistance. Our review summarizes the accumulated evidence for the subpopulations of TAMs and their increasing number of biomarkers, indicating their predictive value for the clinical parameters of carcinogenesis and therapy resistance, with a focus on solid cancers of non-infectious etiology. We present the state-of-the-art knowledge about the tumor-supporting functions of TAMs at all stages of tumor progression and highlight biomarkers, recently identified by single-cell and spatial analytical methods, that discriminate between tumor-promoting and tumor-inhibiting TAMs, where both subtypes express a combination of prototype M1 and M2 genes. Our review focuses on novel mechanisms involved in the crosstalk among epigenetic, signaling, transcriptional and metabolic pathways in TAMs. Particular attention has been given to the recently identified link between cancer cell metabolism and the epigenetic programming of TAMs by histone lactylation, which can be responsible for the unlimited protumoral programming of TAMs. Finally, we explain how TAMs interfere with currently used anticancer therapeutics and summarize the most advanced data from clinical trials, which we divide into four categories: inhibition of TAM survival and differentiation, inhibition of monocyte/TAM recruitment into tumors, functional reprogramming of TAMs, and genetic enhancement of macrophages.
Collapse
Affiliation(s)
- Julia Kzhyshkowska
- Department of Innate Immunity and Tolerance, Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer, 1-3, 68167, Mannheim, Germany.
- German Red Cross Blood Service Baden-Württemberg - Hessen, Friedrich-Ebert Str. 107, 68167, Mannheim, Germany.
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050, Lenina av.36, Tomsk, Russia.
- Bashkir State Medical University of the Ministry of Health of Russia, 450000, Teatralnaya Street, 2a, Ufa, Russia.
| | - Jiaxin Shen
- Department of Innate Immunity and Tolerance, Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer, 1-3, 68167, Mannheim, Germany
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050, Lenina av.36, Tomsk, Russia
- Bashkir State Medical University of the Ministry of Health of Russia, 450000, Teatralnaya Street, 2a, Ufa, Russia
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009, Kooperativnyi st, Tomsk, Russia
| |
Collapse
|
2
|
Chen H, Zhang W, Shi J, Tang Y, Chen X, Li J, Yao X. Study on the mechanism of S100A4-mediated cancer oncogenesis in uveal melanoma cells through the integration of bioinformatics and in vitro experiments. Gene 2024; 911:148333. [PMID: 38431233 DOI: 10.1016/j.gene.2024.148333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/13/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND The elevated metastasis rate of uveal melanoma (UM) is intricately correlated with patient prognosis, significantly affecting the quality of life. S100 calcium-binding protein A4 (S100A4) has tumorigenic properties; therefore, the present study investigated the impact of S100A4 on UM cell proliferation, apoptosis, migration, and invasion using bioinformatics and in vitro experiments. METHODS Bioinformatic analysis was used to screen S100A4 as a hub gene and predict its possible mechanism in UM cells, and the S100A4 silencing cell line was constructed. The impact of S100A4 silencing on the proliferative ability of UM cells was detected using the Cell Counting Kit-8 and colony formation assays. Annexin V-FITC/PI double fluorescence and Hoechst 33342 staining were used to observe the effects of apoptosis on UM cells. The effect of S100A4 silencing on the migratory and invasive capabilities of UM cells was assessed using wound healing and Transwell assays. Western blotting was used to detect the expression of related proteins. RESULTS The present study found that S100A4 is a biomarker of UM, and its high expression is related to poor prognosis. After constructing the S100A4 silencing cell line, cell viability, clone number, proliferating cell nuclear antigen, X-linked inhibitor of apoptosis protein, and survivin expression were decreased in UM cells. The cell apoptosis rate and relative fluorescence intensity increased, accompanied by increased levels of Bax and caspase-3 and decreased levels of Bcl-2. Additionally, a decrease in the cell migration index and relative invasion rate was observed with increased E-cadherin expression and decreased N-cadherin and vimentin protein expression. CONCLUSION S100A4 silencing can inhibit the proliferation, migration, and invasion and synchronously induces apoptosis in UM cells.
Collapse
Affiliation(s)
- Huimei Chen
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Wenqing Zhang
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Jian Shi
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Yu Tang
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiong Chen
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Jiangwei Li
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiaolei Yao
- The First Clinical College of Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Hunan Provincial Key Laboratory for the Prevention and Treatment of Ophthalmology and Otolaryngology Diseases with Traditional Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
3
|
Campanelli G, Deabel RA, Puaar A, Devarakonda LS, Parupathi P, Zhang J, Waxner N, Yang C, Kumar A, Levenson AS. Molecular Efficacy of Gnetin C as Dual-Targeted Therapy for Castrate-Resistant Prostate Cancer. Mol Nutr Food Res 2023; 67:e2300479. [PMID: 37863824 DOI: 10.1002/mnfr.202300479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/29/2023] [Indexed: 10/22/2023]
Abstract
SCOPE Resistance of castrate-resistant prostate cancer (CRPC) to enzalutamide (Enz) involves the expression of constitutively active androgen receptor splice variant (AR-V7). In addition to altered AR pathways, CRPC is characterized by "non-AR-driven" signaling, which includes an overexpression of metastasis-associated protein 1 (MTA1). Combining natural compounds with anticancer drugs may enhance drug effectiveness while reducing adverse effects. In this study, the in vitro and in vivo anticancer effects of Gnetin C (GnC) alone and in combination with Enz against CRPC are examined. METHODS AND RESULTS The effects of GnC alone and in combination with Enz are assessed by cell viability, clonogenic survival, cell migration, and AR and MTA1 expression using 22Rv1 cells. The tumor growth in vivo is assessed by bioluminescent imaging, western blots, RT-PCR, and IHC. GnC alone and in combined treatment inhibit cell viability, clonogenic survival and migration, and AR and MTA1 expression in 22Rv1 cells. The underlying AR- and MTA1-targeted anticancer mechanisms of treatments in vivo involve inhibition of proliferation and angiogenesis, and induction of apoptosis. CONCLUSION The findings demonstrate that GnC alone and GnC combined with Enz effectively inhibits AR- and MTA1-promoted tumor-progression in advanced CRPC, which indicates its potential as a novel therapeutic approach for CRPC.
Collapse
Affiliation(s)
- Gisella Campanelli
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Rabab Al Deabel
- School of Health Professions and Nursing, Long Island University, Brookville, NY, USA
| | - Anand Puaar
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | | | - Prashanth Parupathi
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | | | - Noah Waxner
- College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | - Ching Yang
- College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| | - Avinash Kumar
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | - Anait S Levenson
- College of Veterinary Medicine, Long Island University, Brookville, NY, USA
| |
Collapse
|
4
|
Wei D, Niu B, Zhai B, Liu XB, Yao YL, Liang CC, Wang P. Expression profiles and function prediction of tRNA-derived fragments in glioma. BMC Cancer 2023; 23:1015. [PMID: 37864150 PMCID: PMC10588164 DOI: 10.1186/s12885-023-11532-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive malignant primary brain tumor. The transfer RNA-derived fragments (tRFs) are a new group of small noncoding RNAs, which are dysregulated in many cancers. Until now, the expression and function of tRFs in glioma remain unknown. METHODS The expression profiles of tRF subtypes were analyzed using the Cancer Genome Atlas (TCGA)-low-grade gliomas (LGG)/GBM dataset. The target genes of tRFs were subjected to Gene Ontology, Kyoto Encyclopedia and Gene set enrichment analysis of Genes and Genomes pathway enrichment analysis. The protein-protein interaction enrichment analysis was performed by STRING. QRT-PCR was performed to detect the expressions of tRFs in human glioma cell lines U87, U373, U251, and human astrocyte cell line SVG p12. Western blot assay was used to detect to the expression of S100A11. The interaction between tRF-19-R118LOJX and S100A11 mRNA 3'UTR was detected by dual-luciferase reporter assay. The effects of tRF-19-R118LOJX, tRF-19-6SM83OJX and S100A11 on the glioma cell proliferation, migration and in vitro vasculogenic mimicry formation ability were examined by CCK-8 proliferation assay, EdU assay, HoloMonitor cell migration assay and tube formation assay, respectively. RESULTS tRF-19-R118LOJX and tRF-19-6SM83OJX are the most differentially expressed tRFs between LGG and GBM groups. The functional enrichment analysis showed that the target genes of tRF-19-R118LOJX and tRF-19-6SM83OJX are enriched in regulating blood vessel development. The upregulated target genes are linked to adverse survival outcomes in glioma patients. tRF-19-R118LOJX and tRF-19-6SM83OJX were identified to suppress glioma cell proliferation, migration, and in vitro vasculogenic mimicry formation. The mechanism of tRF-19-R118LOJX might be related to its function as an RNA silencer by targeting the S100A11 mRNA 3'UTR. CONCLUSION tRFs would become novel diagnostic biomarkers and therapeutic targets of glioma, and the mechanism might be related to its post-transcriptionally regulation of gene expression by targeting mRNA 3'UTR.
Collapse
Affiliation(s)
- Deng Wei
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Ben Niu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Bei Zhai
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Xiao-Bai Liu
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yi-Long Yao
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Chan-Chan Liang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Ping Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China.
| |
Collapse
|
5
|
Niu B, Wei D, Liu XY, Zhai B, Liu XB, Yao YL, Xue YX, Wang P. CircMTA1 promotes glioblastoma angiogenesis by encoding MTA1-134aa. FASEB J 2023; 37:e23160. [PMID: 37750502 DOI: 10.1096/fj.202300724r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/08/2023] [Accepted: 08/11/2023] [Indexed: 09/27/2023]
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor with rapid angiogenesis. How to inhibit GBM angiogenesis is a key problem to be solved. To explore the targets of inhibiting GBM angiogenesis, this study confirmed that the expression of circMTA1 (hsa_circ_0033614) was significantly upregulated in human brain microvascular endothelial cells exposed to glioma cell-conditioned medium (GECs). The expression of circMTA1 in the cytoplasm was significantly higher than that in the nucleus. Upregulated circMTA1 in GECs can promote cell proliferation, migration, and tube formation. Further exploration of the circularization mechanism of circMTA1 confirmed that KHDRBS1 protein can bind to the upstream and downstream flanking sequences of circMTA1 and promote circMTA1 biogenesis by coordinating Alu element pairing. KHDRBS1 upregulated the proliferation, migration, and tube formation of GECs by promoting the biogenesis of circMTA1. CircMTA1 can encode the protein MTA1-134aa by internal ribosome entry site sequence-mediated translation mechanism, and promote the proliferation, migration, and tube formation of GECs through the encoded MTA1-134aa. This study provides a new target for inhibiting angiogenesis in brain GBM and a new strategy for improving the therapeutic efficacy of GBM.
Collapse
Affiliation(s)
- Ben Niu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Deng Wei
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Xiao-Yu Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Bei Zhai
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Xiao-Bai Liu
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Long Yao
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi-Xue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| | - Ping Wang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
- Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang, China
| |
Collapse
|
6
|
Pan R, He T, Zhang K, Zhu L, Lin J, Chen P, Liu X, Huang H, Zhou D, Li W, Yang S, Ye G. Tumor-Targeting Extracellular Vesicles Loaded with siS100A4 for Suppressing Postoperative Breast Cancer Metastasis. Cell Mol Bioeng 2023; 16:117-125. [PMID: 37096069 PMCID: PMC10121989 DOI: 10.1007/s12195-022-00757-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/14/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction S100A4 promotes the establishment of tumor microenvironment for malignant cancer cells, and knockdown of S100A4 can inhibit tumorigenesis. However, there is no efficient way to target S100A4 in metastatic tumor tissues. Here, we investigated the role of siS100A4-loaded iRGD-modified extracellular vesicles (siS100A4-iRGD-EVs) in postoperative breast cancer metastasis. Methods siS100A4-iRGD-EVs nanoparticles were engineered and analyzed using TEM and DLS. siRNA protection, cellular uptake, and cytotoxicity of EV nanoparticles were examined in vitro. Postoperative lung metastasis mouse model was created to investigate the tissue distribution and anti-metastasis roles of nanoparticles in vivo. Results siS100A4-iRGD-EVs protected siRNA from RNase degradation, enhanced the cellular uptake and compatibility in vitro. Strikingly, iRGD-modified EVs significantly increased tumor organotropism and siRNA accumulation in lung PMNs compared to siS100A4-EVs in vivo. Moreover, siS100A4-iRGD-EVs treatment remarkedly attenuated lung metastases from breast cancer and increased survival rate of mice through suppressing S100A4 expression in lung. Conclusions siS100A4-iRGD-EVs nanoparticles show more potent anti-metastasis effect in postoperative breast cancer metastasis mouse model. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00757-5.
Collapse
Affiliation(s)
- Ruiling Pan
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Tiancheng He
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Kun Zhang
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Lewei Zhu
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Jiawei Lin
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Peixian Chen
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Xiangwei Liu
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Huiqi Huang
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Dan Zhou
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Wei Li
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Shuqing Yang
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| | - Guolin Ye
- Department of Breast Surgery, The First People’s Hospital of Foshan, No. 81 North Lingnan Avenue, Chancheng, Foshan, 528000 Guangdong China
| |
Collapse
|
7
|
A Simplified and Effective Approach for the Isolation of Small Pluripotent Stem Cells Derived from Human Peripheral Blood. Biomedicines 2023; 11:biomedicines11030787. [PMID: 36979766 PMCID: PMC10045871 DOI: 10.3390/biomedicines11030787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Pluripotent stem cells are key players in regenerative medicine. Embryonic pluripotent stem cells, despite their significant advantages, are associated with limitations such as their inadequate availability and the ethical dilemmas in their isolation and clinical use. The discovery of very small embryonic-like (VSEL) stem cells addressed the aforementioned limitations, but their isolation technique remains a challenge due to their small cell size and their efficiency in isolation. Here, we report a simplified and effective approach for the isolation of small pluripotent stem cells derived from human peripheral blood. Our approach results in a high yield of small blood stem cell (SBSC) population, which expresses pluripotent embryonic markers (e.g., Nanog, SSEA-3) and the Yamanaka factors. Further, a fraction of SBSCs also co-express hematopoietic markers (e.g., CD45 and CD90) and/or mesenchymal markers (e.g., CD29, CD105 and PTH1R), suggesting a mixed stem cell population. Finally, quantitative proteomic profiling reveals that SBSCs contain various stem cell markers (CD9, ITGA6, MAPK1, MTHFD1, STAT3, HSPB1, HSPA4), and Transcription reg complex factors (e.g., STAT5B, PDLIM1, ANXA2, ATF6, CAMK1). In conclusion, we present a novel, simplified and effective isolating process that yields an abundant population of small-sized cells with characteristics of pluripotency from human peripheral blood.
Collapse
|
8
|
Wang W, Ma M, Li L, Huang Y, Zhao G, Zhou Y, Yang Y, Yang Y, Wang B, Ye L. MTA1-TJP1 interaction and its involvement in non-small cell lung cancer metastasis. Transl Oncol 2022; 25:101500. [PMID: 35944414 PMCID: PMC9365954 DOI: 10.1016/j.tranon.2022.101500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
MTA1 was highly expressed in NSCLC tissues and was associated with tumor progression. MTA1 promoted NSCLC cell invasion and migration in vitro and in vivo. TJP1 was found to be an interacting protein of MTA1 involved in cell adhesion. MTA1 promoted NSCLC invasion and metastasis by inhibiting TJP1 protein expression and attenuating intercellular tight junctions. Targeting the MTA1-TJP1 axis may be a promising strategy for inhibiting NSCLC metastasis.
Distant metastasis is the main cause of death in non-small cell lung cancer (NSCLC) patients. The mechanism of metastasis-associated protein 1(MTA1) in NSCLC has not been fully elucidated. This study aimed to reveal the mechanism of MTA1 in the invasion and metastasis of NSCLC. Bioinformatics analysis and our previous results showed that MTA1 was highly expressed in NSCLC tissues and correlated with tumor progression. Knockout of MTA1 by CRISPR/Cas9 significantly inhibited the migration and invasion of H1299 cells, but enhanced cell adhesion. Stable overexpression of MTA1 by lentivirus transfection had opposite effects on migration, invasion and adhesion of A549 cells. The results of in vivo experiments in nude mouse lung metastases model confirmed the promotion of MTA1 on invasion and migration. Tight junction protein 1 (TJP1) was identified by immunoprecipitation and mass spectrometry as an interacting protein of MTA1 involved in cell adhesion. MTA1 inhibited the expression level of TJP1 protein and weakened the tight junctions between cells. More importantly, the rescue assays confirmed that the regulation of MTA1 on cell adhesion, migration and invasion was partially attenuated by TJP1. In Conclusion, MTA1 inhibits the expression level of TJP1 protein co-localized in the cytoplasm and membrane of NSCLC cells, weakens the tight junctions between cells, and changes the adhesion, migration and invasion capabilities of cells, which may be the mechanism of MTA1 promoting the invasion and metastasis of NSCLC. Thus, targeting the MTA1-TJP1 axis may be a promising strategy for inhibiting NSCLC metastasis.
Collapse
Affiliation(s)
- Wei Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, China; Department of Thoracic Surgery, Taihe Hospital (Hubei University of Medicine), Shiyan, China
| | - Mingsheng Ma
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China
| | - Li Li
- Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunchao Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, China
| | - Guangqiang Zhao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, China
| | - Yongchun Zhou
- Molecular Diagnosis Center, Yunnan Cancer Hospital, Kunming, China
| | - Yantao Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, China
| | - Yichen Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, China
| | - Biying Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, China
| | - Lianhua Ye
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, China.
| |
Collapse
|
9
|
Vattem C, Pakala SB. Metastasis-associated protein 1: A potential driver and regulator of the hallmarks of cancer. J Biosci 2022. [DOI: 10.1007/s12038-022-00263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
10
|
Ma J, Li C, Qian H, Zhang Y. MTA1: A Vital Modulator in Prostate Cancer. Curr Protein Pept Sci 2022; 23:456-464. [PMID: 35792131 DOI: 10.2174/1389203723666220705152713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) is the most frequent cancer of the male genitourinary system and the second most common cancer in men worldwide. PCa has become one of the leading diseases endangering men's health in Asia in recent years, with a large increase in morbidity and mortality. MTA1 (metastasis-associated antigen-1), a transcriptional coregulator involved in histone deacetylation and nucleosome remodeling, is a member of the MTA family. MTA1 is involved in cell signaling, chromosomal remodeling, and transcriptional activities, all of which are important for epithelial cell progression, invasion, and growth. MTA1 has been demonstrated to play a significant role in the formation, progression, and metastasis of PCa, and MTA1 expression is specifically linked to PCa bone metastases. Therefore, MTA1 may be a potential target for PCa prevention and treatment. Here, we reviewed the structure, function, and expression of MTA1 in PCa as well as drugs that target MTA1 to highlight a potential new treatment for PCa.
Collapse
Affiliation(s)
- Jialu Ma
- Graduate School of Hebei Medical University, Shijiazhuang, China
| | - Chunxiao Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Zhang
- Graduate School of Hebei Medical University, Shijiazhuang, China
- Department of Urology Surgery, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Ishikawa M, Osaki M, Uno N, Ohira T, Kugoh H, Okada F. MTA1, a metastasis‑associated protein, in endothelial cells is an essential molecule for angiogenesis. Mol Med Rep 2021; 25:11. [PMID: 34779499 PMCID: PMC8600423 DOI: 10.3892/mmr.2021.12527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/14/2021] [Indexed: 11/12/2022] Open
Abstract
Our previous study revealed that metastasis-associated protein 1 (MTA1), which is expressed in vascular endothelial cells, acts as a tube formation promoting factor. The present study aimed to clarify the importance of MTA1 expression in tube formation using MTA1-knockout (KO) endothelial cells (MTA1-KO MSS31 cells). Tube formation was significantly suppressed in MTA1-KO MSS31 cells, whereas MTA1-overexpression MTA1-KO MSS31 cells regained the ability to form tube-like structures. In addition, western blotting analysis revealed that MTA1-KO MSS31 cells showed significantly higher levels of phosphorylation of non-muscle myosin heavy chain IIa, which resulted in suppression of tube formation. This effect was attributed to a decrease of MTA1/S100 calcium-binding protein A4 complex formation. Moreover, inhibition of tube formation in MTA1-KO MSS31 cells could not be rescued by stimulation with vascular endothelial growth factor (VEGF). These results demonstrated that MTA1 may serve as an essential molecule for angiogenesis in endothelial cells and be involved in different steps of the angiogenic process compared with the VEGF/VEGF receptor 2 pathway. The findings showed that endothelial MTA1 and its pathway may serve as promising targets for inhibiting tumor angiogenesis, further supporting the development of MTA1-based antiangiogenic therapies.
Collapse
Affiliation(s)
- Mizuho Ishikawa
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Mitsuhiko Osaki
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Narumi Uno
- Chromosome Engineering Research Center, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Takahito Ohira
- Chromosome Engineering Research Center, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Hiroyuki Kugoh
- Chromosome Engineering Research Center, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| | - Futoshi Okada
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago, Tottori 683‑8503, Japan
| |
Collapse
|
12
|
Larionova I, Kazakova E, Gerashchenko T, Kzhyshkowska J. New Angiogenic Regulators Produced by TAMs: Perspective for Targeting Tumor Angiogenesis. Cancers (Basel) 2021; 13:cancers13133253. [PMID: 34209679 PMCID: PMC8268686 DOI: 10.3390/cancers13133253] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Since the targeting of a single pro-angiogenic factor fails to improve oncological disease outcome, significant efforts have been made to identify new pro-angiogenic factors that could compensate for the deficiency of current therapy or act independently as single drugs. Our review aims to present the state-of-the art for well-known and recently described factors produced by macrophages that induce and regulate angiogenesis. A number of positive and negative regulators of angiogenesis in the tumor microenvironment are produced by tumor-associated macrophages (TAMs). Accumulating evidence has indicated that, apart from the well-known angiogenic factors, there are plenty of novel angiogenesis-regulating proteins that belong to different classes. We summarize the data regarding the direct or indirect mechanisms of the interaction of these factors with endothelial cells during angiogenesis. We highlight the recent findings that explain the limitations in the efficiency of current anti-angiogenic therapy approaches. Abstract Angiogenesis is crucial to the supply of a growing tumor with nutrition and oxygen. Inhibition of angiogenesis is one of the main treatment strategies for colorectal, lung, breast, renal, and other solid cancers. However, currently applied drugs that target VEGF or receptor tyrosine kinases have limited efficiency, which raises a question concerning the mechanism of patient resistance to the already developed drugs. Tumor-associated macrophages (TAMs) were identified in the animal tumor models as a key inducer of the angiogenic switch. TAMs represent a potent source not only for VEGF, but also for a number of other pro-angiogenic factors. Our review provides information about the activity of secreted regulators of angiogenesis produced by TAMs. They include members of SEMA and S100A families, chitinase-like proteins, osteopontin, and SPARC. The COX-2, Tie2, and other factors that control the pro-angiogenic activity of TAMs are also discussed. We highlight how these recent findings explain the limitations in the efficiency of current anti-angiogenic therapy. Additionally, we describe genetic and posttranscriptional mechanisms that control the expression of factors regulating angiogenesis. Finally, we present prospects for the complex targeting of the pro-angiogenic activity of TAMs.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
- Correspondence: (I.L.); (J.K.)
| | - Elena Kazakova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
| | - Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia;
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, 634050 Tomsk, Russia;
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, 68167 Mannheim, Germany
- Correspondence: (I.L.); (J.K.)
| |
Collapse
|
13
|
Lyu T, Wang Y, Li D, Yang H, Qin B, Zhang W, Li Z, Cheng C, Zhang B, Guo R, Song Y. Exosomes from BM-MSCs promote acute myeloid leukemia cell proliferation, invasion and chemoresistance via upregulation of S100A4. Exp Hematol Oncol 2021; 10:24. [PMID: 33789743 PMCID: PMC8011411 DOI: 10.1186/s40164-021-00220-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023] Open
Abstract
Background BM-MSCs play an important role in cancer development through the release of cytokines or exosomes. Studies have shown that extracellular exosomes derived from BM-MSCs are a key pro-invasive factor. However, how BM-MSC-exos influence AML cell proliferation, invasion and chemoresistance remains poorly understood. Methods We isolated exosomes from BM-MSCs and used electron microscopy, particle size separation and western blots to identify the exosomes. The invasion of leukemia cells was observed with a transwell assay. The stemness traits and chemoresistance of the leukemia cells were detected by FCM, colony formation and CCK-8 assays. TCGA database was used to investigate the prognostic relevance of S100A4 and its potential role in AML. Results In this study, we found that BM-MSC-exos increased the metastatic potential, maintained the stemness and contributed to the chemoresistance of leukemia cells. Mechanistically, BM-MSC-exos promoted the proliferation, invasion and chemoresistance of leukemia cells via upregulation of S100A4. Downregulating S100A4 clearly suppressed the proliferation, invasion, and chemoresistance of leukemia cells after treatment with BM-MSC-exos. Bioinformatic analysis with data in TCGA database showed that S100A4 was associated with poor prognosis in AML patients, and functional enrichment revealed its involvement in the processes of cell–cell adhesion and cytokine regulation. Conclusions S100A4 is vital in the BM-MSC-exo-driven proliferation, invasion and chemoresistance of leukemia cells and may serve as a potential target for leukemia therapy.
Collapse
Affiliation(s)
- Tianxin Lyu
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Yinuo Wang
- Translational Cancer Research Center, Peking University First Hospital, Beijing, 100034, China
| | - Ding Li
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Hui Yang
- Translational Cancer Research Center, Peking University First Hospital, Beijing, 100034, China
| | - Bin Qin
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Wenli Zhang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Zhiyue Li
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Cheng Cheng
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Binglei Zhang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China.
| |
Collapse
|
14
|
Takenaga K, Ochiya T, Endo H. Inhibition of the invasion and metastasis of mammary carcinoma cells by NBD peptide targeting S100A4 via the suppression of the Sp1/MMP‑14 axis. Int J Oncol 2021; 58:397-408. [PMID: 33650647 PMCID: PMC7864152 DOI: 10.3892/ijo.2021.5173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/12/2020] [Indexed: 12/30/2022] Open
Abstract
A synthetic peptide that blocks the interaction between the metastasis‑enhancing calcium‑binding protein, S100A4, and its effector protein, methionine aminopeptidase 2 (MetAP2) (the NBD peptide), was previously demonstrated to inhibit the angiogenesis of endothelial cells, leading to the regression of human prostate cancer in a xenograft model. However, the effects of the NBD peptide on the malignant properties of cancer cells that express S100A4 remain to be elucidated. The present study demonstrates that the NBD peptide inhibits the invasiveness and metastasis of highly metastatic human mammary carcinoma cells. The introduction of the peptide into MDA‑MB‑231 variant cells resulted in the suppression of matrix degradation in a gelatin invadopodia assay and invasiveness in a Matrigel invasion assay. In line with these results, the peptide significantly downregulated the expression of matrix metalloproteinase (MMP)‑14 (MT1‑MMP). Mechanistic analysis of the downregulation of MMP‑14 revealed the suppression of the expression of the transcription factor, specificity protein 1 (Sp1), but not that of nuclear factor (NF)‑κB, early growth response 1 (EGR1) or ELK3, all of which were reported to be involved in transcriptional regulation of the MMP‑14 gene. At the same time, evidence suggested that the NBD peptide also suppressed Sp1 and MMP‑14 expression levels in MDA‑MB‑468 cells. Importantly, the intravenous administration of the NBD peptide encapsulated in liposomes inhibited pulmonary metastasis from mammary gland tumors in mice with xenograft tumors. These results indicate that the NBD peptide can suppress malignant tumor growth through the suppression of the Sp1/MMP‑14 axis. Taken together, these results reveal that the NBD peptide acts on not only endothelial cells, but also on tumor cells in an integrated manner, suggesting that the peptide may prove to be a promising cancer therapeutic peptide drug.
Collapse
Affiliation(s)
- Keizo Takenaga
- Department of Life Science, Faculty of Medicine, Shimane University, Shimane 690-0823
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo 160-0023
| | - Hideya Endo
- Division of Cellular and Molecular Biology, Department of Cancer Biology
- Division of Molecular Pathology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
15
|
Takenaga K, Akimoto M, Koshikawa N, Nagase H. Obesity reduces the anticancer effect of AdipoRon against orthotopic pancreatic cancer in diet-induced obese mice. Sci Rep 2021; 11:2923. [PMID: 33536560 PMCID: PMC7859201 DOI: 10.1038/s41598-021-82617-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The antidiabetic adiponectin receptor agonist AdipoRon has been shown to suppress the tumour growth of human pancreatic cancer cells. Because obesity and diabetes affect pancreatic cancer progression and chemoresistance, we investigated the effect of AdipoRon on orthotopic tumour growth of Panc02 pancreatic cancer cells in DIO (diet-induced obese) prediabetic mice. Administration of AdipoRon into DIO mice fed high-fat diets, in which prediabetic conditions were alleviated to some extent, did not reduce either body weight or tumour growth. However, when the DIO mice were fed low-fat diets, body weight and the blood leptin level gradually decreased, and importantly, AdipoRon became effective in suppressing tumour growth, which was accompanied by increases in necrotic areas and decreases in Ki67-positive cells and tumour microvessels. AdipoRon inhibited cell growth and induced necrotic cell death of Panc02 cells and suppressed angiogenesis of endothelial MSS31 cells. Insulin and IGF-1 only slightly reversed the AdipoRon-induced suppression of Panc02 cell survival but had no effect on the AdipoRon-induced suppression of MSS31 cell angiogenesis. Leptin significantly ameliorated AdipoRon-induced suppression of angiogenesis through inhibition of ERK1/2 activation. These results suggest that obesity-associated factors weaken the anticancer effect of AdipoRon, which indicates the importance of weight loss in combating pancreatic cancer.
Collapse
Affiliation(s)
- Keizo Takenaga
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chiba, 260-8717, Japan.
| | - Miho Akimoto
- Department of Biochemistry, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Nobuko Koshikawa
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chiba, 260-8717, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chiba, 260-8717, Japan
| |
Collapse
|
16
|
Qian W, Cai X, Qian Q, Zhang W, Tian L. Metastasis-associated protein 1 promotes epithelial-mesenchymal transition in idiopathic pulmonary fibrosis by up-regulating Snail expression. J Cell Mol Med 2020; 24:5998-6007. [PMID: 32187849 PMCID: PMC7294111 DOI: 10.1111/jcmm.15062] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/05/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and usually fatal lung disease that lacking effective interventions. It is well known that aberrant activation of transforming growth factor‐beta1 (TGF‐β1) frequently promotes epithelial‐mesenchymal transition (EMT) in IPF. Metastasis‐associated gene 1 (MTA1) has identified as an oncogene in several human tumours, and aberrant MTA1 expression has been related to the EMT regulation. However, its expression and function in IPF remain largely unexplored. Using a combination of in vitro and in vivo studies, we found that MTA1 was significantly up‐regulated in bleomycin‐induced fibrosis rats and TGF‐β1‐treated alveolar type Ⅱ epithelial (RLE‐6TN) cells. Overexpression of MTA1 induced EMT of RLE‐6TN cells, as well as facilitates cell proliferation and migration. In contrast, knockdown of MTA1 reversed TGF‐β1‐induced EMT of RLE‐6TN cells. The pro‐fibrotic action of MTA1 was mediated by increasing Snail expression through up‐regulating Snail promoter activity. Moreover, inhibition of MTA1 effectively attenuated bleomycin‐induced fibrosis in rats. Additionally, we preliminarily found astragaloside IV (ASV), which was previously validated having inhibitory effects on TGF‐β1‐induced EMT, could inhibit MTA1 expression in TGF‐β1‐treated RLE‐6TN cells. These findings highlight the role of MTA1 in TGF‐β1‐mediated EMT that offer novel strategies for the prevention and treatment of IPF.
Collapse
Affiliation(s)
- Weibin Qian
- Department of Lung Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinrui Cai
- Department of Traditional Chinese Medicine, Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qiuhai Qian
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wei Zhang
- Department of Lung Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Li Tian
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
17
|
Wen X, Yu X, Tian Y, Liu Z, Cheng W, Li H, Kang J, Wei T, Yuan S, Tian J. Quantitative shear wave elastography in primary invasive breast cancers, based on collagen-S100A4 pathology, indicates axillary lymph node metastasis. Quant Imaging Med Surg 2020; 10:624-633. [PMID: 32269923 DOI: 10.21037/qims.2020.02.18] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background The purpose of this study was to evaluate the value of quantitative shear wave elastography (SWE) in indicating the axillary lymph node metastasis (LNM) of invasive breast cancers (IBCs) and to investigate if S100A4 plays a key role in promoting metastasis and increasing stiffness in IBC. Methods The differences in SWE of 223 IBC patients were compared between the LNM+ and LNM- groups and the optimal cutoff values of SWE for diagnosing LNM were calculated. We searched the gene expression omnibus (GEO) to determine whether S100A4 was more highly expressed in IBCs that were LNM+ than in those that were LNM-. Sirius red and immunohistochemical staining were used to examine the collagen deposition and S100A4 expression of included tissue samples, and correlations of SWE and S100A4 expression with collagen deposition were analyzed. Results The optimal cutoff values for Emax (the maximum stiff value), Emean (the mean stiff value), and EmeanR (the ratio of Emean between mass and parenchyma) for diagnosing axillary LNM were 111.05 kPa, 79.80 kPa, and 6.89, respectively. GSE9893 exhibited more increased S100A4 expression in IBCs that were LNM+ than in those that were LNM-. Collagen volume fraction (CVF) and the average optical density of S100A4 (AODS100A4) in the LNM+ group were significantly higher than those in the LNM- group. Emax, Emean, EmeanR, and AODS100A4 were all positively correlated with CVF. Conclusions SWE in primary IBC could be useful for indicating axillary LNM. S100A4 may be a factor that regulates cancer-associated collagen deposition and metastasis; however, prospective molecular biological studies are needed.
Collapse
Affiliation(s)
- Xin Wen
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xiwen Yu
- Heilongjiang Academy of Medical Sciences, Harbin 150086, China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Hairu Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jia Kang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Tianci Wei
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Shasha Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|