1
|
Li P, Cui Y, Hu K, Wang X, Yu Y. Silencing APLNR enhances the radiosensitivity of prostate cancer by modulating the PI3K/AKT/mTOR signaling pathway. Clin Transl Oncol 2025; 27:1728-1737. [PMID: 39251496 DOI: 10.1007/s12094-024-03692-1] [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: 06/17/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Aberrant expression of apelin receptor (APLNR) has been found to be involved in various cancers' development, however, its function in prostate cancer (PCa) remains unclear. The research aimed to investigate the role and potential mechanism of APLNR in PCa. METHODS The mRNA expression of APLNR was detected via qRT-PCR assay. PCa cell proliferation and apoptosis were determined through plate cloning and flow cytometry. In addition, the expression of apoptosis-related proteins (Bax, Bcl-2, and cleaved caspase-3) was evaluated using western blot. DNA damage marker (γ-H2AX) was analyzed by immunofluorescence and western blot. GSEA analysis was performed for seeking enrichment pathways of APLNR in PCa, and the protein levels of PI3K, p-PI3K, AKT, p-AKT, mTOR, and p-mTOR were tested using western blot. RESULTS APLNR expression was up-regulated in PCa tissues and cells. Silencing APLNR enhanced the sensitivity of PCa cells to radiotherapy, which was manifested by inhibiting cell proliferation, promoting cell apoptosis, and promoting DNA damage. Next, silencing APLNR inhibited the PI3K/AKT/mTOR pathway. Specifically, 740Y-P (the PI3K/AKT/mTOR pathway activator) reversed the effects of silencing APLNR on PCa cell proliferation, apoptosis and DNA damage. CONCLUSION Silencing APLNR inhibited cell proliferation, promoted cell apoptosis, and enhanced the radiosensitivity of PCa cells, which was involved in the PI3K/AKT/mTOR signaling pathway. This study is conducive to the deeper understanding of PCa and further provides a new perspective for the treatment of PCa.
Collapse
Affiliation(s)
- Peng Li
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China
| | - Yanfang Cui
- Department of Ultrasonography, Yantaishan Hospital, Yantai, 264003, Shandong, People's Republic of China
| | - Keyao Hu
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China
| | - Xiaofei Wang
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China
| | - Yizhi Yu
- Department of Urology, Yantaishan Hospital, Laishan District Science and Technology Avenue 10087, Yantai, 264003, Shandong, People's Republic of China.
| |
Collapse
|
2
|
Zhang Y, Liu YJ, Mei J, Yang ZX, Qian XP, Huang W. An Analysis Regarding the Association Between DAZ Interacting Zinc Finger Protein 1 (DZIP1) and Colorectal Cancer (CRC). Mol Biotechnol 2025; 67:527-547. [PMID: 38334905 DOI: 10.1007/s12033-024-01065-1] [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: 11/09/2023] [Accepted: 12/21/2023] [Indexed: 02/10/2024]
Abstract
Colorectal cancer (CRC) is the third most common malignant disease worldwide, and its incidence is increasing, but the molecular mechanisms of this disease are highly heterogeneous and still far from being fully understood. Increasing evidence suggests that fibrosis mediated by abnormal activation of fibroblasts based in the microenvironment is associated with a poor prognosis. However, the function and pathogenic mechanisms of fibroblasts in CRC remain unclear. Here, combining scrna-seq and clinical specimen data, DAZ Interacting Protein 1 (DZIP1) was found to be expressed on fibroblasts and cancer cells and positively correlated with stromal deposition. Importantly, pseudotime-series analysis showed that DZIP1 levels were up-regulated in malignant transformation of fibroblasts and experimentally confirmed that DZIP1 modulates activation of fibroblasts and promotes epithelial-mesenchymal transition (EMT) in tumor cells. Further studies showed that DZIP1 expressed by tumor cells also has a driving effect on EMT and contributes to the recruitment of more fibroblasts. A similar phenomenon was observed in xenografted nude mice. And it was confirmed in xenograft mice that downregulation of DZIP1 expression significantly delayed tumor formation and reduced tumor size in CRC cells. Taken together, our findings suggested that DZIP1 was a regulator of the CRC mesenchymal phenotype. The revelation of targeting DZIP1 provides a new avenue for CRC therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
- Department of Oncology, Nanjing Tianyinshan Hospital, Nanjing, 211199, Jiangsu, China
| | - Yuan-Jie Liu
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jia Mei
- Department of Pathology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Zhao-Xu Yang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Xiao-Ping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Clinical Cancer Institute of Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Wei Huang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Hanzhong Road No.155, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
3
|
Cores Ziskoven P, Nogueira AVB, Eick S, Deschner J. Apelin Counteracts the Effects of Fusobacterium nucleatum on the Migration of Periodontal Ligament Cells In Vitro. Int J Mol Sci 2024; 25:10729. [PMID: 39409058 PMCID: PMC11476847 DOI: 10.3390/ijms251910729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
To better understand the link between periodontitis and metabolic diseases, our in vitro study aimed to assess the influence of the adipokine apelin and/or the periodontal pathogen Fusobacterium nucleatum on periodontal cells. Periodontal ligament (PDL) cells were exposed to F. nucleatum in the presence and absence of apelin. Scratch assays were used to analyze the in vitro wound healing and velocity of cell migration. To investigate if F. nucleatum and/or apelin have a regulatory effect on cell proliferation and apoptosis, proliferation and viability assays were performed as well as an analysis of caspase 9 expression. Both the in vitro wound closure and the cell migration rate were significantly reduced by F. nucleatum. Simultaneous incubation with apelin counteracted the adverse effects of F. nucleatum. The proliferation assay demonstrated that neither apelin nor F. nucleatum significantly affected PDL cell proliferation. Furthermore, neither apelin nor F. nucleatum was cytotoxic or affected apoptosis after 48 h. Apelin could play a modulatory role in the pathogenesis of periodontitis, as it was able to compensate for the inhibitory effects of the periodontal pathogen F. nucleatum on PDL cell migration in vitro.
Collapse
Affiliation(s)
- Pablo Cores Ziskoven
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (P.C.Z.); (A.V.B.N.)
| | - Andressa V. B. Nogueira
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (P.C.Z.); (A.V.B.N.)
| | - Sigrun Eick
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland;
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (P.C.Z.); (A.V.B.N.)
| |
Collapse
|
4
|
Yamada D, Kojima Y, Hosoya A, Suzuki M, Watabe T, Inoue T, Tsugawa N, Asakawa T, Yonemoto Y, Onizawa M, Nemoto Y, Oshima S, Shimonaka M, Kuba K, Ishida J, Fukamizu A, Penninger JM, Watanabe M, Okamoto R, Nagaishi T. Apelin expression is downregulated in T cells in a murine model of chronic colitis. Biochem Biophys Res Commun 2023; 647:72-79. [PMID: 36731336 DOI: 10.1016/j.bbrc.2023.01.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Apelin (APL), an endogenous ligand for APJ, has been reported to be upregulated in a murine model of acute colitis induced by sodium dextran sulfate, as well as inflammatory bowel diseases (IBD) in humans. However, the mechanisms and functions of APL/APJ axis in the pathogenesis of IBD are unclear. We herein analyzed CD4+ T cells to determine the functions of APL in a murine model of chronic colitis induced in Rag deficient mice (Rag-/-). In colonic tissues of wild-type mice (WT), we found that APL was expressed especially in the lamina propria lymphocytes, where CD4+ T cells are dominant, rather than the epithelial cells. Unexpectedly, the APL expression was rather downregulated in the colonic tissue of the chronic colitis group compared to the control groups (Rag-/- before colitis induction and WT). The APL expression was downregulated when naïve T cells were differentiated into effecter T cells. A lack of APL resulted in decreased naïve T cells and increased effecter T cells in secondary lymphoid organs. A synthetic APL peptide, [Pyr1]-APL-13, increased IL-10 and decreased IFN-γ productions by effecter T cells. Administration of [Pyr1]-APL-13 improved survival rate in association with lessened colitis severity and decreased pro-inflammatory cytokine production. This is the first report showing immunological function of APL specifically on T cells, and these results indicate that APL/APJ axis may be a novel therapeutic target for IBD.
Collapse
Affiliation(s)
- Daiki Yamada
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yudai Kojima
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Chemistry, Tokyo University of Science, Tokyo, Japan
| | - Akinori Hosoya
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masahiro Suzuki
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Taro Watabe
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tadahiko Inoue
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Chemistry, Tokyo University of Science, Tokyo, Japan
| | - Naoya Tsugawa
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takehito Asakawa
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuki Yonemoto
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Michio Onizawa
- Department of Advanced Therapeutics for GI Diseases, Graduate School of Medical Science, TMDU, Tokyo, Japan
| | - Yasuhiro Nemoto
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shigeru Oshima
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | | | - Keiji Kuba
- Department of Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junji Ishida
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Bio Center, Vienna, Austria; Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | | - Ryuichi Okamoto
- Department of Gastroenterology, Graduate School of Medical Science, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takashi Nagaishi
- Department of Advanced Therapeutics for GI Diseases, Graduate School of Medical Science, TMDU, Tokyo, Japan.
| |
Collapse
|
5
|
Regulatory role of apelin receptor signaling in migration and differentiation of mouse embryonic stem cell-derived mesoderm cells and mesenchymal stem/stromal cells. Hum Cell 2023; 36:612-630. [PMID: 36692671 DOI: 10.1007/s13577-023-00861-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
Mesoderm-derived cells, including bone, muscle, and mesenchymal stem/stromal cells (MSCs), constitute various parts of vertebrate body. Cell therapy with mesoderm specification in vitro may be a promising treatment for diseases affecting organs of mesodermal origin. Repair and regeneration of damaged organs with in vitro generation of mesoderm-derived tissues and MSCs hold a great potential for regenerative therapy. Therefore, understanding the signaling pathways involving mesoderm and mesoderm-derived cellular differentiation is important. Previous findings indicated the importance of Apelin receptor (Aplnr) signaling, during embryonic development, in gastrulation, cell migration, and differentiation. Nevertheless, regulatory role of Aplnr pathway in differentiation of mesoderm and mesoderm-derived MSCs remains unclear. In the current study, we tried to elucidate the role of Aplnr signaling during mesoderm cell migration and differentiation from mouse embryonic stem cells (mESCs). By activating and suppressing Aplnr signaling pathway via peptide, small molecule, and genetic modifications including siRNA- and shRNA-mediated knockdown and CRISPR-Cas9-mediated knockout (KO), we revealed that Aplnr signaling not only induces migration of cells during germ layer formation but also enhances mesoderm differentiation through FGF/MAPK pathway. Antibody array and LC/MS protein profiling data demonstrated that Apelin-13 treatment enhanced cell cycle, EGFR, FGF, Wnt, and Integrin signaling pathway proteins. Furthermore, Aplelin-13 treatment improved MSC characteristics, with mesenchymal phenotype and high expression of MSC markers, and silencing Aplnr signaling components resulted in significantly reduced expression of MSC markers. Also, Aplnr signaling activity enhanced proliferation and survival of the cells during MSC derivation from mesoderm.
Collapse
|
6
|
Regional immunity of chicken adipose tissue responds to secondary immunity induced by Newcastle disease vaccine via promoting immune activation and weakening lipid metabolism. Poult Sci 2023; 102:102646. [PMID: 37031585 PMCID: PMC10105486 DOI: 10.1016/j.psj.2023.102646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Adipose tissue (AT) is considered as a regional immune organ and plays an important role in the anti-infection immune response. However, the function and mechanism of chicken AT in response to secondary immune response remain poorly understood. Here, we used mRNA and microRNA (miRNA) sequencing technology to survey the transcriptomic landscape of chicken abdominal adipose tissue (AAT) during the first and second immunization with Newcastle disease virus (NDV) vaccine, and carried out bioinformatics analysis, such as Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis, protein-protein interaction (PPI) analysis, and miRNA-mRNA integrated analysis. The results indicated that chicken AAT actively responded to the secondary immune response. DNA replication and cytoskeleton regulation as the regulatory functions of immune activation changed significantly, and weakened lipid metabolism was an effective strategy for the secondary immunity. Mechanically, the regulatory network between the differentially expressed miRNAs (DEMs) and their targeted differentially expressed genes (DEGs), such as miR-206/miR-499-5p-nuclear receptor subfamily 4 group A member 3 (NR4A3)/methylsterol monooxygenase 1 (MSMO1) pathway, was one of the potential key mechanisms by which AAT responded to the secondary immune response. In conclusion, regional immunity of chicken AT responds to secondary immunity by promoting immune activation and weakening lipid metabolism, and this study can instruct future research on antiviral strategy.
Collapse
|
7
|
Chen J, Li Z, Zhao Q, Chen L. Roles of apelin/APJ system in cancer: Biomarker, predictor, and emerging therapeutic target. J Cell Physiol 2022; 237:3734-3751. [DOI: 10.1002/jcp.30845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Jiawei Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| | - Zhiyue Li
- Health Management Center, The Third Xiangya Hospital Central South University Changsha Hunan Province China
| | - Qun Zhao
- Department of Orthopedics Third Xiangya Hospital of Central South University Changsha Hunan China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| |
Collapse
|
8
|
Yang Y, Chen M, Qiu Y, Li X, Huang Y, Zhang W. The Apelin/APLNR system modulates tumor immune response by reshaping the tumor microenvironment. Gene X 2022; 834:146564. [PMID: 35598689 DOI: 10.1016/j.gene.2022.146564] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 11/04/2022] Open
Abstract
Apelin is an endogenous ligand of the Apelin receptor (APLNR), a seven-transmembrane G protein-coupled receptor, which is widely distributed in human tissue. The Apelin/APLNR system is involved in regulating several physiological and pathological processes. The Apelin expression is increased in a variety of cancer and the Apelin/APLNR system could regulate the development of tumors through mediating autophagy, apoptosis, pyroptosis, and other biological processes to regulate tumor cell proliferation, migration, and invasion. The Apelin/APLNR system also participates in immune response and immune regulation through PI3K-Akt, ERK-MAPK, and other signal pathways. The latest research points out that there is a negative regulatory relationship between APLNR and immune checkpoint PD-L1. In this review, we outline the significance of the Apelin/APLNR signaling pathway in tumorigenesis and its immune regulation. These endeavors provide new insights into the translational application of Apelin/APLNR in cancer and may contribute to the promotion of more effective treatments for cancers.
Collapse
Affiliation(s)
- Yuqin Yang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Meilin Chen
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Yanbing Qiu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Xiaoxu Li
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Yumei Huang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China
| | - Wenling Zhang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province 410013, PR China.
| |
Collapse
|
9
|
Keren A, Bertolini M, Keren Y, Ullmann Y, Paus R, Gilhar A. Human organ rejuvenation by VEGF-A: Lessons from the skin. SCIENCE ADVANCES 2022; 8:eabm6756. [PMID: 35749494 PMCID: PMC9232104 DOI: 10.1126/sciadv.abm6756] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Transplanting aged human skin onto young SCID/beige mice morphologically rejuvenates the xenotransplants. This is accompanied by angiogenesis, epidermal repigmentation, and substantial improvements in key aging-associated biomarkers, including ß-galactosidase, p16ink4a, SIRT1, PGC1α, collagen 17A, and MMP1. Angiogenesis- and hypoxia-related pathways, namely, vascular endothelial growth factor A (VEGF-A) and HIF1A, are most up-regulated in rejuvenated human skin. This rejuvenation cascade, which can be prevented by VEGF-A-neutralizing antibodies, appears to be initiated by murine VEGF-A, which then up-regulates VEGF-A expression/secretion within aged human skin. While intradermally injected VEGF-loaded nanoparticles suffice to induce a molecular rejuvenation signature in aged human skin on old mice, VEGF-A treatment improves key aging parameters also in isolated, organ-cultured aged human skin, i.e., in the absence of functional skin vasculature, neural, or murine host inputs. This identifies VEGF-A as the first pharmacologically pliable master pathway for human organ rejuvenation in vivo and demonstrates the potential of our humanized mouse model for clinically relevant aging research.
Collapse
Affiliation(s)
- Aviad Keren
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | - Marta Bertolini
- Monasterium Laboratory, Skin and Hair Research Solutions GmbH, Münster, Germany
| | - Yaniv Keren
- Division of Orthopedic Surgery, Rambam Health Care Campus, Haifa, Israel
| | - Yehuda Ullmann
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | - Ralf Paus
- Monasterium Laboratory, Skin and Hair Research Solutions GmbH, Münster, Germany
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- CUTANEON–Skin & Hair Innovations, Hamburg, Germany
- Corresponding author. (A.G.); (R.P.)
| | - Amos Gilhar
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
- Corresponding author. (A.G.); (R.P.)
| |
Collapse
|
10
|
Aryankalayil MJ, Martello S, Bylicky MA, Chopra S, May JM, Shankardass A, MacMillan L, Sun L, Sanjak J, Vanpouille-Box C, Eke I, Coleman CN. Analysis of lncRNA-miRNA-mRNA expression pattern in heart tissue after total body radiation in a mouse model. J Transl Med 2021; 19:336. [PMID: 34364390 PMCID: PMC8349067 DOI: 10.1186/s12967-021-02998-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/23/2021] [Indexed: 12/14/2022] Open
Abstract
Background Radiation therapy is integral to effective thoracic cancer treatments, but its application is limited by sensitivity of critical organs such as the heart. The impacts of acute radiation-induced damage and its chronic effects on normal heart cells are highly relevant in radiotherapy with increasing lifespans of patients. Biomarkers for normal tissue damage after radiation exposure, whether accidental or therapeutic, are being studied as indicators of both acute and delayed effects. Recent research has highlighted the potential importance of RNAs, including messenger RNAs (mRNAs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as biomarkers to assess radiation damage. Understanding changes in mRNA and non-coding RNA expression will elucidate biological pathway changes after radiation. Methods To identify significant expression changes in mRNAs, lncRNAs, and miRNAs, we performed whole transcriptome microarray analysis of mouse heart tissue at 48 h after whole-body irradiation with 1, 2, 4, 8, and 12 Gray (Gy). We also validated changes in specific lncRNAs through RT-qPCR. Ingenuity Pathway Analysis (IPA) was used to identify pathways associated with gene expression changes. Results We observed sustained increases in lncRNAs and mRNAs, across all doses of radiation. Alas2, Aplnr, and Cxc3r1 were the most significantly downregulated mRNAs across all doses. Among the significantly upregulated mRNAs were cell-cycle arrest biomarkers Gdf15, Cdkn1a, and Ckap2. Additionally, IPA identified significant changes in gene expression relevant to senescence, apoptosis, hemoglobin synthesis, inflammation, and metabolism. LncRNAs Abhd11os, Pvt1, Trp53cor1, and Dino showed increased expression with increasing doses of radiation. We did not observe any miRNAs with sustained up- or downregulation across all doses, but miR-149-3p, miR-6538, miR-8101, miR-7118-5p, miR-211-3p, and miR-3960 were significantly upregulated after 12 Gy. Conclusions Radiation-induced RNA expression changes may be predictive of normal tissue toxicities and may indicate targetable pathways for radiation countermeasure development and improved radiotherapy treatment plans. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02998-w.
Collapse
Affiliation(s)
- Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA.
| | - Shannon Martello
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Michelle A Bylicky
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Sunita Chopra
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Jared M May
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | - Aman Shankardass
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA
| | | | - Landy Sun
- Gryphon Scientific, Takoma Park, MD, 20912, USA
| | | | | | - Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA.,Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Room B3B406, Bethesda, MD, 20892, USA.,Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA
| |
Collapse
|
11
|
Alishahedani ME, Yadav M, McCann KJ, Gough P, Castillo CR, Matriz J, Myles IA. Therapeutic candidates for keloid scars identified by qualitative review of scratch assay research for wound healing. PLoS One 2021; 16:e0253669. [PMID: 34143844 PMCID: PMC8213172 DOI: 10.1371/journal.pone.0253669] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
The scratch assay is an in vitro technique used to analyze cell migration, proliferation, and cell-to-cell interaction. In the assay, cells are grown to confluence and then ‘scratched’ with a sterile instrument. For the cells in the leading edge, the resulting polarity induces migration and proliferation in attempt to ‘heal’ the modeled wound. Keloid scars are known to have an accelerated wound closure phenotype in the scratch assay, representing an overactivation of wound healing. We performed a qualitative review of the recent literature searching for inhibitors of scratch assay activity that were already available in topical formulations under the hypothesis that such compounds may offer therapeutic potential in keloid treatment. Although several shortcomings in the scratch assay literature were identified, caffeine and allicin successfully inhibited the scratch assay closure and inflammatory abnormalities in the commercially available keloid fibroblast cell line. Caffeine and allicin also impacted ATP production in keloid cells, most notably with inhibition of non-mitochondrial oxygen consumption. The traditional Chinese medicine, shikonin, was also successful in inhibiting scratch closure but displayed less dramatic impacts on metabolism. Together, our results partially summarize the strengths and limitations of current scratch assay literature and suggest clinical assessment of the therapeutic potential for these identified compounds against keloid scars may be warranted.
Collapse
Affiliation(s)
- Mohammadali E. Alishahedani
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, North Bethesda, Maryland, United States of America
| | - Manoj Yadav
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, North Bethesda, Maryland, United States of America
| | - Katelyn J. McCann
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, United States of America
| | - Portia Gough
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, North Bethesda, Maryland, United States of America
| | - Carlos R. Castillo
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, North Bethesda, Maryland, United States of America
| | - Jobel Matriz
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, North Bethesda, Maryland, United States of America
| | - Ian A. Myles
- Epithelial Therapeutics Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, North Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
12
|
Apelin Receptor Signaling During Mesoderm Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32648246 DOI: 10.1007/5584_2020_567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The Apelin receptor (Aplnr) is a G-protein coupled receptor which has a wide body distribution and various physiological roles including homeostasis, angiogenesis, cardiovascular and neuroendocrine function. Apelin and Elabela are two peptide components of the Aplnr signaling and are cleaved to give different isoforms which are active in different tissues and organisms.Aplnr signaling is related to several pathologies including obesity, heart disases and cancer in the adult body. However, the developmental role in mammalian embryogenesis is crucial for migration of early cardiac progenitors and cardiac function. Aplnr and peptide components have a role in proliferation, differentiation and movement of endodermal precursors. Although expression of Aplnr signaling is observed in endodermal lineages, the main function is the control of mesoderm cell movement and cardiac development. Mutant of the Aplnr signaling components results in the malformations, defects and lethality mainly due to the deformed heart function. This developmental role share similarity with the cardiovascular functions in the adult body.Determination of Aplnr signaling and underlying mechanisms during mammalian development might enable understanding of regulatory molecular mechanisms which not only control embryonic development process but also control tissue function and disease pathology in the adult body.
Collapse
|