1
|
Pi T, Yi W, Hu M, Quan X, Tian L, Sun H, Yan S. Assessing genomic diversity and signatures of selection in Qingyuan Wapiti. Anim Genet 2025; 56:e13505. [PMID: 39789716 DOI: 10.1111/age.13505] [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: 07/31/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025]
Abstract
Red deer is a species of family Cervidae that is widely distributed in the world and is often raised to provide antlers, as a trophy or traditional medicine materials, and meat. Currently, the whole genomic data for red deer are very limited. Qingyuan Wapiti (QYW), China's first breed of red deer by artificial breeding, is well known for its high yield of antlers and large body size. The phylogenetic tree showed that QYW had a closer genetic relationship with Tarim red deer than European red deer. To explore the genetic diversity and selection signatures, the whole genome of 28 QYW individuals was sequenced, and 19 401 749 biallelic SNPs and 1 849 784 indels were obtained. The value of observed heterozygosity, expected heterozygosity, and nucleotide diversity were 0.258598, 0.268844, and 0.002193, respectively. Based on Tajima's D and integrated haplotype score analyses, the candidate regions containing 187 genes were detected, including PLD1, ANTXR1, PLCL1, CPE, and CTNNA3, which have been reported to be correlated with osteogenesis and mineralization, growth, and body size by previous studies. The results obtained in this study will contribute to elucidating the genetic mechanisms underlying the formation of excellent traits in QYW and provide the whole genome data for future exploration of genomic diversity and adaptation evolution of red deer worldwide.
Collapse
Affiliation(s)
- Te Pi
- College of Animal Science, Jilin University, Changchun, China
| | - Wenfeng Yi
- College of Animal Science, Jilin University, Changchun, China
| | - Mingyue Hu
- College of Animal Science, Jilin University, Changchun, China
| | - Xinjiao Quan
- Jilin Province Animal Husbandry and Veterinary Academy of Sciences, Changchun, China
| | - Laiming Tian
- Jilin Province Animal Husbandry and Veterinary Academy of Sciences, Changchun, China
| | - Hao Sun
- College of Animal Science, Jilin University, Changchun, China
| | - Shouqing Yan
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
2
|
Kaesler N, Cheng M, Nagai J, O’Sullivan J, Peisker F, Bindels EM, Babler A, Moellmann J, Droste P, Franciosa G, Dugourd A, Saez-Rodriguez J, Neuss S, Lehrke M, Boor P, Goettsch C, Olsen JV, Speer T, Lu TS, Lim K, Floege J, Denby L, Costa I, Kramann R. Mapping cardiac remodeling in chronic kidney disease. SCIENCE ADVANCES 2023; 9:eadj4846. [PMID: 38000021 PMCID: PMC10672229 DOI: 10.1126/sciadv.adj4846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023]
Abstract
Patients with advanced chronic kidney disease (CKD) mostly die from sudden cardiac death and recurrent heart failure. The mechanisms of cardiac remodeling are largely unclear. To dissect molecular and cellular mechanisms of cardiac remodeling in CKD in an unbiased fashion, we performed left ventricular single-nuclear RNA sequencing in two mouse models of CKD. Our data showed a hypertrophic response trajectory of cardiomyocytes with stress signaling and metabolic changes driven by soluble uremia-related factors. We mapped fibroblast to myofibroblast differentiation in this process and identified notable changes in the cardiac vasculature, suggesting inflammation and dysfunction. An integrated analysis of cardiac cellular responses to uremic toxins pointed toward endothelin-1 and methylglyoxal being involved in capillary dysfunction and TNFα driving cardiomyocyte hypertrophy in CKD, which was validated in vitro and in vivo. TNFα inhibition in vivo ameliorated the cardiac phenotype in CKD. Thus, interventional approaches directed against uremic toxins, such as TNFα, hold promise to ameliorate cardiac remodeling in CKD.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Mingbo Cheng
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James Nagai
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - James O’Sullivan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Fabian Peisker
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Eric M. J. Bindels
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Patrick Droste
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Giulia Franciosa
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Sabine Neuss
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, Biointerface Laboratory, RWTH Aachen University, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Pathology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jesper V. Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Thimoteus Speer
- Department of Medicine (Nephrology), Goethe University Frankfurt, Frankfurt, Germany
| | - Tzong-Shi Lu
- Brigham and Women’s Hospital, Renal Division, Boston, MA, USA
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Ivan Costa
- Institute for Computational Genomics, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, Aachen, Germany
- Institute of Experimental Medicine and Systems Biology, University Hospital of the RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
3
|
Kareff SA, Corbett V, Hallenbeck P, Chauhan A. TEM8 in Oncogenesis: Protein Biology, Pre-Clinical Agents, and Clinical Rationale. Cells 2023; 12:2623. [PMID: 37998358 PMCID: PMC10670355 DOI: 10.3390/cells12222623] [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: 10/17/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
The TEM8 protein represents an emerging biomarker in many solid tumor histologies. Given the various roles it plays in oncogenesis, including but not limited to angiogenesis, epithelial-to-mesenchymal transition, and cell migration, TEM8 has recently served and will continue to serve as the target of novel oncologic therapies. We review herein the role of TEM8 in oncogenesis. We review its normal function, highlight the additional roles it plays in the tumor microenvironment, and synthesize pre-clinical and clinical data currently available. We underline the protein's prognostic and predictive abilities in various solid tumors by (1) highlighting its association with more aggressive disease biology and poor clinical outcomes and (2) assessing its associated clinical trial landscape. Finally, we offer future directions for clinical studies involving TEM8, including incorporating pre-clinical agents into clinical trials and combining previously tested oncologic therapies with currently available treatments, such as immunotherapy.
Collapse
Affiliation(s)
- Samuel A. Kareff
- University of Miami Sylvester Comprehensive Cancer Center/Jackson Memorial Hospital, Miami, FL 33136, USA
| | | | | | - Aman Chauhan
- Division of Medical Oncology, Department of Medicine, University of Miami Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| |
Collapse
|
4
|
miR-134-5p inhibits osteoclastogenesis through a novel miR-134-5p/Itgb1/MAPK pathway. J Biol Chem 2022; 298:102116. [PMID: 35691339 PMCID: PMC9257423 DOI: 10.1016/j.jbc.2022.102116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Osteoporosis affects approximately 200 million people and severely affects quality of life, but the exact pathological mechanisms behind this disease remain unclear. Various miRNAs have been shown to play a predominant role in the regulation of osteoclast formation. In this study, we explored the role of miR-134-5p in osteoclastogenesis both in vivo and in vitro. We constructed an ovariectomized (OVX) mouse model and performed microarray analysis using bone tissue from OVX mice and their control counterparts. Quantitative RT-PCR data from bone tissue and bone marrow macrophages (BMMs) confirmed the decreased expression of miR-134-5p in OVX mice observed in microarray analysis. In addition, a decrease in miR-134-5p was also observed during induced osteoclastogenesis of BMMs collected from C57BL/6N mice. Through transfection with miR-134-5p agomirs and antagomirs, we found that miR-134-5p knockdown significantly accelerated osteoclast formation and cell proliferation and inhibited apoptosis. Furthermore, a luciferase reporter assay showed that miR-134-5p directly targets the integrin surface receptor gene Itgb1. Cotransfection with Itgb1 siRNA reversed the effect of the miR-134-5p antagomir in promoting osteoclastogenesis. Moreover, the abundance levels of MAPK pathway proteins phosphorylated-p38 (p-p38) and phosphorylated-ERK (p-ERK) were significantly increased after transfection with the miR-134-5p antagomir but decreased after transfection with the miR-134-5p agomir or Itgb1 siRNA, which indicated a potential relationship between the miR-134-5p/Itgb1 axis and the MAPK pathway. Collectively, these results revealed that miR-134-5p inhibits osteoclast differentiation of BMMs both in vivo and in vitro and that the miR-134-5p/Itgb1/MAPK pathway might be a potential target for osteoporosis therapy.
Collapse
|
5
|
Cheng H, Jin S, Huang S, Hu T, Zhao M, Li D, Wu B. Serum Proteomic Analysis by Tandem Mass Tag-Based Quantitative Proteomics in Pediatric Obstructive Sleep Apnea. Front Mol Biosci 2022; 9:762336. [PMID: 35480887 PMCID: PMC9035643 DOI: 10.3389/fmolb.2022.762336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 03/04/2022] [Indexed: 12/02/2022] Open
Abstract
Pediatric obstructive sleep apnea (OSA) is a frequent respiratory disorder with an estimated prevalence of 3–6% in the general population. However, the underlying pathophysiology of OSA remains unclear. Recently, proteomic analysis using high-resolution and high-throughput mass spectrometry has been widely used in the field of medical sciences. In the present study, tandem mass tag (TMT)-based proteomic analysis was performed in the serum of patients with OSA. The proteomic analysis revealed a set of differentially expressed proteins that may be associated with the pathophysiology of OSA. The differentially expressed proteins in patients with OSA were enriched in pathways including phagosome and glycan synthesis/degradation, immune response, and the hedgehog signaling pathway, indicating that such functions are key targets of OSA. Moreover, the experimental validation studies revealed that four proteins including ANTXR1, COLEC10, NCAM1, and VNN1 were reduced in the serum from patients with moderate and severe OSA, while MAN1A1 and CSPG4 protein levels were elevated in the serum from patients with severe OSA. The protein levels of ANTXR1, COLEC10, NCAM1, and VNN1 were inversely correlated with apnea-hypopnea index (AHI) in the recruited subjects, while the protein level of MAN1A1 was positively correlated with AHI, and no significant correlation was detected between CSPG4 protein and AHI. In summary, the present study for the first time identified differentially expressed proteins in the serum from OSA patients with different severities by using TMT-based proteomic analysis. The functional enrichment studies suggested that several signaling pathways may be associated with the pathophysiology of OSA. The experimental validation results indicated that six proteins including ANTXR1, COLEC10, NCAM1, VNN1, CGPG4, and MAN1A1 may play important roles in the pathophysiology of OSA, which requires further mechanistic investigation.
Collapse
Affiliation(s)
- Hanrong Cheng
- Institute of Respiratory Diseases, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Shoumei Jin
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Simin Huang
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Tianyong Hu
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Miao Zhao
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
| | - Dongcai Li
- Longgang ENT Hospital, Institute of ENT and Shenzhen Key Laboratory of ENT, Shenzhen, China
- *Correspondence: Dongcai Li, ; Benqing Wu,
| | - Benqing Wu
- Department of Neonatology, University of Chinese Academy of Science-Shenzhen Hospital, Shenzhen, China
- *Correspondence: Dongcai Li, ; Benqing Wu,
| |
Collapse
|
6
|
Xu D, Gao HJ, Lu CY, Tian HM, Yu XJ. Vitamin D inhibits bone loss in mice with thyrotoxicosis by activating the OPG/RANKL and Wnt/β-catenin signaling pathways. Front Endocrinol (Lausanne) 2022; 13:1066089. [PMID: 36531471 PMCID: PMC9748851 DOI: 10.3389/fendo.2022.1066089] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/04/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE Vitamin D and thyroid hormones have crucial roles in bone metabolism. This study aims to explore the effects of vitamin D on bone metabolism in mice with thyrotoxicosis and its mechanisms. METHODS 12-week-old mice were randomly divided into 6 groups (6 mice/group), the control (CON) group, vitamin D (VD) group, low-dose LT4 (Low LT4) group, low-dose LT4+VD (Low LT4+VD) group, high-dose LT4 (High LT4) group, high-dose LT4+VD (High LT4+VD) group, LT4 was provided every day and vitamin D3 every other day for 12 weeks. Thyroid function, 25-hydroxy vitamin D, type I collagen carboxy-terminal peptide (CTX), and type I procollagen amino-terminal peptide were determined. In addition, microcomputed tomography, bone histology and histomorphometry, a three-point bending test, and the mRNA expression of osteoprotegerin (OPG), receptor activator of nuclear factor-κB ligand (RANKL) and β-catenin in bone were conducted. RESULTS The BMD of lumbar vertebrae and femur decreased and the bone microstructure was destroyed significantly in thyrotoxicosis mice. Addition of vitamin D improved the BMD and bone microstructure only in the low LT4+VD group. Mice with thyrotoxicosis had a significantly higher level of CTX (P<0.05), which was decreased by treatment with vitamin D (P<0.05). The eroded surface per bone surface (Er. S/BS) of the cancellous bone and elongated surface/endocortical perimeter (Er. S/E Pm) of the cortical bone significantly increased in the Low LT4 and High LT4 groups (P<0.05). Treatment with vitamin D significantly decreased the Er. S/BS and Er. S/E Pm. But, treatment with vitamin D did not significantly improve the toughness and rigidity of bones. The ratio of OPG to RANKL and mRNA expression of β-catenin in the Low LT4+VD group were higher than that in the Low LT4 group (P<0.05). CONCLUSION In mice with thyrotoxicosis, treatment with vitamin D can inhibit bone resorption and improve the BMD and trabecular bone architecture by increasing the ratio of OPG to RANKL and upregulating the expression of Wnt/β-catenin.
Collapse
Affiliation(s)
- Dan Xu
- Division of Endocrinology and Metabolism Internal Medicine, West China Hospital, Sichuan University, Chengdu, China
- Division of Endocrinology & Metabolism, People’s Hospital of Le Shan, Le Shan, China
| | - Hong-Jiao Gao
- Division of Endocrinology and Metabolism Internal Medicine, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology, the Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Chun-Yan Lu
- Division of Endocrinology and Metabolism Internal Medicine, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Chun-Yan Lu, ; Hao-Ming Tian,
| | - Hao-Ming Tian
- Division of Endocrinology and Metabolism Internal Medicine, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Chun-Yan Lu, ; Hao-Ming Tian,
| | - Xi-Jie Yu
- Division of Endocrinology and Metabolism Internal Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Si T, Ning X, Chen H, Hu Z, Dun L, Zheng N, Huang P, Yang L, Yi P. ANTXR1 as a potential prognostic biomarker for hepatitis B virus-related hepatocellular carcinoma identified by a weighted gene correlation network analysis. J Gastrointest Oncol 2021; 12:3079-3092. [PMID: 35070431 PMCID: PMC8748048 DOI: 10.21037/jgo-21-764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/17/2021] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND With high incidence and mortality rates, hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors worldwide. Chronic hepatitis B virus (HBV) infection is a leading cause of HCC, especially for Asians and blacks. However, the molecular mechanisms underlying HBV-related HCC are unclear. This study sought to identify novel prognostic biomarkers and explore the potential pathogenesis of HBV-related HCC. METHODS The gene expression profiles and corresponding clinical information of HCC from The Cancer Genome Atlas Liver Hepatocellular Carcinoma data set were analyzed by a weighted gene co-expression network analysis. Correlations between the co-expression modules and clinical traits were calculated. Next, key modules associated with HBV infection were identified. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were conducted for the genes in the key modules. The hub genes were identified based on the protein-protein interaction (PPI) network via the Cytoscape. Finally, an overall survival (OS) analysis was performed. RESULTS The two modules (i.e., the brown and yellow modules) most relevant to HBV infection were constructed. A functional enrichment analysis revealed that the genes in the two modules were mainly enriched in HCC-related pathways, such as the phosphatidylinositol-3-kinase and protein kinase B signaling pathway, focal adhesion, human papillomavirus infection, the Rap1 signaling pathway, and the cyclic guanosine monophosphate-dependent protein kinase (cGMP-PKG) signaling pathway. Ten hub genes [i.e., COL3A1, ANTXR1, COL14A1, THBS2, ADAMTS2, AEBP1, PRELP, EMILIN1, DCN and PODN] in the brown module, and 10 hub genes [i.e., USP34, SEC24C, ZNF770, STAG1, TSTD2, PKD1P6, CCNK, GFT2I, NT5C2 and SMG6] in the yellow module were identified. Among the hub genes, ANTXR1 (Anthrax-toxin receptor 1) was significantly correlated with HBV-related HCC patients' OS. CONCLUSIONS ANTXR1 represents a potential therapeutic target for HBV-related HCC. This study offers novel insights into the molecular mechanisms of HBV-induced tumorigenesis, which needs to be further validated by basic experiments and large-scale cohort studies.
Collapse
Affiliation(s)
- Tao Si
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Xuejian Ning
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Haihui Chen
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Zhengguo Hu
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Linglu Dun
- Department of Neurology Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Na Zheng
- Department of Neurology Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Ping Huang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Liu Yang
- Department of Oncology, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| | - Ping Yi
- Department of Neurology Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, China
| |
Collapse
|
8
|
Hu N, Zeng X, Tang F, Xiong S. Exosomal long non-coding RNA LIPCAR derived from oxLDL-treated THP-1 cells regulates the proliferation of human umbilical vein endothelial cells and human vascular smooth muscle cells. Biochem Biophys Res Commun 2021; 575:65-72. [PMID: 34455222 DOI: 10.1016/j.bbrc.2021.08.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND It has been reported that long non-coding RNA (lncRNA) LIPCAR is involved in the progression of atherosclerosis. However, the mechanism underlying the effects of LIPCAR on regulating the occurrence and development of atherosclerosis remains unclear. METHODS Reverse transcription-quantitative PCR was performed to detect the levels of LIPCAR in the plasma of patients with atherosclerosis and in THP-1 macrophages. THP-1 cells were stimulated with oxidized low-density lipoprotein (ox-LDL) to induce foam cell formation. Furthermore, Transwell assay was carried out to evaluate the migration ability of vascular smooth muscle cells (VSMCs). RESULTS The expression of LIPCAR in the plasma of patients with atherosclerosis was significantly higher compared with that in healthy subjects, while LIPCAR knockdown notably reversed ox-LDL-induced THP-1 cell apoptosis. In addition, LIPCAR was upregulated in exosomes derived from THP-1 cells treated with ox-LDL (THP-1/ox-LDL Exo). Furthermore, THP-1/ox-LDL Exo significantly increased the expression levels of CDK2 and proliferative cell nuclear antigen in human VSMCs, while these effects were reversed following LIPCAR silencing. CONCLUSION The results of the present study suggested that exosomal lncRNA LIPCAR derived from ox-LDL modified THP-1 cells could promote the progression of atherosclerosis. Therefore, LIPCAR may be considered as a novel biomarker for the development of new strategies to treat atherosclerosis.
Collapse
Affiliation(s)
- Nan Hu
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xixi Zeng
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feifei Tang
- Department of Cardiothoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Sizheng Xiong
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, 430030, China.
| |
Collapse
|
9
|
Urbano N, Scimeca M, Crocco A, Mauriello A, Bonanno E, Schillaci O. 18F-Choline PET/CT Identifies High-Grade Prostate Cancer Lesions Expressing Bone Biomarkers. J Clin Med 2019; 8:1657. [PMID: 31614564 PMCID: PMC6832450 DOI: 10.3390/jcm8101657] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/30/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
The main aim of this study was to investigate the possible association between 18F-choline uptake and histopathological features of prostate biopsies such as the Gleason Group and the expression of both epithelial to mesenchymal transition (vimentin) and bone mineralization (bone morphogenetics protein (BMP)-2, runt-related transcription factor 2 (RUNX2), receptor activator of nuclear factor-κB ligand (RANKL), vitamin D receptor (VDR), and pentraxin 3 (PTX3) in situ biomarkers. To this end, we enrolled 79 consecutive prostate cancer patients that underwent both the 18F-choline PET/CT analysis and the prostate bioptic procedure. The standardized uptake value (SUV) average values were collected from 18F-choline PET/CT analysis whereas Gleason Group and immunostaining data were collected from paraffin-embedded sections. Histological classification showed a heterogenous population including both low/intermediate and high-grade prostate cancers. A significant increase of 18F-choline uptake in high-grade prostate lesions (Gleason Score ≥8) was found. Also, linear regression analysis showed a significant correlation between 18F-choline uptake and the number of vimentin, RANKL, VDR, or PTX3 positive prostate cancer cells. Conversely, we observed no significant association between 18F-choline uptake and the expression of bone biomarkers involved in the early phases of osteoblast differentiation (BMP-2, RUNX2). In conclusion, results here reported can lay the foundation for the use of 18F-choline positron emission tomography (PET)/computed tomography (CT) as a diagnostic tool capable of identifying high-grade prostate cancer lesions expressing bone biomarkers.
Collapse
Affiliation(s)
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy.
- Fondazione Umberto Veronesi (FUV), 20122 Milano, Italy.
| | - Antonio Crocco
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Alessandro Mauriello
- Department of Experimental Medicine, University "Tor Vergata", 00133 Rome, Italy.
| | - Elena Bonanno
- Department of Experimental Medicine, University "Tor Vergata", 00133 Rome, Italy.
- Neuromed Group, 'Diagnostica Medica' & 'Villa dei Platani', 83100 Avellino, Italy.
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy.
- IRCCS Neuromed, 86077 Pozzilli, Italy.
| |
Collapse
|