1
|
Li J, Zhao W, Yang J, Lu P, Sun H, Zhang Z, Gu J. Proteomic and serological markers for diagnosing cardia gastric cancer and precursor lesions in a Chinese population. Sci Rep 2024; 14:25309. [PMID: 39455684 PMCID: PMC11512066 DOI: 10.1038/s41598-024-75912-1] [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: 07/03/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Cardia gastric cancer (CGC) is prevalent in East Asia, and noninvasive, cost-effective screening methods are needed. This study investigated the diagnostic value of serum pepsinogen (PG), gastrin-17 (G-17), Helicobacter pylori (H. pylori) antibodies, and proteomic profiling for CGC and precancerous lesions. We conducted a case-control study involving biopsy-confirmed patients with CGC (n = 60), low-grade intraepithelial neoplasia (CLGD, n = 60), high-grade intraepithelial neoplasia (CHGD, n = 64), and healthy controls (n = 120) matched for age and sex from high-incidence areas in China. Serological markers including PGI, PGII, G-17, and H. pylori were measured using ELISA and Western blot, while plasma protein markers were assessed using Olink® technology. The VSOLassoBag algorithm and nine machine learning (ML) algorithms were employed to identify crucial features and construct predictive models. Various evaluation metrics, including the area under the receiver-operating-characteristic curve (AUC), were utilized to compare predictive performance. Elevated PGII levels, decreased PGR, and H. pylori infection were significantly associated with an increased risk of CGC and precancerous lesions (P for trend < 0.05). The eXtreme Gradient Boosting (XGBoost) model performed best in discriminative ability among the 9 ML models. Following feature reduction based on predictive performance, a final explainable XGBoost model was developed, incorporating five protein biomarkers (CDHR2, ICAM4, PTPRM, CDC27, and FLT1). This model exhibited excellent performance in distinguishing individuals with CGC and precancerous lesions from healthy controls (AUC = 0.931 for CGC, 0.867 for CHGD, and 0.763 for CLGD), surpassing the traditional serological marker-based model. This study underscores the diagnostic potential of serological markers and proteomic profiling in the detection of CGC. Further validation and exploration of combined biomarker approaches are warranted to enhance early diagnosis and improve outcomes in high-risk populations.
Collapse
Affiliation(s)
- Jiqing Li
- Qilu Hospital of Shandong University, 107 Wenhua West Road, Lixia District, Jinan, Shandong Province, China
| | - Wei Zhao
- Qilu Hospital of Shandong University, 107 Wenhua West Road, Lixia District, Jinan, Shandong Province, China
| | - Jia Yang
- Liaocheng People's Hospital, Liaocheng, China
| | - Peipei Lu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Heming Sun
- Department of Computer and Information Science and Engineering, University of Florida, Gainesville, USA
| | - Zhenhong Zhang
- Medical Imaging Center, Qilu Hospital of Shandong University Dezhou Hospital, Jinan, China
| | - Jianhua Gu
- Qilu Hospital of Shandong University, 107 Wenhua West Road, Lixia District, Jinan, Shandong Province, China.
- Office of National Central Cancer Registry, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Gal Z, Torok D, Gonda X, Eszlari N, Anderson IM, Deakin B, Petschner P, Juhasz G, Bagdy G. New Evidence for the Role of the Blood-Brain Barrier and Inflammation in Stress-Associated Depression: A Gene-Environment Analysis Covering 19,296 Genes in 109,360 Humans. Int J Mol Sci 2024; 25:11332. [PMID: 39457114 PMCID: PMC11508422 DOI: 10.3390/ijms252011332] [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: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Mounting evidence supports the key role of the disrupted integrity of the blood-brain barrier (BBB) in stress- and inflammation-associated depression. We assumed that variations in genes regulating the expression and coding proteins constructing and maintaining this barrier, along with those involved in inflammation, have a predisposing or protecting role in the development of depressive symptoms after experiencing severe stress. To prove this, genome-by-environment (GxE) interaction analyses were conducted on 6.26 M SNPS covering 19,296 genes on PHQ9 depression in interaction with adult traumatic events scores in the UK Biobank (n = 109,360) in a hypothesis-free setup. Among the 63 genes that were significant in stress-connected depression, 17 were associated with BBB, 23 with inflammatory processes, and 4 with neuroticism. Compared to all genes, the enrichment of significant BBB-associated hits was 3.82, and those of inflammation-associated hits were 1.59. Besides some sex differences, CSMD1 and PTPRD, encoding proteins taking part in BBB integrity, were the most significant hits in both males and females. In conclusion, the identified risk genes and their encoded proteins could provide biomarkers or new drug targets to promote BBB integrity and thus prevent or decrease stress- and inflammation-associated depressive symptoms, and possibly infection, e.g., COVID-19-associated mental and neurological symptoms.
Collapse
Affiliation(s)
- Zsofia Gal
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Dora Torok
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Xenia Gonda
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
- Department of Psychiatry and Psychotherapy, Semmelweis University, 1083 Budapest, Hungary
| | - Nora Eszlari
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Ian Muir Anderson
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester M13 9NT, UK; (I.M.A.); (B.D.)
| | - Bill Deakin
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester M13 9NT, UK; (I.M.A.); (B.D.)
| | - Peter Petschner
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
- Bioinformatics Center, Institute of Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Research Unit for Realization of Sustainable Society, Kyoto University, Gokasho, Uji, Kyoto 611-0011, Japan
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Faculty of Pharmaceutical Sciences, Semmelweis University, 1089 Budapest, Hungary; (Z.G.); (D.T.); (N.E.); (P.P.); (G.J.)
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, 1089 Budapest, Hungary;
| |
Collapse
|
3
|
Qian Z, Song D, Ipsaro JJ, Bautista C, Joshua-Tor L, Yeh JTH, Tonks NK. Manipulating PTPRD function with ectodomain antibodies. Genes Dev 2023; 37:743-759. [PMID: 37669874 PMCID: PMC10546974 DOI: 10.1101/gad.350713.123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/28/2023] [Indexed: 09/07/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are critical regulators of signal transduction but have yet to be exploited fully for drug development. Receptor protein tyrosine phosphatase δ (RPTPδ/PTPRD) has been shown to elicit tumor-promoting functions, including elevating SRC activity and promoting metastasis in certain cell contexts. Dimerization has been implicated in the inhibition of receptor protein tyrosine phosphatases (RPTPs). We have generated antibodies targeting PTPRD ectodomains with the goal of manipulating their dimerization status ectopically, thereby regulating intracellular signaling. We have validated antibody binding to endogenous PTPRD in a metastatic breast cancer cell line, CAL51, and demonstrated that a monoclonal antibody, RD-43, inhibited phosphatase activity and induced the degradation of PTPRD. Similar effects were observed following chemically induced dimerization of its phosphatase domain. Mechanistically, RD-43 triggered the formation of PTPRD dimers in which the phosphatase activity was impaired. Subsequently, the mAb-PTPRD dimer complex was degraded through lysosomal and proteasomal pathways, independently of secretase cleavage. Consequently, treatment with RD-43 inhibited SRC signaling and suppressed PTPRD-dependent cell invasion. Together, these findings demonstrate that manipulating RPTP function via antibodies to the extracellular segments has therapeutic potential.
Collapse
Affiliation(s)
- Zhe Qian
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Graduate Program of Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York 11760, USA
| | - Dongyan Song
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Jonathan J Ipsaro
- Howard Hughes Medical Institute, W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | | - Leemor Joshua-Tor
- Howard Hughes Medical Institute, W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Johannes T-H Yeh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - Nicholas K Tonks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
| |
Collapse
|
4
|
Sers C, Schäfer R. Silencing effects of mutant RAS signalling on transcriptomes. Adv Biol Regul 2023; 87:100936. [PMID: 36513579 DOI: 10.1016/j.jbior.2022.100936] [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: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022]
Abstract
Mutated genes of the RAS family encoding small GTP-binding proteins drive numerous cancers, including pancreatic, colon and lung tumors. Besides the numerous effects of mutant RAS gene expression on aberrant proliferation, transformed phenotypes, metabolism, and therapy resistance, the most striking consequences of chronic RAS activation are changes of the genetic program. By performing systematic gene expression studies in cellular models that allow comparisons of pre-neoplastic with RAS-transformed cells, we and others have estimated that 7 percent or more of all transcripts are altered in conjunction with the expression of the oncogene. In this context, the number of up-regulated transcripts approximates that of down-regulated transcripts. While up-regulated transcription factors such as MYC, FOSL1, and HMGA2 have been identified and characterized as RAS-responsive drivers of the altered transcriptome, the suppressed factors have been less well studied as potential regulators of the genetic program and transformed phenotype in the breadth of their occurrence. We therefore have collected information on downregulated RAS-responsive factors and discuss their potential role as tumor suppressors that are likely to antagonize active cancer drivers. To better understand the active mechanisms that entail anti-RAS function and those that lead to loss of tumor suppressor activity, we focus on the tumor suppressor HREV107 (alias PLAAT3 [Phospholipase A and acyltransferase 3], PLA2G16 [Phospholipase A2, group XVI] and HRASLS3 [HRAS-like suppressor 3]). Inactivating HREV107 mutations in tumors are extremely rare, hence epigenetic causes modulated by the RAS pathway are likely to lead to down-regulation and loss of function.
Collapse
Affiliation(s)
- Christine Sers
- Laboratory of Molecular Tumor Pathology and systems Biology, Institute of Pathology, Charité Universitätstmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany; German Cancer Consortium, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany
| | - Reinhold Schäfer
- Comprehensive Cancer Center, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany; German Cancer Consortium, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany.
| |
Collapse
|
5
|
Ou C, Peng Q, Zeng C. An integrative prognostic and immune analysis of PTPRD in cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:5361-5379. [PMID: 35603359 DOI: 10.3934/mbe.2022251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
PTPRD plays an indispensable role in the occurrence of multiple tumors. However, pan-cancer analysis is unavailable. The purpose of this research was to preliminarily study its prognostic landscape across various tumors and investigate its relationship with immunotherapy. We exhibited the expression profile, survival analysis, and genomic alterations of PTPRD based on the TIMER, GEPIA, UALCAN, PrognoScan and cBioPortal database. The frequency of PTPRD mutation and its correlation with response to immunotherapy were evaluated using the cBioPortal database. The relationship between PTPRD and immune-cell infiltration was analyzed by the TIMER and TISIDB databases. A protein interaction network was constructed by the STRING database. GO and KEGG enrichment analysis was executed by the Metascape database. A correlation between PTPRD expression and prognosis was found in various cancers. Aberrant PTPRD expression was closely related to immune infiltration. In non-small cell lung cancer and melanoma, patients with PTPRD mutations had better overall survival with immune checkpoint inhibitors, and these patients had higher TMB scores. PTPRD mutation was involved in numerous biological processes, including immunological signaling pathways. A PTPRD protein interaction network was constructed, and genes that interacted with PTPRD were identified. Functional enrichment analysis demonstrated that a variety of GO biological processes and KEGG pathways associated with PTPRD were involved in the therapeutic mechanisms. These results revealed that PTPRD might function as a biomarker for prognosis and immune infiltration in cancers, throwing new light on cancer therapeutics.
Collapse
Affiliation(s)
- Chunpei Ou
- Department of Orthopedics and Traumatology, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Qin Peng
- Department of Health Management, Shenzhen Longhua District Central Hospital, Shenzhen 518110, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China
| |
Collapse
|
6
|
Feng X, Cao A, Qin T, Zhang Q, Fan S, Wang B, Song B, Yu X, Li L. Abnormally elevated ubiquilin‑1 expression in breast cancer regulates metastasis and stemness via AKT signaling. Oncol Rep 2021; 46:236. [PMID: 34528694 PMCID: PMC8453688 DOI: 10.3892/or.2021.8187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Ubiquilin-1 (UBQLN1) is an essential factor for the maintenance of proteostasis in cells. It is important for the regulation of different protein degradation mechanisms, including the ubiquitin-proteasome system, autophagy and endoplasmic reticulum-associated protein degradation pathways. However, the role of UBQLN1 in cancer progression remains largely unknown. In the present study, the expression, functions and molecular mechanisms of UBQLN1 in breast cancer tissue samples and cell lines were explored. Immunohistochemical and bioinformatics analyses revealed that UBQLN1 expression was significantly upregulated in breast cancer tissues and cell lines. UBQLN1 expression in breast cancer was significantly associated with lymph node metastasis and TNM stage. Moreover, a high UBQLN1 expression was a predictor of an unfavorable survival in patients with breast cancer. In vitro, UBQLN1 silencing markedly inhibited cell migration and invasion, epithelial-to-mesenchymal transition (EMT) and MMP expression. UBQLN1 silencing attenuated the stem cell-like properties of breast cancer cells, including their mammosphere-forming abilities. UBQLN1 knockdown also enhanced breast cancer cell chemosensitivity to paclitaxel. The expression levels of the stem cell markers. Aldehyde dehydrogenase 1 (ALDH1), Oct-4 and Sox2 were significantly decreased in the cells in which UBQLN1 was silenced, whereas breast cancer stem cells exhibited an increased expression of UBQLN1. Mechanistically, UBQLN1 knockdown inhibited the activation of AKT signaling, as revealed by the increased PTEN expression and the decreased expression of phosphorylated AKT in cells in which UBQLN1 was silenced. On the whole, the present study demonstrates that UBQLN1 is aberrantly upregulated in breast cancer and predicts a poor prognosis. The silencing of UBQLN1 inhibited the invasion, EMT and stemness of breast cancer cells, possibly via AKT signaling.
Collapse
Affiliation(s)
- Xiaoyue Feng
- Department of Pathology and Forensic Medicine, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Anna Cao
- Department of Pathology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310000, P.R. China
| | - Tao Qin
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Qingqing Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Shujun Fan
- Department of Pathology and Forensic Medicine, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Bo Wang
- Department of Pathology and Forensic Medicine, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Bo Song
- Department of Pathology and Forensic Medicine, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xiaotang Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Lianhong Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
7
|
DIAPH2, PTPRD and HIC1 Gene Polymorphisms and Laryngeal Cancer Risk. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147486. [PMID: 34299935 PMCID: PMC8305316 DOI: 10.3390/ijerph18147486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/26/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022]
Abstract
AIM, DIAPH2, PTPRD and HIC1 are the cell glycoprotein, which play an important role in the occurrence and development of tumors. This study was designed to assess the association between DIAPH2, PTPRD and HIC1 SNPs and laryngeal cancer risk. PATIENTS AND METHODS: This study including 267 patients with histologically confirmed laryngeal cancer and 157 controls. The relationship between genetic variations DIAPH2 (rs6620138), PTPRD (rs3765142) and HIC1 (rs9901806) and the onset of laryngeal cancer were investigated. Statistical analysis to calculate the relationship between DIAPH2, PTPRD and HIC1 genes polymorphism and pathogenesis of laryngeal cancer. RESULTS: The results showed that rs6620138 DIAPH2 polymorphism could increase the onset risk of laryngeal cancer. Statistically significant differences in allele distribution of rs6620138 DIAPH2 and rs9901806 HIC1 in the case and control groups subgroups. CONCLUSIONS: This study results suggested that genetic variation of rs6620138 DIAPH2 polymorphism is related to the susceptibility to laryngeal cancer. Our results provide a basis to begin basic research on the role of DIAPH2 gene in the pathogenesis of laryngeal cancer.
Collapse
|
8
|
Huang JW, Jiang X, Li ZL, Jiang CR. MicroRNA-328-5p Alleviates Macrophage Lipid Accumulation through the Histone Deacetylase 3/ATP-binding cassette transporter A1 pathway. Lipids 2021; 56:301-311. [PMID: 33663010 DOI: 10.1002/lipd.12297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/19/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022]
Abstract
MicroRNA-328 (miR-328) was reported to protect against atherosclerosis, but its role in foam cell formation remains unknown. The aim of this study was to investigate the effect of miR-328-5p on macrophage lipid accumulation and the underlying mechanisms. The results showed that miR-328-5p expression was robustly decreased in oxidized low-density lipoprotein (ox-LDL)-treated macrophages. Treatment of human acute monocytic leukemia cel (THP-1) macrophage-derived foam cells with a miR-328-5p mimic markedly increased [3 H]-cholesterol efflux, inhibited lipid droplet accumulation, and decreased intracellular total cholesterol (TC), free cholesterol (FC) and cholesteryl ester (CE) contents. Upregulation of miR-328-5p also reduced the expression of histone deacetylase 3 (HDAC3) but increased the levels of ATP-binding cassette transporter A1 (ABCA1) in THP-1 macrophage-derived foam cells. Mechanistically, miR-328-5p inhibited HDAC3 expression by directly targeting its 3'UTR, thereby promoting ABCA1 expression and the subsequent cholesterol efflux. Furthermore, miR-328-5p mimic treatment did not affect the uptake of Dil-ox-LDL or the expression of scavenger receptor-A (SR-A), thrombospondin receptor (CD36) and ABCG1. Taken together, these findings suggest that miR-328-5p alleviates macrophage lipid accumulation through the HDAC3/ABCA1 pathway.
Collapse
Affiliation(s)
- Jiang-Wei Huang
- Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Xin Jiang
- Department of Emergency, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Zi-Li Li
- Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Chang-Rong Jiang
- Department of Cardiology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|