1
|
Toribio D, Morokuma J, Pellino D, Hardt M, Zoukhri D. Quantitative Changes in the Proteome of Chronically Inflamed Lacrimal Glands From a Sjögren's Disease Animal Model. Invest Ophthalmol Vis Sci 2025; 66:44. [PMID: 40244610 PMCID: PMC12013672 DOI: 10.1167/iovs.66.4.44] [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] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose The lacrimal gland (LG) is the major source of aqueous tears, and insufficient LG secretion leads to aqueous-deficient dry eye (ADDE) disease. To provide a foundational description of LG's protein expression patterns, we prepared protein extracts of LGs from a wild-type and an ADDE mouse model and analyzed the proteome by quantitative mass spectrometry. Methods LGs were isolated from an ADDE mouse model, male non-obese diabetic (NOD) mice and control wild-type BALB/c mice (n = 6 each). Protein samples were prepared in urea-based lysis buffer and protein concentrations determined by the BCA method. The equivalent of 200 µg protein were tryptically digested and analyzed by nanoflow liquid chromatography tandem mass spectrometry (LC-MS/MS). Proteins were identified and quantified using the PEAKS X bioinformatics suite. Downstream differential protein expression analysis was performed using the MS-DAP R package. Selected significantly differentially expressed and detected proteins were subjected to spatial expression analysis using immunohistochemistry. Results Cumulatively, the LC-MS/MS-based proteomics analyses of the murine LG samples identified a total of 31,932 peptide sequences resulting in 2617 protein identifications at a 1% false discovery rate at the peptide and protein level. Principal component analysis (PCA) and hierarchical cluster analysis revealed a separation of NOD and BALB/c samples. Overall, protein diversity was consistently higher in NOD samples. After applying global peptide filter criteria and peptide-to-protein rollup, 1750 remaining proteins were subjected to differential expression analysis using the MSqRob algorithm, which identified 580 proteins with statistically significant expression differences. Data are available via ProteomeXchange with identifier PXD060937. At the cellular level, the up- and downregulation of select proteins were confirmed by immunohistochemistry. Conclusions Our data suggest that chronic inflammation leads to significant alterations in the LG proteome. Ongoing studies aim to identify potentially unique, inflammation-induced proteins that could be amenable to pharmacological modulation.
Collapse
Affiliation(s)
- Danny Toribio
- Department of Basic and Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, Massachusetts, United States
| | - Junji Morokuma
- Department of Basic and Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, Massachusetts, United States
| | - Dante Pellino
- Department of Basic and Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, Massachusetts, United States
| | - Markus Hardt
- Center for Salivary Diagnostics, ADA Forsyth Institute, Cambridge, Massachusetts, United States
- Department of Inflammation and Immunology, ADA Forsyth Institute, Cambridge, Massachusetts, United States
| | - Driss Zoukhri
- Department of Basic and Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, Massachusetts, United States
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
2
|
Liang H, Cao Z, Ren Y, Li Y, Wang H, Sun F, Xue M, Zhu G, Zhou Y. Raman spectroscopy and bioinformatics-based identification of key genes and pathways capable of distinguishing between diffuse large B cell lymphoma and chronic lymphocytic leukemia. Front Immunol 2025; 16:1516946. [PMID: 40070829 PMCID: PMC11893875 DOI: 10.3389/fimmu.2025.1516946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) and chronic lymphocytic leukemia (CLL) are subtypes of non-Hogkin lymphoma (NHL) that are generally distinct form one cases, but the transformation of one of these diseases into the other is possible. Some patients with CLL, for instance, have the potential to develop Richter transformation such that they are diagnosed with a rare, invasive DLBCL subtype. In this study, bioinformatics analyses of these two NHL subtypes were conducted, identifying key patterns of gene expression and then experimentally validating the results. Disease-related gene expression datasets from the GEO database were used to identify differentially expressed genes (DEGs) and DEG functions were examined using GO analysis and protein-protein interaction network construction. This strategy revealed many up- and down-regulated DEGs, with functional enrichment analyses identifying these genes as being closely associated with inflammatory and immune response activity. PPI network analyses and the evaluation of clustered network modules indicated the top 10 up- and down-regulated genes involved in disease onset and development. Serological analyses revealed significantly higher ALB, TT, and WBC levels in CLL patients relative to DLBCL patients, whereas the opposite was true with respect to TG, HDL, GGT, ALP, ALT, and NEUT% levels. In comparison to the CLL and DLBCL groups, the healthy control samples demonstrated higher signals of protein peak positions (621, 643, 848, 853, 869, 935, 1003, 1031, 1221, 1230, 1260, 1344, 1443, 1446, 1548, 1579, 1603, 1647 cm-1), nucleic acid peak positions (726, 781, 786, 1078, 1190, 1415, 1573, 1579 cm-1), beta carotene peak positions (957, 1155, 1162 cm-1), carbohydrate peak positions (842 cm-1), collagen peak positions (1345 cm-1), and lipid peak positions (957, 1078, 1119, 1285, 1299, 1437, 1443, 1446 cm-1) compared to the CLL and DLBCL groups. Verification of these key genes in patient samples yielded results consistent with findings derived from bioinformatics analyses, highlighting their relevance to diagnosing and treating these forms of NHL. Together, these analyses identified genes and pathways involved in both DLBCL and CLL. The set of molecular markers established herein can aid in patient diagnosis and prognostic evaluation, providing a valuable foundation for their therapeutic application.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Computational Biology/methods
- Protein Interaction Maps
- Gene Expression Profiling
- Diagnosis, Differential
- Biomarkers, Tumor/genetics
- Gene Regulatory Networks
- Gene Expression Regulation, Neoplastic
- Female
- Male
Collapse
Affiliation(s)
- Haoyue Liang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhijie Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yansong Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yihan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Haoyu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Fanfan Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mei Xue
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guoqing Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
3
|
Macias-Díaz A, Nieto-Felipe J, Jardín I, Camello PJ, Martinez-Quintana EM, Salido GM, Smani T, Lopez JJ, Rosado JA. Filamin A C-terminal fragment modulates Orai1 expression by inhibition of protein degradation. Am J Physiol Cell Physiol 2025; 328:C657-C669. [PMID: 39764579 DOI: 10.1152/ajpcell.00745.2024] [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: 10/08/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/05/2025]
Abstract
Filamin A (FLNA) is an actin-binding protein that has been reported to interact with STIM1 modulating the activation of Orai1 channels. Cleaving of FLNA by calpain leads to a C-terminal fragment that is involved in a variety of functional and pathological events, including pro-oncogenic activity in different types of cancer. Here, we show that full-length FLNA is downregulated in samples from patients with colon cancer as well as in the adenocarcinoma cell line HT-29. This is consistent with an increased calpain-dependent FLNA cleaving with enhanced expression of the C-terminal FLNA fragment accompanied by enhanced expression of Orai1 and STIM1, as well as store-operated Ca2+ entry (SOCE). To further explore the mechanism underlying the enhancement of SOCE by the C-terminal FLNA fragment, we expressed in HEK-293 cells the C-terminal FLNA region encompassing repeats 16-24 (FLNA16-24 fragment), which enhanced both Orai1 and STIM1 as well as SOCE. Transfection of the FLNA16-24 fragment attenuates Orai1 and STIM1 protein degradation, and, specifically, abrogates Orai1α lysosomal degradation and retains this channel in the plasma membrane. However, the C-terminal FLNA fragment did not induce a detectable modification in Orai1β degradation. Due to the relevance of SOCE in cell physiology, our results provide evidence of a novel mechanism for the regulation of Ca2+ influx with relevant pathophysiological implications.NOTE & NOTEWORTHY FLNA cleaving by calpain has been observed in a variety of tumoral, including prostate and colorectal cancer cells, as well as in nontumoral cells, leading to a C-terminal fragment encompassing repeats 16-24. Expression of the FLNA16-24 fragment in HEK-293 cells enhances Orai1 and STIM1 expression, as well as SOCE, a mechanism mediated by attenuation of Orai1α and STIM1 degradation, providing evidence for a novel mechanism for the regulation of SOCE in normal and malignant cells.
Collapse
Affiliation(s)
- Alvaro Macias-Díaz
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Joel Nieto-Felipe
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Isaac Jardín
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Pedro J Camello
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | | | - Gines M Salido
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, Seville, Spain
| | - Jose J Lopez
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| |
Collapse
|
4
|
Fan H, Tan X, Xu S, Zeng Y, Zhang H, Shao T, Zhao R, Zhou P, Bo X, Fan J, Fu Y, Ding X, Zhou Y. Identification and validation of differentially expressed disulfidptosis-related genes in hypertrophic cardiomyopathy. Mol Med 2024; 30:249. [PMID: 39701955 DOI: 10.1186/s10020-024-01024-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most common cardiovascular diseases with no effective treatment due to its complex pathogenesis. A novel cell death, disulfidptosis, has been extensively studied in the cancer field but rarely in cardiovascular diseases. This study revealed the potential relationship between disulfidptosis and hypertrophic cardiomyopathy and put forward a predictive model containing disulfidptosis-associated genes (DRGs) of GYS1, MYH10, PDMIL1, SLC3A2, CAPZB, showing excellent performance by SVM machine learning model. The results were further validated by western blot, RNA sequencing and immunohistochemistry in a TAC mice model. In addition, resveratrol was selected as a therapeutic drug targeting core genes using the CTD database. In summary, this study provides new perspectives for exploring disulfidptosis-related biomarkers and potential therapeutic targets for hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Huimin Fan
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Xin Tan
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China
- Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Shuai Xu
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China
- Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Yiyao Zeng
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China
- Institute for Hypertension, Soochow University, Suzhou, 215000, China
| | - Hailong Zhang
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Tong Shao
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Runze Zhao
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Peng Zhou
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China
| | - Xiaohong Bo
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Jili Fan
- Department of Cardiovascular Disease, Taihe County People's Hospital, Fuyang, 236600, China
| | - Yangjun Fu
- Department of Neurology, The Third People's Hospital of Hefei, Hefei City, Anhui Province, 230041, China
| | - Xulong Ding
- Center of Translational Medicine and Clinical Laboratory, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital to Soochow University, Suzhou, 215000, China.
| | - Yafeng Zhou
- Department of Cardiology, Suzhou Dushu Lake Hospital, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, 215000, China.
- Institute for Hypertension, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
5
|
Liu Y, Wu Y, Wang C, Hu W, Zou S, Ren H, Zuo Y, Qu L. MiR-127-3p enhances macrophagic proliferation via disturbing fatty acid profiles and oxidative phosphorylation in atherosclerosis. J Mol Cell Cardiol 2024; 193:36-52. [PMID: 38795767 DOI: 10.1016/j.yjmcc.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/05/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic pathology, leading to acute coronary heart disease or stroke. MiR-127 has been found significantly upregulated in advanced atherosclerosis. But its function in atherosclerosis remains unexplored. We explored the role of miR-127-3p in regulating atherosclerosis development and its downstream mechanisms. METHODS The expression profile of miR-127 in carotid atherosclerotic plaques of 23 patients with severe carotid stenosis was detected by RT-qPCR and in situ hybridization. Primary bone marrow-derived macrophages (BMDM) stimulated with oxidized low-density lipoprotein were used as an in vitro model. CCK-8, EdU, RT-qPCR, and flow cytometry were used to detect the proliferative capacity and polarization of BMDM, which were infected by lentivirus-carrying plasmid to upregulate or downregulate miR-127-3p expression, respectively. RNA sequencing combined with bioinformatic analysis and targeted fatty acid metabolomics approach were used to detect the transcriptome and lipid metabolites. The association between miR-127-3p and its target was verified by dual-luciferase activity reporting and Western blotting. Oxygen consumption rate of BMDM were detected using seahorse analysis. High-cholesterol-diet-fed low density lipoprotein deficient (LDLR-/-) mice, with-or-without carotid tandem-stenosis surgery, were treated with miR-127-3p agomir or antagomir to examine its effect on plaque development and stability. RESULTS miR-127-3p, not -5p, is elevated in human advanced carotid atheroma and its expression is positively associated with macrophage accummulation in plaques. In vitro, miR-127-3p-overexpressed macrophage exhibites increased proliferation capacity and facilitates M1 polariztion whereas the contrary trend is present in miR-127-3p-inhibited macrophage. Stearoyl-CoA desaturase-1 (SCD1) is one potential target of miR-127-3p. miR-127-3p mimics decreases the activity of 3' untranslated regions of SCD-1. Furthermore, miR-127-3p downregulates SCD1 expression, and reversing the expression of SCD1 attenuates the increased proliferation induced by miR-127-3p overexpression in macrophage. miR-127-3p overexpression could also lead to decreased content of unsaturated fatty acids (UFAs), increased content of acetyl CoA and increased level of oxidative phosphorylation. In vivo, miR-127-3p agomir significantly increases atherosclerosis progression, macrophage proliferation and decreases SCD1 expression and the content of UFAs in aortic plaques of LDLR-/- mice. Conversely, miR-127-3p antagomir attenuated atherosclerosis, macrophage proliferation in LDLR-/- mice, and enhanced carotid plaque stability in mice with vulnerable plaque induced. CONCLUSION MiR-127-3p enhances proliferation in macrophages through downregulating SCD-1 expression and decreasing the content of unsaturated fatty acid, thereby promoting atherosclerosis development and decreasing plaque stability. miR-127-3p/SCD1/UFAs might provide potential therapeutic target for anti-inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Yandong Liu
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China; Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Yicheng Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Chao Wang
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Weilin Hu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Sili Zou
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China
| | - Huiqiong Ren
- Department of Geriatrics, 905th Hospital of PLA NAVY, Shanghai, China.
| | - Yong Zuo
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
6
|
Wang S, Gong X, Xiao F, Yang Y. Recent advances in host-focused molecular tools for investigating host-gut microbiome interactions. Front Microbiol 2024; 15:1335036. [PMID: 38605718 PMCID: PMC11007152 DOI: 10.3389/fmicb.2024.1335036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Microbial communities in the human gut play a significant role in regulating host gene expression, influencing a variety of biological processes. To understand the molecular mechanisms underlying host-microbe interactions, tools that can dissect signaling networks are required. In this review, we discuss recent advances in molecular tools used to study this interplay, with a focus on those that explore how the microbiome regulates host gene expression. These tools include CRISPR-based whole-body genetic tools for deciphering host-specific genes involved in the interaction process, Cre-loxP based tissue/cell-specific gene editing approaches, and in vitro models of host-derived organoids. Overall, the application of these molecular tools is revolutionizing our understanding of how host-microbiome interactions contribute to health and disease, paving the way for improved therapies and interventions that target microbial influences on the host.
Collapse
Affiliation(s)
- Siyao Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Xu Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yun Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| |
Collapse
|
7
|
Miyazaki T. Calpain and Cardiometabolic Diseases. Int J Mol Sci 2023; 24:16782. [PMID: 38069105 PMCID: PMC10705917 DOI: 10.3390/ijms242316782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Calpain is defined as a member of the superfamily of cysteine proteases possessing the CysPC motif within the gene. Calpain-1 and -2, which are categorized as conventional isozymes, execute limited proteolysis in a calcium-dependent fashion. Accordingly, the calpain system participates in physiological and pathological phenomena, including cell migration, apoptosis, and synaptic plasticity. Recent investigations have unveiled the contributions of both conventional and unconventional calpains to the pathogenesis of cardiometabolic disorders. In the context of atherosclerosis, overactivation of conventional calpain attenuates the barrier function of vascular endothelial cells and decreases the immunosuppressive effects attributed to lymphatic endothelial cells. In addition, calpain-6 induces aberrant mRNA splicing in macrophages, conferring atheroprone properties. In terms of diabetes, polymorphisms of the calpain-10 gene can modify insulin secretion and glucose disposal. Moreover, conventional calpain reportedly participates in amino acid production from vascular endothelial cells to induce alteration of amino acid composition in the liver microenvironment, thereby facilitating steatohepatitis. Such multifaceted functionality of calpain underscores its potential as a promising candidate for pharmaceutical targets for the treatment of cardiometabolic diseases. Consequently, the present review highlights the pivotal role of calpains in the complications of cardiometabolic diseases and embarks upon a characterization of calpains as molecular targets.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Tokyo 142-8555, Japan
| |
Collapse
|
8
|
Wang Y, Li M, Chen J, Yu Y, Yu Y, Shi H, Liu X, Chen Z, Chen R, Ge J. Macrophage CAPN4 regulates CVB3-induced cardiac inflammation and injury by promoting NLRP3 inflammasome activation and phenotypic transformation to the inflammatory subtype. Free Radic Biol Med 2023; 208:430-444. [PMID: 37660839 DOI: 10.1016/j.freeradbiomed.2023.08.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023]
Abstract
Exploring the immune mechanism of coxsackievirus B3 (CVB3)-induced myocarditis may provide a promising therapeutic strategy. Here, we investigated the regulatory role of macrophage CAPN4 in the phenotypic transformation of macrophages and NOD-like receptor protein 3 (NLRP3) inflammasome activation. We found that CAPN4 was the most upregulated subtype of the calpain family in CVB3-infected bone marrow-derived macrophages (BMDMs) and Raw 264.7 cells after CVB3 infection and was upregulated in cardiac macrophages from CVB3-infected mice. Conditional knockout of CAPN4 (CAPN4flox/flox; LYZ2-Cre, CAPN4-cKO mice) ameliorated inflammation and myocardial injury and improved cardiac function and survival after CVB3 infection. Enrichment analysis revealed that macrophage differentiation and the interleukin signaling pathway were the most predominant biological processes in macrophages after CVB3 infection. We further found that CVB3 infection and the overexpression of CAPN4 promoted macrophage M1 polarization and NLRP3 inflammasome activation, while CAPN4 knockdown reversed these changes. Correspondingly, CAPN4-cKO alleviated CVB3-induced M1 macrophage transformation and NLRP3 expression and moderately increased M2 transformation in vivo. The culture supernatant of CAPN4-overexpressing or CVB3-infected macrophages impaired cardiac fibroblast function and viability. Moreover, macrophage CAPN4 could upregulate C/EBP-homologous protein (chop) expression, which increased proinflammatory cytokine release by activating the phosphorylation of transducer of activator of transcription 1 (STAT1) and 3 (STAT3). Overall, these results suggest that CAPN4 increases M1-type and inhibits M2-type macrophage polarization through the chop-STAT1/STAT3 signaling pathway to mediate CVB3-induced myocardial inflammation and injury. CAPN4 may be a novel target for viral myocarditis treatment.
Collapse
Affiliation(s)
- Yucheng Wang
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Minghui Li
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Jun Chen
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Ying Yu
- Department of General Practice, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Yong Yu
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Hui Shi
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Xiaoxiao Liu
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Zhiwei Chen
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| | - Ruizhen Chen
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China.
| | - Junbo Ge
- Key Laboratory of Viral Cardiovascular Diseases, Ministry of Health, China & Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Medical College of Fudan University, Xuhui District, Shanghai, 200010, China
| |
Collapse
|
9
|
Qin R, Huang L, Xu W, Qin Q, Liang X, Lai X, Huang X, Xie M, Chen L. Identification of disulfidptosis-related genes and analysis of immune infiltration characteristics in ischemic strokes. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:18939-18959. [PMID: 38052584 DOI: 10.3934/mbe.2023838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Immune infiltration plays a pivotal role in the pathogenesis of ischemic stroke. A novel form of cell death known as disulfidptosis has emerged in recent studies. However, there is currently a lack of research investigating the regulatory mechanism of disulfidptosis-related genes in immune infiltration during ischemic stroke. Using machine learning methods, we identified candidate key disulfidptosis-related genes (DRGs). Subsequently, we performed an analysis of immune cell infiltration to investigate the dysregulation of immune cells in the context of ischemic stroke. We assessed their diagnostic value by employing receiver operating characteristic (ROC) curves. To gain further insights, we conducted functional enrichment analyses to elucidate the signaling pathways associated with these seven DRGs. We identified two distinct subclusters based on the expression patterns of these seven DRGs. The unique roles of these subclusters were further evaluated through KEGG analysis and immune infiltration studies. Furthermore, we validated the expression profiles of these seven DRGs using both single-cell datasets and external datasets. Lastly, molecular docking was performed to explore potential drugs for the treatment of ischemic stroke. We identified seven DRGs. The seven DRGs are related to immune cells. Additionally, these seven DRGs also demonstrate potential diagnostic value in ischemic stroke. Functional enrichment analysis highlighted pathways such as platelet aggregation and platelet activation. Two subclusters related to disulfidptosis were defined, and functional enrichment analysis of their differentially expressed genes (DEGs) primarily involved pathways like cytokine-cytokine receptor interaction. Single-cell analysis indicated that these seven DRGs were primarily distributed among immune cell types. Molecular docking results suggested that genistein might be a potential therapeutic drug. This study has opened up new avenues for exploring the causes of ischemic stroke and developing potential therapeutic targets.
Collapse
Affiliation(s)
- Rongxing Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Lijuan Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Wei Xu
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Qingchun Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Xiaojun Liang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xinyu Lai
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xiaoying Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Minshan Xie
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Li Chen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
10
|
Shi Y, Qiu T, Wu C, Yuan W, Yao X, Jiang L, Wang N, Wang L, Han Q, Yang G, Liu X, Sun X. Filamin A facilitates NLRP3 inflammasome activation during arsenic-induced nonalcoholic steatohepatitis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:107703-107715. [PMID: 37740811 DOI: 10.1007/s11356-023-29702-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 08/31/2023] [Indexed: 09/25/2023]
Abstract
Prolonged exposure to arsenic can cause nonalcoholic steatohepatitis (NASH). The NOD-like receptor protein 3 (NLRP3) inflammasome plays an essential role in the process of NASH. However, the mechanism by which arsenic promotes NLRP3 expression remains unclear. Three-month NaAsO2 gavage led to the nuclear factor-κB (NF-κB) signaling pathway activation and NASH. Additionally, NaAsO2 upregulated the level of Filamin A (FLNA) and pyroptosis, thereby activating the NLRP3 inflammasome in SD rat liver. Using FLNA siRNA, NASH-associated inflammation and pyroptosis were clearly mitigated by reducing activation of the NLRP3 inflammasome. Furthermore, arsenic treatment facilitated activation of the NF-κB signaling pathway and promoted p-p65 translocation into the nucleus. Chromatin immunoprecipitation (Ch-IP) assay indicated that FLNA promoted p65 binding to the NLRP3 gene and upregulated the transcription of NLRP3, ultimately leading to pyroptosis and NASH. Our findings indicate that FLNA and pyroptosis are strongly associated with NASH induced by NaAsO2. Collectively, the findings of this study indicated that FLNA mediates NF-κB signaling pathway-induced activation of the NLRP3 inflammasome and ultimately activates pyroptosis and NASH upon NaAsO2 exposure. This information may be useful for improving therapeutic strategies against arsenic-induced NASH.
Collapse
Affiliation(s)
- Yan Shi
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
- Xi'an Center for Disease Control and Prevention, No. 599 Xiying Road, Xi'an, 710000, People's Republic of China
| | - Tianming Qiu
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Chenbing Wu
- Preventive Medicine Laboratory, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Weizhuo Yuan
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Xiaofeng Yao
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Liping Jiang
- Preventive Medicine Laboratory, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Ningning Wang
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Lu Wang
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Qiuyue Han
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Guang Yang
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Xiaofang Liu
- Department of Nutrition and Food Safety, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China
| | - Xiance Sun
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China.
- Global Health Research Center, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, People's Republic of China.
| |
Collapse
|
11
|
Bi L, Wacker BK, Komandur K, Sanford N, Dichek DA. Apolipoprotein A-I vascular gene therapy reduces vein-graft atherosclerosis. Mol Ther Methods Clin Dev 2023; 30:558-572. [PMID: 37693942 PMCID: PMC10482902 DOI: 10.1016/j.omtm.2023.08.018] [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: 04/21/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023]
Abstract
Coronary artery venous bypass grafts typically fail because of atherosclerosis driven by lipid and macrophage accumulation. Therapy for vein-graft atherosclerosis is limited to statin drugs, which are only modestly effective. We hypothesized that transduction of vein-graft endothelium of fat-fed rabbits with a helper-dependent adenovirus expressing apolipoprotein AI (HDAdApoAI) would reduce lipid and macrophage accumulation. Fat-fed rabbits received bilateral external jugular vein-to-carotid artery interposition grafts. Four weeks later, one graft per rabbit (n = 23 rabbits) was infused with HDAdApoAI and the contralateral graft with HDAdNull. Grafts were harvested 12 weeks later. Paired analyses of grafts were performed, with vein graft cholesterol, intimal lipid, and macrophage content as the primary endpoints. HDAd genomes were detected in all grafts. APOAI mRNA was median 63-fold higher in HDAdApoAI grafts versus HDAdNull grafts (p < 0.001). HDAdApoAI grafts had a mean 15% lower total cholesterol (by mass spectrometry; p = 0.003); mean 19% lower intimal lipid (by oil red O staining; p = 0.02); and mean 13% lower expression of the macrophage marker CD68 (by reverse transcriptase-mediated quantitative PCR; p = 0.008). In vivo transduction of vein-graft endothelium achieves persistent APOAI expression and reduces vein-graft cholesterol, intimal lipid, and CD68 expression. Vascular gene therapy with APOAI has promise for preventing vein-graft failure caused by atherosclerosis.
Collapse
Affiliation(s)
- Lianxiang Bi
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Bradley K. Wacker
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Kaushik Komandur
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Nicole Sanford
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - David A. Dichek
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| |
Collapse
|
12
|
Li XY, Kuang DD, Guo AJ, Deng YY, Pan LH, Li QM, Luo JP, Zha XQ. Inhibition of Ca 2+-calpain signaling is a new mechanism using Laminaria japonica polysaccharide to prevent macrophage foam cell formation and atherosclerosis. Food Funct 2023; 14:4036-4048. [PMID: 37067393 DOI: 10.1039/d2fo04099a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The Ca2+-calpain signaling plays a pivotal role in regulating the upstream signaling pathway of cellular autophagy. The aim of the current work was to investigate the role of Ca2+-calpain signaling in the regulation of macrophage autophagy by a Laminaria japonica polysaccharide (LJP61A) in Ox-LDL induced macrophages and high fat diet fed atherosclerotic mice. Results revealed that the LJP61A markedly decreased the levels of intracellular Ca2+, calpain1, calpain2 and their downstream effectors (Gsα, cAMP and IP3), and simultaneously enhanced autophagy activity and lipid metabolism, thereby reducing lipid accumulation in the Ox-LDL stimulated macrophages and lipid-laden plaques in atherosclerotic mice. Moreover, BAPTA-AM (a Ca2+ chelator) and calpeptin (a calpain inhibitor) synergistically strengthened the beneficial effects of LJP61A on autophagy and lipid metabolism by decreasing the levels of intracellular Ca2+, calpain1, calpain2, and their downstream effectors (Gsα, cAMP and IP3) induced by Ox-LDL. These findings suggested that the LJP61A suppressed macrophage derived foam cell formation and atherosclerosis by modulating the Ca2+-calpain-mediated autophagy.
Collapse
Affiliation(s)
- Xue-Ying Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| | - Dan-Dan Kuang
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| | - An-Jun Guo
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| | - Yuan-Yuan Deng
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences, Guangzhou 510610, People's Republic of China
- Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangzhou 510610, People's Republic of China
- Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, People's Republic of China
| | - Li-Hua Pan
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| | - Qiang-Ming Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| | - Jian-Ping Luo
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| | - Xue-Qiang Zha
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
- Key Laboratory of Metabolism and Regulation for Major Disease of Anhui Higher Education Institutes, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| |
Collapse
|
13
|
Lu Y, Wang M, Zhao M, Zhang Q, Qian R, Hu Z, Ke Q, Yu L, Wang L, Lai Q, Liu Z, Jiang X, Zhang B, Yang J, Yao Y. Filamin A is overexpressed in non-alcoholic steatohepatitis and contributes to the progression of inflammation and fibrosis. Biochem Biophys Res Commun 2023; 653:93-101. [PMID: 36863213 DOI: 10.1016/j.bbrc.2023.02.048] [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: 02/08/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic and progressive liver disease characterized by steatosis, inflammation, and fibrosis. Filamin A (FLNA), an actin-binding protein, is involved in various cell functions, including the regulation of immune cells and fibroblasts. However, its role in the development of NASH through inflammation and fibrogenesis is not fully understood. In this study, we found that FLNA expression was increased in liver tissues of patients with cirrhosis and mice with non-alcoholic fatty liver disease (NAFLD)/NASH and fibrosis. Immunofluorescence analysis showed that FLNA was primarily expressed in macrophages and hepatic stellate cells (HSCs). Knocking down of FLNA by specific shRNA in phorbol-12-myristate-13-acetate (PMA)-derived THP-1 macrophages reduced lipopolysaccharide (LPS)-stimulated inflammatory response. The decreased mRNA levels of inflammatory cytokines and chemokines and suppression of the STAT3 signaling were observed in FLNA-downregulated macrophages. In addition, knockdown of FLNA in immortalized human hepatic stellate cells (LX-2 cells) resulted in decreased mRNA levels of fibrotic cytokines and enzymes involved in collagen synthesis, as well as increased levels of metalloproteinases and pro-apoptotic proteins. Overall, these results suggest that FLNA may contribute to the pathogenesis of NASH through its role in the regulation of inflammatory and fibrotic mediators.
Collapse
Affiliation(s)
- Ying Lu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengzhu Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Manyu Zhao
- Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Qianru Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China; Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Qian
- Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Zan Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Qi Ke
- Department of Pathology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Lin Yu
- Department of Clinical Laboratory, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, 621000, China
| | - Liqun Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenmi Liu
- Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Xia Jiang
- Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Ben Zhang
- Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yuqin Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China; Molecular Toxicology Laboratory of Sichuan Provincial Education Office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Wang Z, Liu M, Lei H, Lei P, Liu X, Zhang J, Xiao S, Zheng Y, Feng YG. Serum Proteomics Combined with Metabolomics Analysis Explore the Molecular Biological Characteristics of Eruptive Syringoma. Clin Cosmet Investig Dermatol 2023; 16:17-26. [PMID: 36636632 PMCID: PMC9830079 DOI: 10.2147/ccid.s393620] [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: 11/04/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Eruptive syringoma (ES) is a clinical variant of the appendageal tumor syringoma. Around 75% of ES arise in the head or neck, which makes them unsightly. ES is common in patients with amyloidosis, diabetes, and Down's syndrome, suggesting that it may be associated with potential systemic effects. ES is a rare tumor with the unclear pathogenesis and no effective treatment. METHODS A PubMed search of ES was conducted. Plasma samples of patients with ES were acquired from the Department of Dermatology at Xi'an Jiaotong University's Second Affiliated Hospital. After removing highly abundant proteins, plasma samples were subjected to proteomics and metabolomics analysis using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS LC-MS/MS revealed 71 differentially expressed proteins and 18 differentially abundant metabolites. The functional analysis highlighted the importance of complement binding, coagulation, secretory granules and vesicle lumen. Further, the study revealed 15 hub genes associated with FGG, GC, APOE, FGA, FGB, C4A, C3, CRP, C4B, FLNA, TAGLN2, ANXA5, MYL6, MYL12B, and TLN1 organized into three clusters. The seed genes in each cluster were GC, FLNA, and MYL6. In addition, glycol metabolism was associated with variable abundance of serum metabolites, which explains the relatively high rate of ES among diabetics. CONCLUSION This study suggests that immunological inflammation and tumor glycol metabolism may play significant role in the pathophysiology of ES.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Dermatology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Meng Liu
- Department of Dermatology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Hao Lei
- Department of Dermatology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Panpan Lei
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi’an Jiaotong University School of Life Science and Technology, Xi’an, People’s Republic of China
| | - Xinyi Liu
- Department of Dermatology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jing Zhang
- Department of Dermatology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shengxiang Xiao
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yan Zheng
- Department of Dermatology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yi-Guo Feng
- Department of Dermatology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| |
Collapse
|
15
|
Ganekal P, Vastrad B, Vastrad C, Kotrashetti S. Identification of biomarkers, pathways, and potential therapeutic targets for heart failure using next-generation sequencing data and bioinformatics analysis. Ther Adv Cardiovasc Dis 2023; 17:17539447231168471. [PMID: 37092838 PMCID: PMC10134165 DOI: 10.1177/17539447231168471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Heart failure (HF) is the most common cardiovascular diseases and the leading cause of cardiovascular diseases related deaths. Increasing molecular targets have been discovered for HF prognosis and therapy. However, there is still an urgent need to identify novel biomarkers. Therefore, we evaluated biomarkers that might aid the diagnosis and treatment of HF. METHODS We searched next-generation sequencing (NGS) dataset (GSE161472) and identified differentially expressed genes (DEGs) by comparing 47 HF samples and 37 normal control samples using limma in R package. Gene ontology (GO) and pathway enrichment analyses of the DEGs were performed using the g: Profiler database. The protein-protein interaction (PPI) network was plotted with Human Integrated Protein-Protein Interaction rEference (HiPPIE) and visualized using Cytoscape. Module analysis of the PPI network was done using PEWCC1. Then, miRNA-hub gene regulatory network and TF-hub gene regulatory network were constructed by Cytoscape software. Finally, we performed receiver operating characteristic (ROC) curve analysis to predict the diagnostic effectiveness of the hub genes. RESULTS A total of 930 DEGs, 464 upregulated genes and 466 downregulated genes, were identified in HF. GO and REACTOME pathway enrichment results showed that DEGs mainly enriched in localization, small molecule metabolic process, SARS-CoV infections, and the citric acid tricarboxylic acid (TCA) cycle and respiratory electron transport. After combining the results of the PPI network miRNA-hub gene regulatory network and TF-hub gene regulatory network, 10 hub genes were selected, including heat shock protein 90 alpha family class A member 1 (HSP90AA1), arrestin beta 2 (ARRB2), myosin heavy chain 9 (MYH9), heat shock protein 90 alpha family class B member 1 (HSP90AB1), filamin A (FLNA), epidermal growth factor receptor (EGFR), phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), cullin 4A (CUL4A), YEATS domain containing 4 (YEATS4), and lysine acetyltransferase 2B (KAT2B). CONCLUSIONS This discovery-driven study might be useful to provide a novel insight into the diagnosis and treatment of HF. However, more experiments are needed in the future to investigate the functional roles of these genes in HF.
Collapse
Affiliation(s)
- Prashanth Ganekal
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. College of Pharmacy, Gadag, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, #253, Bharthinagar, Dharwad 580001, India
| | | |
Collapse
|
16
|
Abstract
RNA is not always a faithful copy of DNA. Advances in tools enabling the interrogation of the exact RNA sequence have permitted revision of how genetic information is transferred. We now know that RNA is a dynamic molecule, amenable to chemical modifications of its four canonical nucleotides by dedicated RNA-binding enzymes. The ever-expanding catalogue of identified RNA modifications in mammals has led to a burst of studies in the past 5 years that have explored the biological relevance of the RNA modifications, also known as epitranscriptome. These studies concluded that chemical modification of RNA nucleotides alters several properties of RNA molecules including sequence, secondary structure, RNA-protein interaction, localization and processing. Importantly, a plethora of cellular functions during development, homeostasis and disease are controlled by RNA modification enzymes. Understanding the regulatory interface between a single-nucleotide modification and cellular function will pave the way towards the development of novel diagnostic, prognostic and therapeutic tools for the management of diseases, including cardiovascular disease. In this Review, we use two well-studied and abundant RNA modifications - adenosine-to-inosine RNA editing and N6-methyladenosine RNA methylation - as examples on which to base the discussion about the current knowledge on installation or removal of RNA modifications, their effect on biological processes related to cardiovascular health and disease, and the potential for development and application of epitranscriptome-based prognostic, diagnostic and therapeutic tools for cardiovascular disease.
Collapse
|
17
|
Shahinuzzaman ADA, Kamal AHM, Chakrabarty JK, Rahman A, Chowdhury SM. Identification of Inflammatory Proteomics Networks of Toll-like Receptor 4 through Immunoprecipitation-Based Chemical Cross-Linking Proteomics. Proteomes 2022; 10:proteomes10030031. [PMID: 36136309 PMCID: PMC9506174 DOI: 10.3390/proteomes10030031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/14/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptor 4 (TLR4) is a receptor on an immune cell that can recognize the invasion of bacteria through their attachment with bacterial lipopolysaccharides (LPS). Hence, LPS is a pro-immune response stimulus. On the other hand, statins are lipid-lowering drugs and can also lower immune cell responses. We used human embryonic kidney (HEK 293) cells engineered to express HA-tagged TLR-4 upon treatment with LPS, statin, and both statin and LPS to understand the effect of pro- and anti-inflammatory responses. We performed a monoclonal antibody (mAb) directed co-immunoprecipitation (CO-IP) of HA-tagged TLR4 and its interacting proteins in the HEK 293 extracted proteins. We utilized an ETD cleavable chemical cross-linker to capture weak and transient interactions with TLR4 protein. We tryptic digested immunoprecipitated and cross-linked proteins on beads, followed by liquid chromatography–mass spectrometry (LC-MS/MS) analysis of the peptides. Thus, we utilized the label-free quantitation technique to measure the relative expression of proteins between treated and untreated samples. We identified 712 proteins across treated and untreated samples and performed protein network analysis using Ingenuity Pathway Analysis (IPA) software to reveal their protein networks. After filtering and evaluating protein expression, we identified macrophage myristoylated alanine-rich C kinase substrate (MARCKSL1) and creatine kinase proteins as a potential part of the inflammatory networks of TLR4. The results assumed that MARCKSL1 and creatine kinase proteins might be associated with a statin-induced anti-inflammatory response due to possible interaction with the TLR4.
Collapse
Affiliation(s)
- A. D. A. Shahinuzzaman
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Pharmaceutical Sciences Research Division, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| | - Abu Hena Mostafa Kamal
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Advanced Technology Cores, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jayanta K. Chakrabarty
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Quantitative Proteomics and Metabolomics Center, Columbia University, New York, NY 10027, USA
| | - Aurchie Rahman
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Saiful M. Chowdhury
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Correspondence: ; Tel.: +1-817-272-5439
| |
Collapse
|
18
|
Stevens TW, Khalaf FK, Soehnlen S, Hegde P, Storm K, Meenakshisundaram C, Dworkin LD, Malhotra D, Haller ST, Kennedy DJ, Dube P. Dirty Jobs: Macrophages at the Heart of Cardiovascular Disease. Biomedicines 2022; 10:1579. [PMID: 35884884 PMCID: PMC9312498 DOI: 10.3390/biomedicines10071579] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the greatest public health concerns and is the leading cause of morbidity and mortality in the United States and worldwide. CVD is a broad yet complex term referring to numerous heart and vascular conditions, all with varying pathologies. Macrophages are one of the key factors in the development of these conditions. Macrophages play diverse roles in the maintenance of cardiovascular homeostasis, and an imbalance of these mechanisms contributes to the development of CVD. In the current review, we provide an in-depth analysis of the diversity of macrophages, their roles in maintaining tissue homeostasis within the heart and vasculature, and the mechanisms through which imbalances in homeostasis may lead to CVD. Through this review, we aim to highlight the potential importance of macrophages in the identification of preventative, diagnostic, and therapeutic strategies for patients with CVD.
Collapse
Affiliation(s)
- Travis W. Stevens
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Fatimah K. Khalaf
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
- Department of Clinical Pharmacy, University of Alkafeel, Najaf 54001, Iraq
| | - Sophia Soehnlen
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Prajwal Hegde
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Kyle Storm
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Chandramohan Meenakshisundaram
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Lance D. Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Deepak Malhotra
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Steven T. Haller
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - David J. Kennedy
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| | - Prabhatchandra Dube
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43606, USA; (T.W.S.); (F.K.K.); (S.S.); (P.H.); (K.S.); (C.M.); (L.D.D.); (D.M.); (S.T.H.)
| |
Collapse
|
19
|
Observation on the Efficacy of Ginkgo Ketone Ester Drop Pill in Improving Hypertension Combined with Carotid Atherosclerotic Plaque. Emerg Med Int 2022; 2022:8650537. [PMID: 35811610 PMCID: PMC9262540 DOI: 10.1155/2022/8650537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose. To observe and analyze the efficacy of Ginkgo ketone ester drop pill intervention in patients with hypertension combined with carotid atherosclerotic plaque. Methods. The subjects were 300 patients with hypertension complicated with carotid atherosclerotic plaque treated in our hospital from January 2019 to September 2021. The grouping was done by the random number table method and 300 patients were divided equally into 2 groups. One group was treated with Western medicine alone (clopidogrel sulfate tablets, phenyl amlodipine tablets, irbesartan tablets, and resorvastatin) as the Western medicine group (WM group, n = 150), and one group was added to this intervention with Ginkgo ketone ester drop pill as the Chinese medicine group (CM group, n = 150). The observation indexes were the improvement of blood pressure (systolic blood pressure (SBP) and diastolic blood pressure (DBP)), blood lipids (low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglyceride (TG), and total cholesterol (TC)), vascular endothelial function (nitric oxide (NO) and endothelin-1 (ET-1)), inflammatory factors (C-reactive protein (CRP) and interleukin-6 (IL-6)), plaque (intimal medial thickness (IMT) of carotid artery and plaque area), and efficacy after intervention and adverse effects during intervention in both groups. Results. After intervention, SBP, DBP, LDL-C, TG, and TC levels were lower and HDL-C levels were higher in both groups than before intervention in the same group, and both CM groups improved significantly compared with the WM group (
). After intervention, NO levels were higher and ET-1 levels were lower in both groups than before the intervention in the same group, and both CM groups improved significantly compared with the WM group (
). After intervention, CRP and IL-6 levels were lower in both groups than before intervention in the same group, and both CM groups improved significantly compared with the WM group (
). After intervention, IMT and plaque area were lower in both groups than before intervention in the same group and both CM groups improved significantly compared with the WM group (
). The total effective number of the CM group was better than the WM group (
), and there was no significant difference in the adverse reactions number in both groups (
). Conclusions. The treatment of hypertension combined with carotid atherosclerotic plaque with Ginkgo ketone ester drop pill helps to improve the blood pressure, blood lipid, and vascular endothelial function of patients and helps to inhibit the inflammation level and atherosclerotic plaque of patients, with significant efficacy and no significant adverse effects in patients, which is worthy of clinical promotion.
Collapse
|
20
|
Liu Q, Pan J, Bao L, Xu C, Qi Y, Jiang B, Wang D, Zhu X, Li X, Zhang H, Bai H, Yang Q, Ma J, Wiemer EAC, Ben J, Chen Q. Major Vault Protein Prevents Atherosclerotic Plaque Destabilization by Suppressing Macrophage ASK1-JNK Signaling. Arterioscler Thromb Vasc Biol 2022; 42:580-596. [PMID: 35387478 DOI: 10.1161/atvbaha.121.316662] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Macrophages are implicated in atherosclerotic plaque instability by inflammation and degradation of extracellular matrix. However, the regulatory mechanisms driving these macrophage-associated processes are not well understood. Here, we aimed to identify the plaque destabilization-associated cytokines and signaling pathways in macrophages. METHODS The atherosclerotic models of myeloid-specific MVP (major vault protein) knockout mice and control mice were generated. Atherosclerotic instability, macrophage inflammatory signaling, and active cytokines released by macrophages were examined in vivo and in vitro by using cellular and molecular biological approaches. RESULTS MVP deficiency in myeloid cells exacerbated murine plaque instability by increasing production of both MMP (matrix metallopeptidase)-9 and proinflammatory cytokines in artery wall. Mechanistically, expression of MMP-9 was mediated via ASK1 (apoptosis signal-regulating kinase 1)-MKK-4 (mitogen-activated protein kinase kinase 4)-JNK (c-Jun N-terminal kinase) signaling in macrophages. MVP and its α-helical domain could bind with ASK1 and inhibit its dimerization and phosphorylation. A 62 amino acid peptide (MVP-[686-747]) in the α-helical domain of MVP showed a crucial role in preventing macrophage MMP-9 production and plaque instability. CONCLUSIONS MVP may act as an inhibitor for ASK1-JNK signaling-mediated MMP-9 production in macrophages and, thereby, attenuate unstable plaque formation. Our findings suggest that suppression of macrophage ASK1-JNK signaling may be a useful strategy antagonizing atherosclerotic diseases.
Collapse
Affiliation(s)
- Qingling Liu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Junlu Pan
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Linrui Bao
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Chunxiang Xu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Yu Qi
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Bin Jiang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Dongdong Wang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Xudong Zhu
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Xiaoyu Li
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Hanwen Zhang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Hui Bai
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Qing Yang
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Junqing Ma
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Erik A C Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands (E.A.C.W.)
| | - Jingjing Ben
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| | - Qi Chen
- Department of Pathophysiology, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China (Q.L., J.P., L.B., C.X., Y.Q., B.J., D.W., X.Z., X.L., H.Z., H.B., Q.Y., J.M., J.B., Q.C.)
| |
Collapse
|
21
|
Guenther C. β2-Integrins - Regulatory and Executive Bridges in the Signaling Network Controlling Leukocyte Trafficking and Migration. Front Immunol 2022; 13:809590. [PMID: 35529883 PMCID: PMC9072638 DOI: 10.3389/fimmu.2022.809590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
Leukocyte trafficking is an essential process of immunity, occurring as leukocytes travel within the bloodstream and as leukocyte migration within tissues. While it is now established that leukocytes can utilize the mesenchymal migration mode or amoeboid migration mode, differences in the migratory behavior of leukocyte subclasses and how these are realized on a molecular level in each subclass is not fully understood. To outline these differences, first migration modes and their dependence on parameters of the extracellular environments will be explained, as well as the intracellular molecular machinery that powers migration in general. Extracellular parameters are detected by adhesion receptors such as integrins. β2-integrins are surface receptors exclusively expressed on leukocytes and are essential for leukocytes exiting the bloodstream, as well as in mesenchymal migration modes, however, integrins are dispensable for the amoeboid migration mode. Additionally, the balance of different RhoGTPases - which are downstream of surface receptor signaling, including integrins - mediate formation of membrane structures as well as actin dynamics. Individual leukocyte subpopulations have been shown to express distinct RhoGTPase profiles along with their differences in migration behavior, which will be outlined. Emerging aspects of leukocyte migration include signal transduction from integrins via actin to the nucleus that regulates DNA status, gene expression profiles and ultimately leukocyte migratory phenotypes, as well as altered leukocyte migration in tumors, which will be touched upon.
Collapse
Affiliation(s)
- Carla Guenther
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
22
|
MK2206 attenuates atherosclerosis by inhibiting lipid accumulation, cell migration, proliferation, and inflammation. Acta Pharmacol Sin 2022; 43:897-907. [PMID: 34316032 PMCID: PMC8976090 DOI: 10.1038/s41401-021-00729-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is a common comorbidity in patients with cancer, and the main leading cause of noncancer-related deaths in cancer survivors. Considering that current antitumor drugs usually induce cardiovascular injury, the quest for developing new antitumor drugs, especially those with cardiovascular protection, is crucial for improving cancer prognosis. MK2206 is a phase II clinical anticancer drug and the role of this drug in cardiovascular disease is still unclear. Here, we revealed that MK2206 significantly reduced vascular inflammation, atherosclerotic lesions, and inhibited proliferation of vascular smooth muscle cell in ApoE-/- mice in vivo. We demonstrated that MK2206 reduced lipid accumulation by promoting cholesterol efflux but did not affect lipid uptake and decreased inflammatory response by modulating inflammation-related mRNA stability in macrophages. In addition, we revealed that MK2206 suppressed migration, proliferation, and inflammation in vascular smooth muscle cells. Moreover, MK2206 inhibited proliferation and inflammation of endothelial cells. The present results suggest that MK2206, as a promising drug in clinical antitumor therapy, exhibits anti-inflammatory and antiatherosclerotic potential. This report provides a novel strategy for the prevention of cardiovascular comorbidities in cancer survivors.
Collapse
|
23
|
Bandaru S, Prajapati B, Juvvuna PK, Dosa S, Kogner P, Johnsen JI, Chandrasekhar K, Akyürek LM. Filamin A increases aggressiveness of human neuroblastomas. Neurooncol Adv 2022; 4:vdac028. [PMID: 35441138 PMCID: PMC9012446 DOI: 10.1093/noajnl/vdac028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abstract
Background
The actin-binding protein filamin A (FLNA) regulates oncogenic signal transduction important for tumor growth, but the role of FLNA in the progression of neuroblastoma (NB) has not been explored.
Methods
We analyzed FLNA mRNA expression in the R2 NB-database and FLNA protein expression in human NB tumors. We then silenced FLNA expression in human SKNBE2 and IMR32 NB cells by lentiviral vector encoding shRNA FLNA and assayed the cells for proliferation, migration, colony, spheroid formation, and apoptosis. SKNBE2 xenografts expressing or lacking FLNA in BALB/c nude mice were analyzed by both routine histopathology and immunohistochemistry.
Results
We observed shorter patient survival with higher expression of FLNA mRNA than patients with lower FLNA mRNA expression, and high-risk NB tumors expressed higher FLNA levels. SKNBE2 cells expressing higher FLNA levels proliferated more than IMR32 cells expressing lower FLNA levels. NB cell lines transfected with siRNA FLNA proliferated and migrated less, expressed lower levels of phosphorylated AKT and ERK1/2, formed smaller colonies and spheroids, as well as increased apoptosis. After inoculation of SKNBE2 cells infected with lentivirus expressing shRNA FLNA, size of NB tumors and number of proliferating cells were decreased. Furthermore, we identified STAT3 as an interacting partner of FLNA. Silencing FLNA mRNA reduced levels of phosphorylated STAT3 and MYCN, and induced expression NF-κB, cleaved caspase 3, and p53.
Conclusion
Inhibition of FLNA impaired NB cell signaling and function and reduced NB tumor size in vivo, suggesting that drugs targeting either FLNA or its interaction with STAT3 may be useful in the treatment of NB.
Collapse
Affiliation(s)
- Sashidar Bandaru
- Department of Laboratory Medicine, Institute of Biomedicine, Västra Götalandsregionen, Göteborg, Sweden
- University of Gothenburg, Department of Clinical Pathology, Sahlgrenska University Hospital, Västra Götalandsregionen, Göteborg, Sweden
| | - Bharat Prajapati
- Department of Laboratory Medicine, Institute of Biomedicine, Västra Götalandsregionen, Göteborg, Sweden
| | - Prasanna Kumar Juvvuna
- Department of Laboratory Medicine, Institute of Biomedicine, Västra Götalandsregionen, Göteborg, Sweden
| | - Sandor Dosa
- University of Gothenburg, Department of Clinical Pathology, Sahlgrenska University Hospital, Västra Götalandsregionen, Göteborg, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Children's and Women's Health, Pediatric Oncology, Astrid Lindgrens Childrens Hospital, Stockholm, Sweden
| | - John I Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health Karolinska University Hospital, Stockholm, Sweden
| | - Kanduri Chandrasekhar
- Department of Laboratory Medicine, Institute of Biomedicine, Västra Götalandsregionen, Göteborg, Sweden
| | - Levent M Akyürek
- Department of Laboratory Medicine, Institute of Biomedicine, Västra Götalandsregionen, Göteborg, Sweden
- University of Gothenburg, Department of Clinical Pathology, Sahlgrenska University Hospital, Västra Götalandsregionen, Göteborg, Sweden
| |
Collapse
|
24
|
Li Y, Che J, Chang L, Guo M, Bao X, Mu D, Sun X, Zhang X, Lu W, Xie J. CD47- and Integrin α4/β1-Comodified-Macrophage-Membrane-Coated Nanoparticles Enable Delivery of Colchicine to Atherosclerotic Plaque. Adv Healthc Mater 2022; 11:e2101788. [PMID: 34786845 DOI: 10.1002/adhm.202101788] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/26/2021] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease and the major pathological factor of most cardiovascular diseases, leading to ≈1/3 of deaths worldwide. Improving local delivery of anti-inflammatory drugs to the site of atherosclerosis has significant promise to prevent the development of atherosclerotic plaque clinically. Here, a modified-macrophage-membrane-coated nanoparticle drug delivery able to transport colchicine to the atherosclerotic site is reported. This hybrid system efficiently targets endothelial cells under an inflammatory environment while escaping the endocytosis of macrophages. Furthermore, the anti-inflammatory effect of the modified-macrophage-membrane-coated nanoparticles on foam cells is studied. In vivo, the migration of the modified-macrophage-membrane-coated nanoparticles to atherosclerotic lesions is confirmed in a vulnerable atherosclerotic plaque mouse model. Intravenous injections of the hybrid system successfully reduce the lipid plaque load and improve the plaque stability. This strategy provides a potential therapeutic system for the targeted delivery of anti-inflammatory drugs to the atherosclerotic site for the treatment of atherosclerosis in cardiovascular diseases.
Collapse
Affiliation(s)
- Yuyu Li
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Junyi Che
- Institute of Translational Medicine Department of Science and Technology Drum Tower Hospital Medical School of Nanjing University Nanjing 211800 China
| | - Lei Chang
- Department of Cardiology Affiliated Nanjing Drum Tower Hospital of Nanjing Medical University Nanjing 211800 China
| | - Meng Guo
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Xue Bao
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Dan Mu
- Department of Radiology Drum Tower Hospital Medical School of Nanjing University Nanjing 211800 China
| | - Xuan Sun
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Xin Zhang
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| | - Wen Lu
- Department of Cardiology Xu Zhou Central Hospital Xu Zhou 221009 China
| | - Jun Xie
- Department of Cardiology Drum Tower Hospital MOE Key Laboratory of Model Animal for Disease Medical School of Nanjing University Nanjing 211800 China
| |
Collapse
|
25
|
Yang C, Yang P, Liu P, Wang H, Ke E, Li K, Yan H. Targeting Filamin A alleviates ovariectomy-induced bone loss in mice via the WNT/β-catenin signaling pathway. Cell Signal 2021; 90:110191. [PMID: 34774991 DOI: 10.1016/j.cellsig.2021.110191] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/22/2021] [Accepted: 11/08/2021] [Indexed: 11/03/2022]
Abstract
Osteoporosis (OP) is a worldwide prevalent chronic metabolic bone disease, causing by a disruption of the balance between bone resorption and formation. Estrogen deficiency and aging are the main causes for disturbances in bone remodeling activity and bone loss, however, the mechanisms underlying bone remodeling regulation require clarification if novel targets for OP treatment are to be identified. In this investigation, we showed that filamin A (FLNA) accumulated in osteoblasts (OBs) and osteoclasts (OC) in bone from human OP samples, and mice with age-related and postmenopausal OP. FLNA negatively modulated in vitro osteogenic differentiation and positively promoted RANKL-induced osteoclastic differentiation. Mechanistically, FLNA interacted with low-density lipoprotein receptor-related proteins 6 (LRP6) to inhibit β-catenin expression, and enhanced nuclear factor of activated T cell c1 (NFATc1)-dependent osteoclastogenic gene expression to inhibit osteogenesis, and promote osteoclastogenesis. Inhibiting FLNA with calpeptin activated WNT/β-catenin signaling, resulting in prominent protective effects of bone loss in ovariectomy (OVX)-induced postmenopausal OP mice. Our findings revealed that FLNA not only participated in OP pathogenesis, but could be a new target to stimulate bone formation and inhibit bone resorption. Targeting FLNA with calpeptin may be a promising therapeutic approach for postmenopausal OP in the future.
Collapse
Affiliation(s)
- Changsheng Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, Guangdong Province 510000, China
| | - Panpan Yang
- Academy of Orthopedics Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Peilin Liu
- Academy of Orthopedics Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Hong Wang
- Academy of Orthopedics Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Ee Ke
- Guangdong Provincial People's Hospital, Guangdong, Academy of Medical Sciences, Guangzhou 510080, China.
| | - Kai Li
- Academy of Orthopedics Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China.
| | - Huibo Yan
- Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, Guangdong Province 510000, China.
| |
Collapse
|
26
|
Bandaru S, Ala C, Zhou AX, Akyürek LM. Filamin A Regulates Cardiovascular Remodeling. Int J Mol Sci 2021; 22:ijms22126555. [PMID: 34207234 PMCID: PMC8235345 DOI: 10.3390/ijms22126555] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 01/25/2023] Open
Abstract
Filamin A (FLNA) is a large actin-binding cytoskeletal protein that is important for cell motility by stabilizing actin networks and integrating them with cell membranes. Interestingly, a C-terminal fragment of FLNA can be cleaved off by calpain to stimulate adaptive angiogenesis by transporting multiple transcription factors into the nucleus. Recently, increasing evidence suggests that FLNA participates in the pathogenesis of cardiovascular and respiratory diseases, in which the interaction of FLNA with transcription factors and/or cell signaling molecules dictate the function of vascular cells. Localized FLNA mutations associate with cardiovascular malformations in humans. A lack of FLNA in experimental animal models disrupts cell migration during embryogenesis and causes anomalies, including heart and vessels, similar to human malformations. More recently, it was shown that FLNA mediates the progression of myocardial infarction and atherosclerosis. Thus, these latest findings identify FLNA as an important novel mediator of cardiovascular development and remodeling, and thus a potential target for therapy. In this update, we summarized the literature on filamin biology with regard to cardiovascular cell function.
Collapse
Affiliation(s)
- Sashidar Bandaru
- Division of Clinical Pathology, Sahlgrenska Academy Hospital, 413 45 Gothenburg, Sweden;
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (C.A.); (A.-X.Z.)
| | - Chandu Ala
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (C.A.); (A.-X.Z.)
| | - Alex-Xianghua Zhou
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (C.A.); (A.-X.Z.)
| | - Levent M. Akyürek
- Division of Clinical Pathology, Sahlgrenska Academy Hospital, 413 45 Gothenburg, Sweden;
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden; (C.A.); (A.-X.Z.)
- Correspondence:
| |
Collapse
|
27
|
Liu M, Xu Z, Zhang C, Yang C, Feng J, Lu Y, Zhang W, Chen W, Xu X, Sun X, Yang M, Liu W, Zhou T, Yang Y. NudC L279P Mutation Destabilizes Filamin A by Inhibiting the Hsp90 Chaperoning Pathway and Suppresses Cell Migration. Front Cell Dev Biol 2021; 9:671233. [PMID: 34262899 PMCID: PMC8273881 DOI: 10.3389/fcell.2021.671233] [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: 02/23/2021] [Accepted: 05/31/2021] [Indexed: 11/29/2022] Open
Abstract
Filamin A, the first discovered non-muscle actin filament cross-linking protein, plays a crucial role in regulating cell migration that participates in diverse cellular and developmental processes. However, the regulatory mechanism of filamin A stability remains unclear. Here, we find that nuclear distribution gene C (NudC), a cochaperone of heat shock protein 90 (Hsp90), is required to stabilize filamin A in mammalian cells. Immunoprecipitation-mass spectrometry and western blotting analyses reveal that NudC interacts with filamin A. Overexpression of human NudC-L279P (an evolutionarily conserved mutation in NudC that impairs its chaperone activity) not only decreases the protein level of filamin A but also results in actin disorganization and the suppression of cell migration. Ectopic expression of filamin A is able to reverse these defects induced by the overexpression of NudC-L279P. Furthermore, Hsp90 forms a complex with filamin A. The inhibition of Hsp90 ATPase activity by either geldanamycin or radicicol decreases the protein stability of filamin A. In addition, ectopic expression of Hsp90 efficiently restores NudC-L279P overexpression-induced protein stability and functional defects of filamin A. Taken together, these data suggest NudC L279P mutation destabilizes filamin A by inhibiting the Hsp90 chaperoning pathway and suppresses cell migration.
Collapse
Affiliation(s)
- Min Liu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhangqi Xu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Zhang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunxia Yang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxing Feng
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiqing Lu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Zhang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenwen Chen
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyang Xu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxia Sun
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingyang Yang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,The Cancer Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Yuehong Yang
- Department of Cell Biology, and Institute of Gastroenterology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Merinopoulos I, Gunawardena T, Stirrat C, Cameron D, Eccleshall SC, Dweck MR, Newby DE, Vassiliou VS. Diagnostic Applications of Ultrasmall Superparamagnetic Particles of Iron Oxide for Imaging Myocardial and Vascular Inflammation. JACC Cardiovasc Imaging 2021; 14:1249-1264. [PMID: 32861658 DOI: 10.1016/j.jcmg.2020.06.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/22/2020] [Accepted: 06/04/2020] [Indexed: 01/03/2023]
Abstract
Cardiac magnetic resonance (CMR) is at the forefront of noninvasive methods for the assessment of myocardial anatomy, function, and most importantly tissue characterization. The role of CMR is becoming even more significant with an increasing recognition that inflammation plays a major role for various myocardial diseases such as myocardial infarction, myocarditis, and takotsubo cardiomyopathy. Ultrasmall superparamagnetic particles of iron oxide (USPIO) are nanoparticles that are taken up by monocytes and macrophages accumulating at sites of inflammation. In this context, USPIO-enhanced CMR can provide valuable additional information regarding the cellular inflammatory component of myocardial and vascular diseases. Here, we will review the recent diagnostic applications of USPIO in terms of imaging myocardial and vascular inflammation, and highlight some of their future potential.
Collapse
Affiliation(s)
- Ioannis Merinopoulos
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Tharusha Gunawardena
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Colin Stirrat
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Donnie Cameron
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; C.J. Gorter Centre for High Field MRI, Department of Radiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Simon C Eccleshall
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Marc R Dweck
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - David E Newby
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Vassilios S Vassiliou
- Norwich Medical School, University of East Anglia, Norfolk and Norwich University Hospital, Norwich, United Kingdom; Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom.
| |
Collapse
|
29
|
Isolated Eosinophilic Myometritis: A Case Report of an Extremely Rare Phenomenon. Int J Gynecol Pathol 2021; 41:e3-e7. [PMID: 33935159 DOI: 10.1097/pgp.0000000000000790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Increased number of eosinophils in the uterus has been reported under physiological and pathologic conditions. However, eosinophilic infiltration limited to the myometrium is very unusual. A rare finding of isolated eosinophilic infiltration in the myometrium without involvement of endometrium or pathologies in the cervix or ovaries was observed in a 31-yr-old woman seeking medical attention for unexplained infertility, abnormal uterine bleeding, and dysmenorrhea. The patient had no allergies, parasitic disease, or other systemic disorders. This rare manifestation of eosinophilic infiltration expands the differential diagnosis of inflammatory conditions of the myometrium in patients with gynecological issues.
Collapse
|
30
|
Giannella A, Ceolotto G, Radu CM, Cattelan A, Iori E, Benetti A, Fabris F, Simioni P, Avogaro A, Vigili de Kreutzenberg S. PAR-4/Ca 2+-calpain pathway activation stimulates platelet-derived microparticles in hyperglycemic type 2 diabetes. Cardiovasc Diabetol 2021; 20:77. [PMID: 33812377 PMCID: PMC8019350 DOI: 10.1186/s12933-021-01267-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Background Patients with type 2 diabetes (T2DM) have a prothrombotic state that needs to be fully clarified; microparticles (MPs) have emerged as mediators and markers of this condition. Thus, we investigate, in vivo, in T2DM either with good (HbA1c ≤ 7.0%; GGC) or poor (HbA1c > 7.0%; PGC) glycemic control, the circulating levels of MPs, and in vitro, the molecular pathways involved in the release of MPs from platelets (PMP) and tested their pro-inflammatory effects on THP-1 transformed macrophages. Methods In 59 T2DM, and 23 control subjects with normal glucose tolerance (NGT), circulating levels of CD62E+, CD62P+, CD142+, CD45+ MPs were determined by flow cytometry, while plasma levels of ICAM-1, VCAM-1, IL-6 by ELISA. In vitro, PMP release and activation of isolated platelets from GGC and PGC were investigated, along with their effect on IL-6 secretion in THP-1 transformed macrophages. Results We found that MPs CD62P+ (PMP) and CD142+ (tissue factor-bearing MP) were significantly higher in PGC T2DM than GGC T2DM and NGT. Among MPs, PMP were also correlated with HbA1c and IL-6. In vitro, we showed that acute thrombin exposure stimulated a significantly higher PMP release in PGC T2DM than GGC T2DM through a more robust activation of PAR-4 receptor than PAR-1 receptor. Treatment with PAR-4 agonist induced an increased release of PMP in PGC with a Ca2+-calpain dependent mechanism since this effect was blunted by calpain inhibitor. Finally, the uptake of PMP derived from PAR-4 treated PGC platelets into THP-1 transformed macrophages promoted a marked increase of IL-6 release compared to PMP derived from GGC through the activation of the NF-kB pathway. Conclusions These results identify PAR-4 as a mediator of platelet activation, microparticle release, and inflammation, in poorly controlled T2DM. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-021-01267-w.
Collapse
Affiliation(s)
- Alessandra Giannella
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Giulio Ceolotto
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Claudia Maria Radu
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Arianna Cattelan
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Elisabetta Iori
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Andrea Benetti
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Fabrizio Fabris
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Paolo Simioni
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | - Angelo Avogaro
- Metabolic Disease Unit, Department of Medicine-DIMED, Via Giustiniani, 2, 35128, Padova, Italy
| | | |
Collapse
|
31
|
Zhou J, Kang X, An H, Lv Y, Liu X. The function and pathogenic mechanism of filamin A. Gene 2021; 784:145575. [PMID: 33737122 DOI: 10.1016/j.gene.2021.145575] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022]
Abstract
Filamin A(FLNa) is an actin-binding protein, which participates in the formation of the cytoskeleton, anchors a variety of proteins in the cytoskeleton and regulates cell adhesion and migration. It is involved in signal transduction, cell proliferation and differentiation, pseudopodia formation, vesicle transport, tumor resistance and genetic diseases by binding with interacting proteins. In order to fully elucidate the structure, function and pathogenesis of FLNa, we summarized all substances which directly or indirectly act on FLNa so far, upstream and downstream targets which having effect on it, signaling pathways and their functions. It also recorded the expression and effect of FLNa in different diseases, including hereditary disease and tumors.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China.
| | - Xinmei Kang
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China.
| | - Hanxiang An
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China.
| | - Yun Lv
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China.
| | - Xin Liu
- Department of Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China.
| |
Collapse
|
32
|
Lamsoul I, Dupré L, Lutz PG. Molecular Tuning of Filamin A Activities in the Context of Adhesion and Migration. Front Cell Dev Biol 2020; 8:591323. [PMID: 33330471 PMCID: PMC7714767 DOI: 10.3389/fcell.2020.591323] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023] Open
Abstract
The dynamic organization of actin cytoskeleton meshworks relies on multiple actin-binding proteins endowed with distinct actin-remodeling activities. Filamin A is a large multi-domain scaffolding protein that cross-links actin filaments with orthogonal orientation in response to various stimuli. As such it plays key roles in the modulation of cell shape, cell motility, and differentiation throughout development and adult life. The essentiality and complexity of Filamin A is highlighted by mutations that lead to a variety of severe human disorders affecting multiple organs. One of the most conserved activity of Filamin A is to bridge the actin cytoskeleton to integrins, thereby maintaining the later in an inactive state. We here review the numerous mechanisms cells have developed to adjust Filamin A content and activity and focus on the function of Filamin A as a gatekeeper to integrin activation and associated adhesion and motility.
Collapse
Affiliation(s)
- Isabelle Lamsoul
- Centre de Physiopathologie de Toulouse Purpan, INSERM, CNRS, Université de Toulouse, UPS, Toulouse, France
| | - Loïc Dupré
- Centre de Physiopathologie de Toulouse Purpan, INSERM, CNRS, Université de Toulouse, UPS, Toulouse, France.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Pierre G Lutz
- Centre de Physiopathologie de Toulouse Purpan, INSERM, CNRS, Université de Toulouse, UPS, Toulouse, France
| |
Collapse
|
33
|
Song A, Wang J, Tong Y, Fang J, Zhang Y, Zhang H, Ruan H, Wang K, Liu Y. BKCa channels regulate the immunomodulatory properties of WJ-MSCs by affecting the exosome protein profiles during the inflammatory response. Stem Cell Res Ther 2020; 11:440. [PMID: 33059770 PMCID: PMC7560248 DOI: 10.1186/s13287-020-01952-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) from the human umbilical cord have been studied extensively due to their immunomodulatory functions. Large-conductance Ca2+-activated K+ (BKCa channels) channels are involved in many inflammatory responses, but their involvement in the anti-inflammatory activity of WJ-MSCs is unknown. The underlying molecular mechanism, through which BKCa channels mediate the immunomodulation of WJ-MSC, which may include changes in exosomes proteomics, has not yet been clarified. Methods Alizarin staining, Oil Red O staining, and flow cytometry were used to identify WJ-MSCs, which were isolated from human umbilical cord Wharton’s jelly. BKCa channels were detected in WJ-MSCs using western blotting, real-time polymerase chain reaction (real-time PCR), and electrophysiology, and cytokine expression was examined using real-time PCR and enzyme-linked immunosorbent assays (ELISAs). Exosomes were characterized using transmission electron microscopy and nanoparticle tracking analysis. Proteomics analysis was performed to explore exosomal proteomic profiles. Results The cells derived from human umbilical cord Wharton’s jelly were identified as MSCs. BKCa channels were detected in the isolated WJ-MSCs, and the expression of these channels increased after lipopolysaccharide (LPS) stimulation. BKCa channels blockade in LPS-treated WJ-MSCs induced apoptosis and decreased interleukin-6 (IL-6) expression. Furthermore, THP-1 cells (human monocytic cell line) stimulated with LPS/interferon gamma (IFN-γ) produced more anti-inflammatory cytokines after treatment with exosomes derived from BKCa channel-knockdown WJ-MSCs (si-exo). We also observed altered expression of mitochondrial ATP synthase alpha subunit (ATP5A1), filamin B, and other proteins in si-exo, which might increase the anti-inflammatory activity of macrophages. Conclusions Our study described the functional expression of BKCa channels in WJ-MSCs, and BKCa channels regulated the immunomodulatory properties of WJ-MSCs by affecting the exosomal protein profiles during the inflammatory response.
Collapse
Affiliation(s)
- Ahui Song
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Jingjing Wang
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Yan Tong
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Junyan Fang
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China
| | - Yi Zhang
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Huiping Zhang
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Hongqiang Ruan
- Shanghai Applied Protein Technology Co., Ltd.,Research & Development Center, 58 Yuanmei Road, Shanghai, People's Republic of China
| | - Kai Wang
- The Clinical and Translational Research Center Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yingli Liu
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, People's Republic of China.
| |
Collapse
|
34
|
Bandaru S, Ala C, Ekstrand M, Akula MK, Pedrelli M, Liu X, Bergström G, Håversen L, Borén J, Bergo MO, Akyürek LM. Lack of RAC1 in macrophages protects against atherosclerosis. PLoS One 2020; 15:e0239284. [PMID: 32941503 PMCID: PMC7498073 DOI: 10.1371/journal.pone.0239284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
Abstract
The Rho GTPase RAC1 is an important regulator of cytoskeletal dynamics, but the role of macrophage-specific RAC1 has not been explored during atherogenesis. We analyzed RAC1 expression in human carotid atherosclerotic plaques using immunofluorescence and found higher macrophage RAC1 expression in advanced plaques compared with intermediate human atherosclerotic plaques. We then produced mice with Rac1-deficient macrophages by breeding conditional floxed Rac1 mice (Rac1fl/fl) with mice expressing Cre from the macrophage-specific lysosome M promoter (LC). Atherosclerosis was studied in vivo by infecting Rac1fl/fl and Rac1fl/fl/LC mice with AdPCSK9 (adenoviral vector overexpressing proprotein convertase subtilisin/kexin type 9). Rac1fl/fl/LC macrophages secreted lower levels of IL-6 and TNF-α and exhibited reduced foam cell formation and lipid uptake. The deficiency of Rac1 in macrophages reduced the size of aortic atherosclerotic plaques in AdPCSK9-infected Rac1fl/fl/LC mice. Compare with controls, intima/media ratios, the size of necrotic cores, and numbers of CD68-positive macrophages in atherosclerotic plaques were reduced in Rac1-deficient mice. Moreover, we found that RAC1 interacts with actin-binding filamin A. Macrophages expressed increased RAC1 levels in advanced human atherosclerosis. Genetic inactivation of RAC1 impaired macrophage function and reduced atherosclerosis in mice, suggesting that drugs targeting RAC1 may be useful in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Sashidar Bandaru
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Chandu Ala
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Matias Ekstrand
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Murali K. Akula
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Matteo Pedrelli
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xi Liu
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Västra Götalandregionen, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Liliana Håversen
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Martin O. Bergo
- Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Levent M. Akyürek
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Pathology, Västra Götalandregionen, Sahlgrenska University Hospital, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
35
|
Myriocin and d-PDMP ameliorate atherosclerosis in ApoE-/- mice via reducing lipid uptake and vascular inflammation. Clin Sci (Lond) 2020; 134:439-458. [PMID: 32091078 DOI: 10.1042/cs20191028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/17/2020] [Accepted: 02/21/2020] [Indexed: 02/05/2023]
Abstract
Sphingolipids have been implicated in the etiology of atherosclerosis. The commonly used sphingolipid inhibitors, myriocin (a ceramide inhibitor) and d-PDMP (d-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol, a glycosphingolipid inhibitor), have shown therapeutic potential but their efficacy and their underlying mechanisms remain unclear. Here, apolipoprotein E-deficient (apoE-/-) mice were fed a high-fat diet (HFD) and treated with a control, myriocin, d-PDMP, or atorvastatin for 12 weeks. We analyzed the effects of these drugs on the size and detailed composition of atherosclerotic plaques. Molecular biological approaches were used to explore how the inhibitors affect lipid metabolism and foam-cell formation. Treatment with myriocin or d-PDMP led to smaller and less vulnerable atherosclerotic lesions and was almost as effective as atorvastatin. Sphingolipid inhibitors down-regulated the expression of monocyte chemotactic protein 1 (MCP-1) and its receptor chemoattractant cytokine receptor 2 (CCR2), which play a key role in monocyte recruitment. They also decreased pro-inflammatory Ly-6chigh monocytes and influenced the uptake of modified LDL by down-regulating the expression of cluster of differentiation 36 (CD36) and lectin-like oxidized LDL (ox-LDL) receptor-1 (LOX-1). The inhibitors exhibited the advantage of maintaining normal glucose homeostasis compared with atorvastatin. These findings reveal for the first time that the modulation of sphingolipid synthesis can effectively alleviate atherosclerosis progression by preventing lipid uptake and reducing inflammatory responses in the arterial walls.
Collapse
|
36
|
Haider P, Kral-Pointner JB, Mayer J, Richter M, Kaun C, Brostjan C, Eilenberg W, Fischer MB, Speidl WS, Hengstenberg C, Huber K, Wojta J, Hohensinner P. Neutrophil Extracellular Trap Degradation by Differently Polarized Macrophage Subsets. Arterioscler Thromb Vasc Biol 2020; 40:2265-2278. [PMID: 32673525 PMCID: PMC7447175 DOI: 10.1161/atvbaha.120.314883] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Supplemental Digital Content is available in the text. Macrophages are immune cells, capable to remodel the extracellular matrix, which can harbor extracellular DNA incorporated into neutrophil extracellular traps (NETs). To study the breakdown of NETs we studied the capability of macrophage subsets to degrade these structures in vitro and in vivo in a murine thrombosis model. Furthermore, we analyzed human abdominal aortic aneurysm samples in support of our in vitro and in vivo results.
Collapse
Affiliation(s)
- Patrick Haider
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Julia B Kral-Pointner
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Austria (J.B.K.-P., J.W.)
| | - Julia Mayer
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Manuela Richter
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christoph Kaun
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christine Brostjan
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery (C.B., W.E.), Medical University of Vienna, Austria
| | - Wolf Eilenberg
- Division of Vascular Surgery and Surgical Research Laboratories, Department of Surgery (C.B., W.E.), Medical University of Vienna, Austria
| | - Michael B Fischer
- Department of Blood Group Serology and Transfusion Medicine (M.B.F.), Medical University of Vienna, Austria.,Department of Biomedical Research, Danube University Krems, Austria (M.B.F.)
| | - Walter S Speidl
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Christian Hengstenberg
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| | - Kurt Huber
- Wilhelminenhospital, 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Vienna, Austria (K.H.).,Sigmund Freud University, Medical Faculty, Vienna, Austria (K.H.)
| | - Johann Wojta
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, Austria (J.B.K.-P., J.W.).,Medical University of Vienna, Core Facilities, Austria (J.W.)
| | - Philipp Hohensinner
- From the Division of Cardiology, Department of Medicine II (P. Haider, J.B.K.-P., J.M., M.R., C.K., W.S.S., C.H., J.W., P. Hohensinner), Medical University of Vienna, Austria
| |
Collapse
|
37
|
Arkorful MA, Noren Hooten N, Zhang Y, Hewitt AN, Barrientos Sanchez L, Evans MK, Dluzen DF. MicroRNA-1253 Regulation of WASF2 (WAVE2) and its Relevance to Racial Health Disparities. Genes (Basel) 2020; 11:E572. [PMID: 32443852 PMCID: PMC7288301 DOI: 10.3390/genes11050572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/13/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
The prevalence of hypertension among African Americans (AAs) in the US is among the highest of any demographic and affects over two-thirds of AA women. Previous data from our laboratory suggest substantial differential gene expression (DGE) of mRNAs and microRNAs (miRNAs) exists within peripheral blood mononuclear cells (PBMCs) isolated from AA and white women with or without hypertension. We hypothesized that DGE by race may contribute to racial differences in hypertension. In a reanalysis of our previous dataset, we found that the Wiskott-Aldrich syndrome protein Verprolin-homologous protein 2 (WASF2 (also known as WAVE2)) is differentially expressed in AA women with hypertension, along with several other members of the actin cytoskeleton signaling pathway that plays a role in cell shape and branching of actin filaments. We performed an in silico miRNA target prediction analysis that suggested miRNA miR-1253 regulates WASF2. Transfection of miR-1253 mimics into human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs) significantly repressed WASF2 mRNA and protein levels (p < 0.05), and a luciferase reporter assay confirmed that miR-1253 regulates the WASF2 3' UTR (p < 0.01). miR-1253 overexpression in HUVECs significantly increased HUVEC lamellipodia formation (p < 0.01), suggesting the miR-1253-WASF2 interaction may play a role in cell shape and actin cytoskeleton function. Together, we have identified novel roles for miR-1253 and WASF2 in a hypertension-related disparities context. This may ultimately lead to the discovery of additional actin-related genes which are important in the vascular-related complications of hypertension and influence the disproportionate susceptibility to hypertension among AAs in general and AA women in particular.
Collapse
Affiliation(s)
- Mercy A. Arkorful
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA;
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute on Aging, Baltimore, MD 21224, USA;
| | - Amirah N. Hewitt
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Lori Barrientos Sanchez
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Baltimore, MD 21224, USA; (N.N.H.); (A.N.H.); (L.B.S.); (M.K.E.)
| | - Douglas F. Dluzen
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA;
| |
Collapse
|
38
|
Akyürek LM, Bergo MO, Bandaru S. Response by Akyürek et al to Letter Regarding Article, "Targeting Filamin A Reduces Macrophage Activity and Atherosclerosis". Circulation 2019; 140:e814-e815. [PMID: 31765260 DOI: 10.1161/circulationaha.119.043865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Levent M Akyürek
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden (L.M.A., S.B.).,Department of Clinical Pathology and Cytology, Sahlgrenska University Hospital, Västra Götalandregionen, Göteborg, Sweden (L.M.A.).,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.O.B.)
| | - Martin O Bergo
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden (L.M.A., S.B.).,Department of Clinical Pathology and Cytology, Sahlgrenska University Hospital, Västra Götalandregionen, Göteborg, Sweden (L.M.A.).,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.O.B.)
| | - Sashidar Bandaru
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Sweden (L.M.A., S.B.).,Department of Clinical Pathology and Cytology, Sahlgrenska University Hospital, Västra Götalandregionen, Göteborg, Sweden (L.M.A.).,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.O.B.)
| |
Collapse
|
39
|
Wang D, Yang X, Chen T. Letter by Wang et al Regarding Article, "Targeting Filamin A Reduces Macrophage Activity and Atherosclerosis". Circulation 2019; 140:e812-e813. [PMID: 31765256 DOI: 10.1161/circulationaha.119.043075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Daxin Wang
- Clinical Medical College, Yangzhou University, Jiangsu, China (D.W., X.Y.)
| | - Xinquan Yang
- Clinical Medical College, Yangzhou University, Jiangsu, China (D.W., X.Y.).,Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (X.Y.)
| | - Tian Chen
- Department of Cardiology, Dalian Medical University, Liaoning, China (T.C.)
| |
Collapse
|