1
|
Liu H, Wang F, Hu Z, Wei J. Potential Drug Targets for Diabetic Retinopathy Identified Through Mendelian Randomization Analysis. Transl Vis Sci Technol 2024; 13:17. [PMID: 39541108 PMCID: PMC11572760 DOI: 10.1167/tvst.13.11.17] [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: 07/23/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose This study aimed to investigate the causal effect of plasma proteins on diabetic retinopathy (DR) risk and identify potential drug targets for this disease. Methods Two-sample Mendelian randomization was performed to explore potential drug targets for DR. A total of 734 proteins were selected as instrumental variables. The Steiger filtering test and colocalization analysis were conducted to determine the causal direction and genetic pleiotropy. Plasma proteins from the decode study were used to validate the findings. Results Eleven plasma proteins were associated with DR risk. Genetically predicted high levels of CCL3L1 (odds ratio [OR] = 0.582; 95% confidence interval [CI], 0.343-0.986; P = 0.044), PAM (OR = 0.782; 95% CI, 0.652-0.937; P = 0.008), GP1BA (OR = 0.793; 95% CI, 0.632-0.994; P = 0.044), GALNT16 (OR = 0.832; 95% CI, 0.727-0.952; P = 0.008), POGLUT1 (OR = 0.836; 95% CI = 0.703-0.995; P = 0.043), and DKK3 (OR = 0.859; 95% CI, 0.777-0.950; P = 0.003) have the protective effect on DR risk. Genetically predicted high levels of GFRA2 (OR = 1.104; 95% CI, 1.028-1.187; P = 0.007), PATE4 (OR = 1.405; 95% CI, 1.060-1.860; P = 0.018), GSTA1 (OR = 1.464; 95% CI, 1.163-1.842; P = 0.001), SIRPG (OR = 1.600, 95% CI, 1.244-2.057; P = 2.51E-04), and MAPK13 (OR = 1.731; 95% CI, 1.233-2.431; P = 0.002) were associated with an increased risk of DR. However, the colocalization analysis results suggested that SIRPG and GP1BA have a shared causal variant with DR. Conclusions CCL3L1, PAM, GALNT16, POGLUT1, DKK3, GFRA2, PATE4, GSTA1, and MAPK13 were associated with DR risk and were identified as potential drug targets for DR. Translational Relevance The present study has highlighted the role of CCL3L1, PAM, GALNT16, POGLUT1, DKK3, GFRA2, PATE4, GSTA1, and MAPK13 in the development of DR.
Collapse
Affiliation(s)
- Huan Liu
- Department of Ophthalmology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, PR China
| | - Feiyan Wang
- Department of Ophthalmology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, PR China
| | - Ziqing Hu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, USA
| | - Jing Wei
- Department of Ophthalmology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, PR China
| |
Collapse
|
2
|
Hu Y, Li W, Cheng X, Yang H, She ZG, Cai J, Li H, Zhang XJ. Emerging Roles and Therapeutic Applications of Arachidonic Acid Pathways in Cardiometabolic Diseases. Circ Res 2024; 135:222-260. [PMID: 38900855 DOI: 10.1161/circresaha.124.324383] [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] [Indexed: 06/22/2024]
Abstract
Cardiometabolic disease has become a major health burden worldwide, with sharply increasing prevalence but highly limited therapeutic interventions. Emerging evidence has revealed that arachidonic acid derivatives and pathway factors link metabolic disorders to cardiovascular risks and intimately participate in the progression and severity of cardiometabolic diseases. In this review, we systemically summarized and updated the biological functions of arachidonic acid pathways in cardiometabolic diseases, mainly focusing on heart failure, hypertension, atherosclerosis, nonalcoholic fatty liver disease, obesity, and diabetes. We further discussed the cellular and molecular mechanisms of arachidonic acid pathway-mediated regulation of cardiometabolic diseases and highlighted the emerging clinical advances to improve these pathological conditions by targeting arachidonic acid metabolites and pathway factors.
Collapse
Affiliation(s)
- Yufeng Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Xu Cheng
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Hailong Yang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Zhi-Gang She
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Jingjing Cai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China (H.L.)
| | - Xiao-Jing Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- School of Basic Medical Sciences, Wuhan University, China (X.-J.Z.)
| |
Collapse
|
3
|
Niu X, Zhang Z, Zhou Q, Wuethrich A, Lobb R, Trau M. Analysis of secreted small extracellular vesicles from activated human microglial cell lines reveals distinct pro- and anti-inflammatory proteomic profiles. Proteomics 2024; 24:e2300094. [PMID: 38343172 DOI: 10.1002/pmic.202300094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/12/2023] [Accepted: 01/26/2024] [Indexed: 06/04/2024]
Abstract
Microglia are a specialized population of innate immune cells located in the central nervous system. In response to physiological and pathological changes in their microenvironment, microglia can polarize into pro-inflammatory or anti-inflammatory phenotypes. A dysregulation in the pro-/anti-inflammatory balance is associated with many pathophysiological changes in the brain and nervous system. Therefore, the balance between microglia pro-/anti-inflammatory polarization can be a potential biomarker for the various brain pathologies. A non-invasive method of detecting microglia polarization in patients would have promising clinical applications. Here, we perform proteomic analysis of small extracellular vesicles (sEVs) derived from microglia cells to identify sEVs biomarkers indicative of pro-inflammatory and anti-inflammatory phenotypic changes. sEVs were isolated from microglia cell lines under different inflammatory conditions and analyzed by proteomics by liquid chromatography with mass spectrometry. Our findings provide the potential roles of sEVs that could be related to the pathogenesis of various brain diseases.
Collapse
Affiliation(s)
- Xueming Niu
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Zhen Zhang
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Quan Zhou
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Alain Wuethrich
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Richard Lobb
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
| | - Matt Trau
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
4
|
Mocci G, Sukhavasi K, Örd T, Bankier S, Singha P, Arasu UT, Agbabiaje OO, Mäkinen P, Ma L, Hodonsky CJ, Aherrahrou R, Muhl L, Liu J, Gustafsson S, Byandelger B, Wang Y, Koplev S, Lendahl U, Owens GK, Leeper NJ, Pasterkamp G, Vanlandewijck M, Michoel T, Ruusalepp A, Hao K, Ylä-Herttuala S, Väli M, Järve H, Mokry M, Civelek M, Miller CJ, Kovacic JC, Kaikkonen MU, Betsholtz C, Björkegren JL. Single-Cell Gene-Regulatory Networks of Advanced Symptomatic Atherosclerosis. Circ Res 2024; 134:1405-1423. [PMID: 38639096 PMCID: PMC11122742 DOI: 10.1161/circresaha.123.323184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND While our understanding of the single-cell gene expression patterns underlying the transformation of vascular cell types during the progression of atherosclerosis is rapidly improving, the clinical and pathophysiological relevance of these changes remains poorly understood. METHODS Single-cell RNA sequencing data generated with SmartSeq2 (≈8000 genes/cell) in 16 588 single cells isolated during atherosclerosis progression in Ldlr-/-Apob100/100 mice with human-like plasma lipoproteins and from humans with asymptomatic and symptomatic carotid plaques was clustered into multiple subtypes. For clinical and pathophysiological context, the advanced-stage and symptomatic subtype clusters were integrated with 135 tissue-specific (atherosclerotic aortic wall, mammary artery, liver, skeletal muscle, and visceral and subcutaneous, fat) gene-regulatory networks (GRNs) inferred from 600 coronary artery disease patients in the STARNET (Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task) study. RESULTS Advanced stages of atherosclerosis progression and symptomatic carotid plaques were largely characterized by 3 smooth muscle cells (SMCs), and 3 macrophage subtype clusters with extracellular matrix organization/osteogenic (SMC), and M1-type proinflammatory/Trem2-high lipid-associated (macrophage) phenotypes. Integrative analysis of these 6 clusters with STARNET revealed significant enrichments of 3 arterial wall GRNs: GRN33 (macrophage), GRN39 (SMC), and GRN122 (macrophage) with major contributions to coronary artery disease heritability and strong associations with clinical scores of coronary atherosclerosis severity. The presence and pathophysiological relevance of GRN39 were verified in 5 independent RNAseq data sets obtained from the human coronary and aortic artery, and primary SMCs and by targeting its top-key drivers, FRZB and ALCAM in cultured human coronary artery SMCs. CONCLUSIONS By identifying and integrating the most gene-rich single-cell subclusters of atherosclerosis to date with a coronary artery disease framework of GRNs, GRN39 was identified and independently validated as being critical for the transformation of contractile SMCs into an osteogenic phenotype promoting advanced, symptomatic atherosclerosis.
Collapse
MESH Headings
- Humans
- Single-Cell Analysis
- Animals
- Gene Regulatory Networks
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Male
- Plaque, Atherosclerotic
- Disease Progression
- Female
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Knockout
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
Collapse
Affiliation(s)
- Giuseppe Mocci
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
| | - Katyayani Sukhavasi
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Estonia (K.S., A.R., H.J.)
| | - Tiit Örd
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
| | - Sean Bankier
- Computational Biology Unit, Department of Informatics, University of Bergen, Norway (S.B., T.M.)
| | - Prosanta Singha
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
| | - Uma Thanigai Arasu
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
| | - Olayinka Oluwasegun Agbabiaje
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
| | - Petri Mäkinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
| | - Lijiang Ma
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York (L. Ma, S.K., K.H., J.L.M.B.)
| | - Chani J. Hodonsky
- Robert M. Berne Cardiovascular Research Center (C.J.H., G.K.O., C.J.M.), University of Virginia, Charlottesville
- Center for Public Health Genomics (C.J.H., R.A., M.C.), University of Virginia, Charlottesville
| | - Redouane Aherrahrou
- Center for Public Health Genomics (C.J.H., R.A., M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (R.A., M.C.), University of Virginia, Charlottesville
| | - Lars Muhl
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
| | - Jianping Liu
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
| | - Sonja Gustafsson
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
| | - Byambajav Byandelger
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
| | - Ying Wang
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, CA (Y.W., N.J.L.)
- Stanford Cardiovascular Institute, Stanford University, CA (Y.W., N.J.L.)
| | - Simon Koplev
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York (L. Ma, S.K., K.H., J.L.M.B.)
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, United Kingdom (S.K.)
| | - Urban Lendahl
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center (C.J.H., G.K.O., C.J.M.), University of Virginia, Charlottesville
| | - Nicholas J. Leeper
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, CA (Y.W., N.J.L.)
- Stanford Cardiovascular Institute, Stanford University, CA (Y.W., N.J.L.)
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology (G.P., M.M.), University Medical Center Utrecht, the Netherlands
- Central Diagnostics Laboratory (G.P., M.M.), University Medical Center Utrecht, the Netherlands
| | - Michael Vanlandewijck
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
| | - Tom Michoel
- Computational Biology Unit, Department of Informatics, University of Bergen, Norway (S.B., T.M.)
| | - Arno Ruusalepp
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Estonia (K.S., A.R., H.J.)
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York (L. Ma, S.K., K.H., J.L.M.B.)
| | - Seppo Ylä-Herttuala
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
| | - Marika Väli
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.V., C.B.)
- Department of Pathological anatomy and Forensic medicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Estonia (M.V.)
| | - Heli Järve
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital and Department of Cardiology, Institute of Clinical Medicine, Tartu University, Estonia (K.S., A.R., H.J.)
| | - Michal Mokry
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
- Laboratory of Experimental Cardiology (G.P., M.M.), University Medical Center Utrecht, the Netherlands
| | - Mete Civelek
- Center for Public Health Genomics (C.J.H., R.A., M.C.), University of Virginia, Charlottesville
- Department of Biomedical Engineering (R.A., M.C.), University of Virginia, Charlottesville
| | - Clint J. Miller
- Robert M. Berne Cardiovascular Research Center (C.J.H., G.K.O., C.J.M.), University of Virginia, Charlottesville
| | - Jason C. Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (J.C.K.)
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia (J.C.K.)
- St. Vincent’s Clinical School, University of NSW, Sydney, Australia (J.C.K.)
| | - Minna U. Kaikkonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio (T.O., P.S., U.T.A., O.O.A., P.M., S.Y.-H., M.U.K.)
| | - Christer Betsholtz
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.V., C.B.)
| | - Johan L.M. Björkegren
- Department of Medicine (Huddinge), Karolinska Institutet, Sweden (G.M., L. Muhl, J.L., S.G., B.B., U.L., M.V., C.B., J.L.M.B.)
- Department of Genetics and Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York (L. Ma, S.K., K.H., J.L.M.B.)
- Clinical Gene Networks AB, Stockholm, Sweden (J.L.M.B.)
| |
Collapse
|
5
|
Xu P, Cao Y, Zhang S, Liu X, Zhang M, Zhang C. The predictive value of serum Dickkopf-1, Dickkopf-3 level to coronary artery disease and acute coronary syndrome. Int J Cardiol 2024; 403:131887. [PMID: 38382851 DOI: 10.1016/j.ijcard.2024.131887] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 01/22/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Previous studies have already confirmed the association between Dickkopf (Dkk) protein and the occurrence and progression of atherosclerosis. However, there is limited clinical evidence regarding the serum levels of Dickkopf-1 (Dkk1) and Dickkopf-3 (Dkk3) in relation to atherosclerotic cardiovascular disease (ASCVD), particularly acute coronary syndrome (ACS). MATERIALS AND METHODS A total of 88 healthy volunteers and 280 patients with coronary artery disease (CAD) undergoing coronary angiography for angina between October 2021 and October 2022, including 96 cases of stable angina (SA), 96 of unstable angina (UA) and 88 of acute myocardial infarction (AMI) were included finally. The serum concentrations of Dkk1 and Dkk3 were measured using electrochemiluminescence of Meso Scale Discovery. The predictive value of single or combined application of serum Dkk1 and Dkk3 in CAD and ACS were evaluated. RESULTS The serum levels of Dkk1 were significantly higher in the SA group, UA group, and AMI group compared to the control group. Multivariable logistic regression analysis demonstrated that elevated serum Dkk1 levels were independent predictive factors for increased risk of CAD and ACS (OR = 1.027, 95%CI = 1.019-1.034, p < 0.001; OR = 1.045, 95%CI = 1.028-1.053, p < 0.001, respectively). Receiver operating characteristic curve (ROC) analysis showed that the optimal cutoff value of serum Dkk1 for predicting ACS was 205 ng/dl, with a sensitivity of 82.6% and specificity of 96.6%. The area under the curve (AUC) was 0.930 (95%CI: 0.899-0.961, p < 0.001). Regarding Dkk3, serum Dkk3 levels were elevated in CAD patients compared to the healthy control group, and significantly higher in ACS patients compared to SA patients. Serum Dkk3 was significantly associated with increased risk of CAD and ACS (OR = 1.131, 95%CI = 1.091-1.173, p < 0.001; OR = 1.201, 95%CI = 1.134-1.271, p < 0.001, respectively). ROC curve analysis showed that the optimal cutoff value of serum Dkk3 for predicting ACS was 50.82 ng/ml, with a sensitivity of 85.9% and specificity of 87.5%. The AUC was 0.925 (95%CI: 0.894-0.956, p < 0.001). When serum Dkk1 and Dkk3 are combined as predictive factors for ACS, the AUC was 0.975. CONCLUSION Serum levels of Dkk1 and Dkk3 are significantly associated with an increased risk of CAD and ACS, and they possess predictive value for CAD and ACS. The combination of serum Dkk1 and Dkk3 is a superior predictive factor for CAD and ACS.
Collapse
Affiliation(s)
- Panpan Xu
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Cao
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shuai Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoling Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Meng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Cheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
6
|
Dziamałek-Macioszczyk P, Winiarska A, Pawłowska A, Wojtacha P, Stompór T. Patterns of Dickkopf-3 Serum and Urine Levels at Different Stages of Chronic Kidney Disease. J Clin Med 2023; 12:4705. [PMID: 37510820 PMCID: PMC10380869 DOI: 10.3390/jcm12144705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/08/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Dickkopf 3 (Dkk3) is a WNT/β-catenin signaling pathway regulator secreted by tubular epithelial cells upon the influence of different stressors. Recently Dkk3 was described as a biomarker of tubular cell injury and a tool that may estimate the risk of chronic kidney disease (CKD) progression. The data about Dkk3 concentrations at particular stages of CKD are lacking. The aim of this study was to measure serum and urine Dkk3 levels in patients with different 'renal status' and evaluate its role as a biomarker of renal damage. One hundred individuals, aged between 24 and 85 years (mean 53.1 ± 17.1), were enrolled in the study. Five groups of 20 subjects each were recruited based on their kidney function. Serum and urine Dkk3 levels were measured by ELISA. The highest median urinary Dkk3 normalized to urinary creatinine was found in patients with established CKD (7051 pg/mg). It was two times higher in renal transplant patients (5705 pg/mg) than in healthy individuals (2654 pg/mg) and the glomerulonephritis group (2470 pg/mg). Urinary Dkk3 was associated with serum creatinine in participants with established CKD and following transplantation. Our results confirm the potential role of Dkk3 as a biomarker of an ongoing renal injury.
Collapse
Affiliation(s)
- Paulina Dziamałek-Macioszczyk
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Agata Winiarska
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Anna Pawłowska
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| | - Paweł Wojtacha
- Department of Industrial and Food Microbiology, University of Warmia and Mazury in Olsztyn, 10-726 Olsztyn, Poland
| | - Tomasz Stompór
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-561 Olsztyn, Poland
| |
Collapse
|
7
|
Manta CP, Leibing T, Friedrich M, Nolte H, Adrian M, Schledzewski K, Krzistetzko J, Kirkamm C, David Schmid C, Xi Y, Stojanovic A, Tonack S, de la Torre C, Hammad S, Offermanns S, Krüger M, Cerwenka A, Platten M, Goerdt S, Géraud C. Targeting of Scavenger Receptors Stabilin-1 and Stabilin-2 Ameliorates Atherosclerosis by a Plasma Proteome Switch Mediating Monocyte/Macrophage Suppression. Circulation 2022; 146:1783-1799. [PMID: 36325910 DOI: 10.1161/circulationaha.121.058615] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Scavenger receptors Stabilin-1 (Stab1) and Stabilin-2 (Stab2) are preferentially expressed by liver sinusoidal endothelial cells. They mediate the clearance of circulating plasma molecules controlling distant organ homeostasis. Studies suggest that Stab1 and Stab2 may affect atherosclerosis. Although subsets of tissue macrophages also express Stab1, hematopoietic Stab1 deficiency does not modulate atherogenesis. Here, we comprehensively studied how targeting Stab1 and Stab2 affects atherosclerosis. METHODS ApoE-KO mice were interbred with Stab1-KO and Stab2-KO mice and fed a Western diet. For antibody targeting, Ldlr-KO mice were also used. Unbiased plasma proteomics were performed and independently confirmed. Ligand binding studies comprised glutathione-S-transferase-pulldown and endocytosis assays. Plasma proteome effects on monocytes were studied by single-cell RNA sequencing in vivo, and by gene expression analyses of Stabilin ligand-stimulated and plasma-stimulated bone marrow-derived monocytes/macrophages in vitro. RESULTS Spontaneous and Western diet-associated atherogenesis was significantly reduced in ApoE-Stab1-KO and ApoE-Stab2-KO mice. Similarly, inhibition of Stab1 or Stab2 by monoclonal antibodies significantly reduced Western diet-associated atherosclerosis in ApoE-KO and Ldlr-KO mice. Although neither plasma lipid levels nor circulating immune cell numbers were decisively altered, plasma proteomics revealed a switch in the plasma proteome, consisting of 231 dysregulated proteins comparing wildtype with Stab1/2-single and Stab1/2-double KO, and of 41 proteins comparing ApoE-, ApoE-Stab1-, and ApoE-Stab2-KO. Among this broad spectrum of common, but also disparate scavenger receptor ligand candidates, periostin, reelin, and TGFBi (transforming growth factor, β-induced), known to modulate atherosclerosis, were independently confirmed as novel circulating ligands of Stab1/2. Single-cell RNA sequencing of circulating myeloid cells of ApoE-, ApoE-Stab1-, and ApoE-Stab2-KO mice showed transcriptomic alterations in patrolling (Ccr2-/Cx3cr1++/Ly6Clo) and inflammatory (Ccr2+/Cx3cr1+/Ly6Chi) monocytes, including downregulation of proatherogenic transcription factor Egr1. In wildtype bone marrow-derived monocytes/macrophages, ligand exposure alone did not alter Egr1 expression in vitro. However, exposure to plasma from ApoE-Stab1-KO and ApoE-Stab2-KO mice showed a reverted proatherogenic macrophage activation compared with ApoE-KO plasma, including downregulation of Egr1 in vitro. CONCLUSIONS Inhibition of Stab1/Stab2 mediates an anti-inflammatory switch in the plasma proteome, including direct Stabilin ligands. The altered plasma proteome suppresses both patrolling and inflammatory monocytes and, thus, systemically protects against atherogenesis. Altogether, anti-Stab1- and anti-Stab2-targeted therapies provide a novel approach for the future treatment of atherosclerosis.
Collapse
Affiliation(s)
- Calin-Petru Manta
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Thomas Leibing
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Section of Clinical and Molecular Dermatology (T.L., M.A., J.K., C.K., Y.X., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Mirco Friedrich
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Department of Neurology, MCTN (M.F., M.P.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany (M.F., M.P.)
| | - Hendrik Nolte
- European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Institute for Genetics and CECAD, University of Cologne, Germany (H.N., M.K.).,Max Planck Institute for Biology of Ageing, Cologne, Germany (H.N.)
| | - Monica Adrian
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Section of Clinical and Molecular Dermatology (T.L., M.A., J.K., C.K., Y.X., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Kai Schledzewski
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Jessica Krzistetzko
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Section of Clinical and Molecular Dermatology (T.L., M.A., J.K., C.K., Y.X., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Christof Kirkamm
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Section of Clinical and Molecular Dermatology (T.L., M.A., J.K., C.K., Y.X., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Christian David Schmid
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Yannick Xi
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Section of Clinical and Molecular Dermatology (T.L., M.A., J.K., C.K., Y.X., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Ana Stojanovic
- European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience, MI3 (A.S., A.C.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Sarah Tonack
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany (S.T., S.O., M.K.)
| | - Carolina de la Torre
- Centre for Medical Research (ZMF) (C.d.l.T.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Seddik Hammad
- Department of Medicine II (S.H.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany (S.T., S.O., M.K.)
| | - Marcus Krüger
- Institute for Genetics and CECAD, University of Cologne, Germany (H.N., M.K.).,Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany (S.T., S.O., M.K.)
| | - Adelheid Cerwenka
- European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience, MI3 (A.S., A.C.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Michael Platten
- European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Department of Neurology, MCTN (M.F., M.P.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany (M.F., M.P.)
| | - Sergij Goerdt
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Cyrill Géraud
- Department of Dermatology, Venereology, and Allergology (C.-P.M., T.L., M.A., K.S., J.K., C.K., C.D.S., Y.X., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,Section of Clinical and Molecular Dermatology (T.L., M.A., J.K., C.K., Y.X., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany.,European Center for Angioscience (ECAS) (C.-P.M., T.L., M.F., M.A., K.S., J.K., C.K., C.D.S., Y.X., A.S., A.C., M.P., S.G., C.G.), University Medical Center and Medical Faculty Mannheim, Heidelberg University, Germany
| |
Collapse
|
8
|
Bachmann JC, Baumgart SJ, Uryga AK, Bosteen MH, Borghetti G, Nyberg M, Herum KM. Fibrotic Signaling in Cardiac Fibroblasts and Vascular Smooth Muscle Cells: The Dual Roles of Fibrosis in HFpEF and CAD. Cells 2022; 11:1657. [PMID: 35626694 PMCID: PMC9139546 DOI: 10.3390/cells11101657] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/11/2022] Open
Abstract
Patients with heart failure with preserved ejection fraction (HFpEF) and atherosclerosis-driven coronary artery disease (CAD) will have ongoing fibrotic remodeling both in the myocardium and in atherosclerotic plaques. However, the functional consequences of fibrosis differ for each location. Thus, cardiac fibrosis leads to myocardial stiffening, thereby compromising cardiac function, while fibrotic remodeling stabilizes the atherosclerotic plaque, thereby reducing the risk of plaque rupture. Although there are currently no drugs targeting cardiac fibrosis, it is a field under intense investigation, and future drugs must take these considerations into account. To explore similarities and differences of fibrotic remodeling at these two locations of the heart, we review the signaling pathways that are activated in the main extracellular matrix (ECM)-producing cells, namely human cardiac fibroblasts (CFs) and vascular smooth muscle cells (VSMCs). Although these signaling pathways are highly overlapping and context-dependent, effects on ECM remodeling mainly act through two core signaling cascades: TGF-β and Angiotensin II. We complete this by summarizing the knowledge gained from clinical trials targeting these two central fibrotic pathways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kate M. Herum
- Research and Early Development, Novo Nordisk A/S, Novo Nordisk Park, 2760 Maaloev, Denmark; (J.C.B.); (S.J.B.); (A.K.U.); (M.H.B.); (G.B.); (M.N.)
| |
Collapse
|
9
|
Zhang Y, Zuo X. miR-25-3p protects renal tubular epithelial cells from apoptosis induced by renal IRI by targeting DKK3. Open Life Sci 2022; 16:1393-1404. [PMID: 35174294 PMCID: PMC8812715 DOI: 10.1515/biol-2021-0127] [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: 07/08/2021] [Revised: 10/10/2021] [Accepted: 10/20/2021] [Indexed: 11/15/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is one of the main causes of acute kidney injury (AKI). So far, there have been many studies on renal IRI, although an effective treatment method has not been developed. In recent years, growing evidence has shown that small noncoding RNAs play an important regulatory role in renal IRI. This article aims to explore whether microRNA-25-3p (miR-25-3p) plays a role in the molecular mechanism of renal IRI. The results showed that the expression level of miR-25-3p was significantly downregulated in a rat renal IRI model, and this result was confirmed with in vitro experiments. After the hypoxia-reoxygenation treatment, the apoptosis level of NRK-52E cells transfected with miR-25-3p mimics decreased significantly, and this antiapoptotic effect was antagonized by miR-25-3p inhibitors. In addition, we confirmed that DKK3 is a target of miR-25-3p. miR-25-3p exerts its protective effect against apoptosis on NRK-52E cells by inhibiting the expression of DKK3, and downregulating the expression level of miR-25-3p could disrupt this protective effect. In addition, we reconfirmed the role of miR-25-3p in rats. Therefore, we confirmed that miR-25-3p may target DKK3 to reduce renal cell damage caused by hypoxia and that miR-25-3p may be a new potential treatment for renal IRI.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacology, Nanjing Medical University, Nanjing 210000, Jiangsu Province, P. R. China
| | - Xiangrong Zuo
- Department of Intensive Care Medical, Jiangsu Provincial People's Hospital, Nanjing 210000, Jiangsu Province, P. R. China
| |
Collapse
|
10
|
Tan W, Wang G, Liu G, You D, Wei M, Jin X, Zhao W, Zheng M. The elevation of miR-185-5p alleviates high-fat diet-induced atherosclerosis and lipid accumulation in vivo and in vitro via SREBP2 activation. Aging (Albany NY) 2022; 14:1729-1742. [PMID: 35172278 PMCID: PMC8908921 DOI: 10.18632/aging.203896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE SREBP2, a member of the SREBP family, is a primary regulator of lipid metabolism. In recent years, an increasing number of studies have suggested that miRNAs regulate lipid metabolism-related genes. It was speculated in this study that miRNAs may be implicated in the regulation of lipid accumulation in macrophages by SREBP2 protein. METHODS AND RESULTS GSE34812, GSE132651 and GSE28829 datasets comprised of atherosclerosis samples were downloaded to explore the gene expression profiles related to the miRNAs and SREBP2, and miR-185-5p was predicted to be a target of SREBP2. The GO annotations and KEGG pathway analysis were adopted for functional classification of differentially expressed genes, and lipid metabolic process was an enriched pathway in atherosclerosis. Besides, the effects of SREBP2 on increasing lipid accumulation were investigated in vivo using miR-185-5p mimic/apoE-/- mice and miR-185-5p NC/apoE-/- mice. All mice fed with a HFD suffered from atherosclerosis. Moreover, the effects of miR-185-5p on atherosclerotic plaque formation in mice were analyzed. An in vitro assay was also performed to determine the effect of miR-185-5p on ox-LDL-stimulated RAW 264.7 macrophages. Finally, miR-185-5p mimic was transfected into cultured macrophages. The results showed that the miR-185-5p elevation might regulate lipid accumulation in mice by targeting SREBP2. Furthermore, miR-185-5p mimic repressed the activation of SREBP1, SREBP2, LDLR, SCD-1, HMGCR as well as NLRP3, IL-1β, TNF-α in HFD fed mice or ox-LDL-stimulated macrophages. CONCLUSIONS our study demonstrated that miR-185-5p effectively alleviates atherosclerosis and lipid accumulation by regulating the miR-185-5p/SREBP2 axis.
Collapse
Affiliation(s)
- Wenyun Tan
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Gang Wang
- Department of Cardiology, 980 Hospital of PLA Joint Logistics Support Forces, Shijiazhuang, Hebei, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Daofeng You
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Mei Wei
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaojing Jin
- Department of Emergency, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wei Zhao
- Department of Ultrasound, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
11
|
van Ingen E, Foks AC, Woudenberg T, van der Bent ML, de Jong A, Hohensinner PJ, Wojta J, Bot I, Quax PHA, Nossent AY. Inhibition of microRNA-494-3p activates Wnt signaling and reduces proinflammatory macrophage polarization in atherosclerosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:1228-1239. [PMID: 34853722 PMCID: PMC8607137 DOI: 10.1016/j.omtn.2021.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/24/2021] [Accepted: 10/28/2021] [Indexed: 12/31/2022]
Abstract
We have previously shown that treatment with third-generation antisense oligonucleotides against miR-494-3p (3GA-494) reduces atherosclerotic plaque progression and stabilizes lesions, both in early and established plaques, with reduced macrophage content in established plaques. Within the plaque, different subtypes of macrophages are present. Here, we aimed to investigate whether miR-494-3p directly influences macrophage polarization and activation. Human macrophages were polarized into either proinflammatory M1 or anti-inflammatory M2 macrophages and simultaneously treated with 3GA-494 or a control antisense (3GA-ctrl). We show that 3GA-494 treatment inhibited miR-494-3p in M1 macrophages and dampened M1 polarization, while in M2 macrophages miR-494-3p expression was induced and M2 polarization enhanced. The proinflammatory marker CCR2 was reduced in 3GA-494-treated atherosclerosis-prone mice. Pathway enrichment analysis predicted an overlap between miR-494-3p target genes in macrophage polarization and Wnt signaling. We demonstrate that miR-494-3p regulates expression levels of multiple Wnt signaling components, such as LRP6 and TBL1X. Wnt signaling appears activated upon treatment with 3GA-494, both in cultured M1 macrophages and in plaques of hypercholesterolemic mice. Taken together, 3GA-494 treatment dampened M1 polarization, at least in part via activated Wnt signaling, while M2 polarization was enhanced, which is both favorable in reducing atherosclerotic plaque formation and increasing plaque stability.
Collapse
Affiliation(s)
- Eva van Ingen
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Division of BioTherapeutics, LACDR, Leiden University, 2333 CC Leiden, The Netherlands
| | - Amanda C Foks
- Division of BioTherapeutics, LACDR, Leiden University, 2333 CC Leiden, The Netherlands
| | - Tamar Woudenberg
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - M Leontien van der Bent
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Alwin de Jong
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Philipp J Hohensinner
- Department of Internal Medicine II, Medical, University of Vienna, 1090 Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical, University of Vienna, 1090 Vienna, Austria.,Ludwig Boltzmann Institute for Cardiovascular Research, 1090 Vienna, Austria
| | - Ilze Bot
- Division of BioTherapeutics, LACDR, Leiden University, 2333 CC Leiden, The Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Anne Yaël Nossent
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.,Department of Internal Medicine II, Medical, University of Vienna, 1090 Vienna, Austria.,Department of Laboratory Medicine, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
12
|
Ji Y, Yan T, Zhu S, Wu R, Zhu M, Zhang Y, Guo C, Yao K. The Integrative Analysis of Competitive Endogenous RNA Regulatory Networks in Coronary Artery Disease. Front Cardiovasc Med 2021; 8:647953. [PMID: 34631806 PMCID: PMC8492936 DOI: 10.3389/fcvm.2021.647953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 08/25/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Coronary artery disease (CAD) is the leading cause of cardiovascular death. The competitive endogenous RNAs (ceRNAs) hypothesis is a new theory that explains the relationship between lncRNAs and miRNAs. The mechanism of ceRNAs in the pathological process of CAD has not been fully elucidated. The objective of this study was to explore the ceRNA mechanism in CAD using the integrative bioinformatics analysis and provide new research ideas for the occurrence and development of CAD. Methods: The GSE113079 dataset was downloaded, and differentially expressed lncRNAs (DElncRNAs) and genes (DEGs) were identified using the limma package in the R language. Weighted gene correlation network analysis (WGCNA) was performed on DElncRNAs and DEGs to explore lncRNAs and genes associated with CAD. Functional enrichment analysis was performed on hub genes in the significant module identified via WGCNA. Four online databases, including TargetScan, miRDB, miRTarBase, and Starbase, combined with an online tool, miRWalk, were used to construct ceRNA regulatory networks. Results: DEGs were clustered into ten co-expression modules with different colors using WGCNA. The brown module was identified as the key module with the highest correlation coefficient. 188 hub genes were identified in the brown module for functional enrichment analysis. DElncRNAs were clustered into sixteen modules, including seven modules related to CAD with the correlation coefficient more than 0.5. Three ceRNA networks were identified, including OIP5-AS1-miR-204-5p/miR-211-5p-SMOC1, OIP5-AS1-miR-92b-3p-DKK3, and OIP5-AS1-miR-25-3p-TMEM184B. Conclusion: Three ceRNA regulatory networks identified in this study may play crucial roles in the occurrence and development of CAD, which provide novel insights into the ceRNA mechanism in CAD.
Collapse
Affiliation(s)
- Yuyao Ji
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Tao Yan
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijie Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Runda Wu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Miao Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yangyang Zhang
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Changfa Guo
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kang Yao
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Jiang L, Sun X, Deng J, Hu Y, Xu Q. Different Roles of Stem/Progenitor Cells in Vascular Remodeling. Antioxid Redox Signal 2021; 35:192-203. [PMID: 33107320 DOI: 10.1089/ars.2020.8199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Significance: Since the discovery of vascular stem cells, there has been considerable advancement in comprehending the nature and functions of these cells. Due to their differentiation potential to repair endothelial cells and to participate in lesion formation during vascular remodeling, it is crucial to elucidate vascular stem cell behaviors and the mechanisms underlying this process, which could provide new chances for the design of clinical therapeutic application of stem cells. Recent Advances: Over the past decades, major progress has been made on progenitor/vascular stem cells in the field of cardiovascular research. Vascular stem cells are mostly latent in their niches and can be bioactivated in response to damage and get involved in endothelial repair and smooth muscle cell aggregation to generate neointima. Accumulating evidence has been shown recently, using genetic lineage tracing mouse models, to particularly provide solutions to the nature of vascular stem cells and to monitor both cell migration and the process of differentiation during physiological angiogenesis and in vascular diseases. Critical Issues: This article reviews and summarizes the current research progress of vascular stem cells in this field and highlights future prospects for stem cell research in regenerative medicine. Future Directions: Despite recent advances and achievements of stem cells in cardiovascular research, the nature and cell fate of vascular stem cells remain elusive. Further comprehensive studies using new techniques including genetic cell lineage tracing and single-cell RNA sequencing are essential to fully illuminate the role of stem cells in vascular development and diseases. Antioxid. Redox Signal. 35, 192-203.
Collapse
Affiliation(s)
- Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaolei Sun
- Vascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiacheng Deng
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Zhang Y, Wu H, He R, Ye C, Chen H, Wang J, Li Z. Dickkopf-2 knockdown protects against classic macrophage polarization and lipid loading by activation of Wnt/β-catenin signaling. J Cardiol 2021; 78:328-333. [PMID: 34030936 DOI: 10.1016/j.jjcc.2021.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Wnt/β-catenin signaling pathway plays an important role in regulation of macrophage activation implicated in the development of atherosclerosis. However, as a negative regulator of Wnt/β-catenin, the potential role of Dickkopf-2 (Dkk2) on macrophage activation remains unexplored. MATERIALS AND METHODS Bone marrow-derived macrophages (BMDMs) and mouse peritoneal macrophages (MPMs) collected from ApoE knockout mice upon oxidation low lipoprotein (Ox-LDL) administration were performed to test the expression of Dkk2. The loss-of-function strategy using siRNA-Dkk2 was further utilized for the function of Dkk2. Inhibition of β-catenin with XAV939 (a β-catenin specific inhibitor) was further used for testing its effect on macrophage activation mediated by Dkk2 knockdown. RESULTS AND CONCLUSION In the current study, real time-polymerase chain reaction analysis demonstrated that an up-regulated Dkk2 expression was observed in BMDMs and MPMs of ApoE knockout mice upon Ox-LDL administration, which was confirmed by western blot. The double immunofluorescence staining further exhibited that Dkk2 showed a strong immunoreactivity in BMDMs and primarily located in cytoplasm of macrophages. Dkk2 knockdown significantly decreased the genes related to classic M1 polarized macrophage but increased alternative M2 polarized macrophage markers. Moreover, Dkk2 silencing dramatically attenuated foam cell formation which was contributed by promoted markers' expression associated with cholesterol efflux but attenuated markers to cholesterol influx. Mechanistically, we observed that Dkk2 knockdown activated Wnt/β-catenin signaling by promoting β-catenin to translocate into the nuclei of macrophages, and XAV939 reversed the ameliorated effect of Dkk2 silencing macrophage activation. Taken together, these results suggested that downregulated Dkk2 expression in macrophages was responsible for the inactivation of macrophage through targeting Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yuan Zhang
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Hongkun Wu
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Rui He
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Changlun Ye
- Department of Cardiology, Chongqing Qijiang District People's Hospital, Chongqing, PR China
| | - Hao Chen
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Jiao Wang
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China
| | - Zhenggong Li
- Center of Cardiology, Chong Qing General Hospital, University of Chinese Academy of Sciences, Chongqing, PR China.
| |
Collapse
|
15
|
The emerging plasma biomarker Dickkopf-3 (DKK3) and its association with renal and cardiovascular disease in the general population. Sci Rep 2021; 11:8642. [PMID: 33883651 PMCID: PMC8060267 DOI: 10.1038/s41598-021-88107-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/05/2021] [Indexed: 12/29/2022] Open
Abstract
Dickkopf-3 (DKK3) is an emerging biomarker for cardiovascular disease (CVD) and chronic kidney disease (CKD). Herein, baseline DKK3 plasma levels were measured in 8420 subjects from the Prevention of Renal and Vascular ENd-stage Disease (PREVEND) cohort, a large general population cohort, using enzyme-linked immunosorbent assays. Associations with clinical variables and outcomes were analysed. Median DKK3 level was 32.8 ng/ml (28.0–39.0). In multivariable linear regression analysis, the strongest correlates for plasma DKK3 were age, body mass index and estimated glomerular filtration rate (eGFR). At baseline, 564 (6.7%) subjects had CVD (defined as a myocardial infarction and/or cerebrovascular accident) and 1361 (16.2%) subjects had CKD (defined as eGFR < 60 ml/min/1.73m2 and/or urinary albumin excretion (UAE) > 30 mg/24 h). Of subjects with known CVD and CKD follow-up status (respectively 7828 and 5548), 669 (8.5%) developed CVD and 951 (17.1%) developed CKD (median follow-up respectively 12.5 and 10.2 years). Crude logistic regression analysis revealed that DKK3 levels were associated with prevalent CVD (Odds ratio: 2.14 [1.76–2.61] per DKK3 doubling, P < 0.001) and CKD (Odds ratio: 1.84 [1.59–2.13] per DKK3 doubling, P < 0.001). In crude Cox proportional hazard regression analysis, higher DKK3 levels were associated with higher risk for new-onset CVD (Hazard ratio: 1.47 [1.13–1.91] per DKK3 doubling, P = 0.004) and CKD (Hazard ratio: 1.45, [1.25–1.69] per DKK3 doubling, P < 0.001). However, these associations remained no longer significant after correction for common clinical variables and risk factors, though independently predicted for new-onset CKD in a subgroup of subjects with the lowest UAE values. Together, DKK3 plasma levels are associated with cardiovascular risk factors, but are generally not independently associated with prevalent and new-onset CVD and CKD and only predicted for new-onset CKD in those subjects with the lowest UAE values.
Collapse
|
16
|
Li SX, Li J, Dong LW, Guo ZY. Cytoskeleton-Associated Protein 4, a Promising Biomarker for Tumor Diagnosis and Therapy. Front Mol Biosci 2021; 7:552056. [PMID: 33614703 PMCID: PMC7892448 DOI: 10.3389/fmolb.2020.552056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Cytoskeleton-associated protein 4 (CKAP4) is located in the rough endoplasmic reticulum (ER) and plays an important role in stabilizing the structure of ER. Meanwhile, CKAP4 is also found to act as an activated receptor at the cell surface. The multifunction of CKAP4 was gradually discovered with growing research evidence. In addition to the involvement in various physiological events including cell proliferation, cell migration, and stabilizing the structure of ER, CKAP4 has been implicated in tumorigenesis. However, the role of CKAP4 is still controversial in tumor biology, which may be related to different signal transduction pathways mediated by binding to different ligands in various microenvironments. Interestingly, CKAP4 has been recently recognized as a serological marker of several tumors and CKAP4 is expected to be a tumor therapeutic target. Therefore, deciphering the gene status, expression regulation, functions of CKAP4 in different diseases may shed new light on CKAP4-based cancer diagnosis and therapeutic strategy. This review discusses the publications that describe CKAP4 in various diseases, especially on tumor promotion and suppression, and provides a detailed discussion on the discrepancy.
Collapse
Affiliation(s)
- Shuang-Xi Li
- Department of Nephrology, Changhai Hospital, The Navy Military Medical University, Shanghai, China
| | - Juan Li
- Department of Nephrology, Changhai Hospital, The Navy Military Medical University, Shanghai, China
| | - Li-Wei Dong
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, The Navy Military Medical University, Shanghai, China.,National Center for Liver Cancer, Shanghai, China
| | - Zhi-Yong Guo
- Department of Nephrology, Changhai Hospital, The Navy Military Medical University, Shanghai, China
| |
Collapse
|
17
|
Tuttolomondo A, Puleo MG, Velardo MC, Corpora F, Daidone M, Pinto A. Molecular Biology of Atherosclerotic Ischemic Strokes. Int J Mol Sci 2020; 21:9372. [PMID: 33317034 PMCID: PMC7763838 DOI: 10.3390/ijms21249372] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Among the causes of global death and disability, ischemic stroke (also known as cerebral ischemia) plays a pivotal role, by determining the highest number of worldwide mortality, behind cardiomyopathies, affecting 30 million people. The etiopathogenetic burden of a cerebrovascular accident could be brain ischemia (~80%) or intracranial hemorrhage (~20%). The most common site when ischemia occurs is the one is perfused by middle cerebral arteries. Worse prognosis and disablement consequent to brain damage occur in elderly patients or affected by neurological impairment, hypertension, dyslipidemia, and diabetes. Since, in the coming years, estimates predict an exponential increase of people who have diabetes, the disease mentioned above constitutes together with stroke a severe social and economic burden. In diabetic patients after an ischemic stroke, an exorbitant activation of inflammatory molecular pathways and ongoing inflammation is responsible for more severe brain injury and impairment, promoting the advancement of ischemic stroke and diabetes. Considering that the ominous prognosis of ischemic brain damage could by partially clarified by way of already known risk factors the auspice would be modifying poor outcome in the post-stroke phase detecting novel biomolecules associated with poor prognosis and targeting them for revolutionary therapeutic strategies.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties, “G. D’Alessandro”, University of Palermo, Piazza delle Cliniche n.2, 90127 Palermo, Italy; (M.G.P.); (M.C.V.); (F.C.); (M.D.); (A.P.)
| | | | | | | | | | | |
Collapse
|
18
|
Ackers I, Szymanski C, Silver MJ, Malgor R. Oxidized Low-Density Lipoprotein Induces WNT5A Signaling Activation in THP-1 Derived Macrophages and a Human Aortic Vascular Smooth Muscle Cell Line. Front Cardiovasc Med 2020; 7:567837. [PMID: 33330641 PMCID: PMC7710548 DOI: 10.3389/fcvm.2020.567837] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of atherosclerosis is complex, evolves, and involves many cell types. Macrophages and vascular smooth muscle cells (VSMCs) are critically involved in atherosclerosis development and progression. Several studies have shown that WNT5A protein is abundantly expressed in human atherosclerotic lesions; however, the mechanism and role of WNT signaling pathway activation is not clearly known. Using THP-1 derived macrophages, and human aortic VSMC cells, we evaluated in vitro how oxidized low-density lipoprotein (oxLDL) and WNT5A signaling interact in these two cell lines. We used western blot, scratch assay, metabolic proliferation assay, as well as immunostaining to analyze the effect of Wnt signaling activation. The results demonstrated that oxLDL, as well as WNT5A (control), induced Disheveled-2 (DVL2) activation and Kif26b degradation, indicating activation of non-canonical Wnt signaling. We found that oxLDL and WNT5A induced FZD5-ROR2 co-localization at the cellular membrane in vitro in THP-1 derived macrophages. Box5 (FZD5 receptor antagonist) inhibited oxLDL-induced DVL2/JNK activation secondary to newly secreted WNT protein from THP-1 derived macrophages. We found that WNT3A (canonical Wnt) and WNT5A showed different roles in this VSMC cell line. These findings indicate that WNT5A is upregulated by oxLDL, promotes foam cell formation, and affects VSMC phenotype and migration in these two cell lines. Also, in these cell lines FZD5 signaling seems to be necessary for lipid accumulation and, through this mechanism, WNT5A could modulate foam cell formation. Thus, our results suggest that WNT5A may contribute to the pathogenesis of vascular disease through modulating macrophage and VSMC behavior.
Collapse
Affiliation(s)
- Ian Ackers
- Osteopathic Heritage Foundation, Translational Biomedical Sciences Program, Ohio University, Athens, OH, United States
| | - Candice Szymanski
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | | | - Ramiro Malgor
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
- The Diabetes Institute, Ohio University, Athens, OH, United States
- Molecular and Cellular Biology Graduate Program, Ohio University, Athens, OH, United States
| |
Collapse
|
19
|
Liu Y, Neogi A, Mani A. The role of Wnt signalling in development of coronary artery disease and its risk factors. Open Biol 2020; 10:200128. [PMID: 33081636 PMCID: PMC7653355 DOI: 10.1098/rsob.200128] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/23/2020] [Indexed: 02/05/2023] Open
Abstract
The Wnt signalling pathways are composed of a highly conserved cascade of events that govern cell differentiation, apoptosis and cell orientation. Three major and distinct Wnt signalling pathways have been characterized: the canonical Wnt pathway (or Wnt/β-catenin pathway), the non-canonical planar cell polarity pathway and the non-canonical Wnt/Ca2+ pathway. Altered Wnt signalling pathway has been associated with diverse diseases such as disorders of bone density, different malignancies, cardiac malformations and heart failure. Coronary artery disease is the most common type of heart disease in the United States. Atherosclerosis is a multi-step pathological process, which starts with lipid deposition and endothelial cell dysfunction, triggering inflammatory reactions, followed by recruitment and aggregation of monocytes. Subsequently, monocytes differentiate into tissue-resident macrophages and transform into foam cells by the uptake of modified low-density lipoprotein. Meanwhile, further accumulations of lipids, infiltration and proliferation of vascular smooth muscle cells, and deposition of the extracellular matrix occur under the intima. An atheromatous plaque or hyperplasia of the intima and media is eventually formed, resulting in luminal narrowing and reduced blood flow to the myocardium, leading to chest pain, angina and even myocardial infarction. The Wnt pathway participates in all different stages of this process, from endothelial dysfunction to lipid deposit, and from initial inflammation to plaque formation. Here, we focus on the role of Wnt cascade in pathophysiological mechanisms that take part in coronary artery disease from both clinical and experimental perspectives.
Collapse
Affiliation(s)
- Ya Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Arpita Neogi
- Yale Cardiovascular Genetics Program, Yale University, New Haven, CT, USA
| | - Arya Mani
- Yale Cardiovascular Genetics Program, Yale University, New Haven, CT, USA
- Yale Cardiovascular Research Center, Department of Medicine, Yale University, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
20
|
Boucher P, Matz RL, Terrand J. atherosclerosis: gone with the Wnt? Atherosclerosis 2020; 301:15-22. [PMID: 32289618 DOI: 10.1016/j.atherosclerosis.2020.03.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/19/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis, a pathology affecting large and medium-sized arteries, is the major cause of cardiovascular morbidity/mortality in industrialized countries. During atherosclerosis, cells accumulate large amounts of cholesterol through the uptake of modified low-density lipoprotein particles to form foam cells. This accumulation forms the basis for the development of the disease and for a large spectrum of other diseases in various organs. Massive research efforts have yielded valuable information about the underlying molecular mechanisms of atherosclerosis. In particular, newer discoveries on the early stage of lesion formation, cholesterol accumulation, reverse cholesterol transport, and local inflammation in the vascular wall have opened unanticipated horizons of understanding and raised novel questions and therapeutic opportunities. In this review, we focus on Wnt signaling, which has received little attention so far, yet affects lysosomal function and signalling pathways that limit cholesterol accumulation. This occurs in different tissues and cell types, including smooth muscle cells, endothelial cells and macrophages in the arterial wall, and thus profoundly impacts on atherosclerotic disease development and progression.
Collapse
Affiliation(s)
- Philippe Boucher
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France.
| | - Rachel L Matz
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| | - Jérôme Terrand
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| |
Collapse
|
21
|
Zhu Z, Guo D, Zhong C, Wang A, Xu T, Peng Y, Peng H, Li Q, Ju Z, Geng D, Chen J, Zhang Y, He J. Serum dickkopf-3 is associated with death and vascular events after ischemic stroke: an observational study from CATIS. J Neuroinflammation 2020; 17:12. [PMID: 31918729 PMCID: PMC6953290 DOI: 10.1186/s12974-019-1680-4] [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: 08/07/2019] [Accepted: 12/17/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dickkopf-3 (Dkk-3) is implicated in the progression of atherosclerosis. This study aimed to investigate the association between serum Dkk-3 and the prognosis of ischemic stroke. METHODS We measured serum Dkk-3 levels in 3344 ischemic stroke patients from CATIS (China Antihypertensive Trial in Acute Ischemic Stroke). The primary outcome was a combination of death and vascular events within 3 months after ischemic stroke. RESULTS During 3 months of follow-up, the cumulative incidence rates of primary outcome among ischemic stroke patients in five quintiles of serum Dkk-3 (from low to high) were 4.49%, 3.74%, 2.54%, 5.23%, and 6.73%, respectively (log-rank p = 0.004). Multivariable Cox proportional hazards regression analyses showed that compared with the third quintile of serum Dkk-3, the adjusted hazard ratios (95% confidence intervals) associated with the first and fifth quintile were 3.49 (1.46-8.34) and 4.23 (1.86-9.64) for primary outcome, 3.47 (1.06-11.36) and 5.30 (1.81-15.51) for death, and 2.66 (1.01-7.01) and 3.35 (1.33-8.40) for vascular events, respectively. Multivariable-adjusted Cox proportional hazards regression model with restricted cubic splines showed a U-shaped association between serum Dkk-3 and the risk of primary outcome (p for nonlinearity = 0.030). Moreover, adding serum Dkk-3 to conventional risk factors could improve the predictive power for primary outcome (net reclassification improvement 28.44%, p < 0.001; integrated discrimination improvement 0.48%, p = 0.001). CONCLUSIONS Both low and high serum Dkk-3 levels are associated with increased risks of death and vascular events within 3 months after ischemic stroke, indicating that serum Dkk-3 may have a special effect on the prognosis of ischemic stroke. We also found that serum Dkk-3 might be a prognostic biomarker for ischemic stroke. Further studies are needed to replicate our findings and to determine the optimal levels of serum Dkk-3.
Collapse
Affiliation(s)
- Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province, 215123, China.,Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Daoxia Guo
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province, 215123, China
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province, 215123, China
| | - Aili Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province, 215123, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province, 215123, China
| | - Yanbo Peng
- Department of Neurology, Affiliated Hospital of North China University of Science and Technology, Tangshan, Hebei, China
| | - Hao Peng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province, 215123, China
| | - Qunwei Li
- Department of Epidemiology, School of Public Health, Taishan Medical College, Tai'an, Shandong, China
| | - Zhong Ju
- Department of Neurology, Kerqin District First People's Hospital of Tongliao City, Tongliao City, Inner Mongolia, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Chen
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.,Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province, 215123, China.
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.,Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| |
Collapse
|
22
|
Abstract
Clinical and preclinical studies over the past 3 decades have uncovered a multitude of signaling pathways involved in the initiation and progression of atherosclerosis. From these studies, signaling by proteins of the Wnt family has recently emerged as an important player in the development of atherosclerosis. Wnt signaling is characterized by a large number of ligands, receptors, and coreceptors and can be regulated at many different levels. Among Wnt modulators, the evolutionary conserved Dkk (Dickkopf) proteins, and especially Dkk-1, the founding member of the family, are the best characterized. The role of Dkks in the pathophysiology of the arterial wall is only partially understood, but their involvement in atherosclerosis is becoming increasingly evident. This review introduces recent key findings on Dkk proteins and their functions in atherosclerosis and discusses the potential importance of modulating Dkk signaling as part of a novel, improved strategy for preventing and treating atherosclerosis-related diseases.
Visual Overview—
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Roberta Baetta
- From the Centro Cardiologico Monzino, IRCCS, Milano, Italy
| | - Cristina Banfi
- From the Centro Cardiologico Monzino, IRCCS, Milano, Italy
| |
Collapse
|
23
|
Kajiwara C, Fumoto K, Kimura H, Nojima S, Asano K, Odagiri K, Yamasaki M, Hikita H, Takehara T, Doki Y, Morii E, Kikuchi A. p63-Dependent Dickkopf3 Expression Promotes Esophageal Cancer Cell Proliferation via CKAP4. Cancer Res 2018; 78:6107-6120. [PMID: 30181180 DOI: 10.1158/0008-5472.can-18-1749] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/20/2018] [Accepted: 08/28/2018] [Indexed: 11/16/2022]
Abstract
Dickkopf3 (DKK3) is a secretory protein that belongs to the DKK family, but exhibits structural divergence from other family members, and its corresponding receptors remain to be identified. Although DKK3 has been shown to have oncogenic functions in certain cancer types, the underlying mechanism by which DKK3 promotes tumorigenesis remains to be clarified. We show here that DKK3 stimulates esophageal cancer cell proliferation via cytoskeleton-associated protein 4 (CKAP4), which acts as a receptor for DKK3. DKK3 was expressed in approximately 50% of tumor lesions of esophageal squamous cell carcinoma (ESCC) cases; simultaneous expression of DKK3 and CKAP4 was associated with poor prognosis. Anti-CKAP4 antibody inhibited both binding of DKK3 to CKAP4 and xenograft tumor formation induced by ESCC cells. p63, a p53-related transcriptional factor frequently amplified in ESCC, bound to the upstream region of the DKK3 gene. Knockdown of p63 decreased DKK3 expression in ESCC cells, and reexpression of DKK3 partially rescued cell proliferation in p63-depleted ESCC cells. Expression of ΔNp63α and DKK3 increased the size of tumor-like esophageal organoids, and anti-CKAP4 antibody inhibited growth of esophageal organoids. Taken together, these results suggest that the DKK3-CKAP4 axis might serve as a novel molecular target for ESCC.Significance: In esophageal cancer, findings identify DKK3 as a poor prognostic indicator and demonstrate CKAP4 inhibition as an effective therapeutic strategy. Cancer Res; 78(21); 6107-20. ©2018 AACR.
Collapse
Affiliation(s)
- Chihiro Kajiwara
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Katsumi Fumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hirokazu Kimura
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Keita Asano
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kazuki Odagiri
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Suita, Japan.
| |
Collapse
|
24
|
Abstract
Vascular, resident stem cells are present in all 3 layers of the vessel wall; they play a role in vascular formation under physiological conditions and in remodeling in pathological situations. Throughout development and adult early life, resident stem cells participate in vessel formation through vasculogenesis and angiogenesis. In adults, the vascular stem cells are mostly quiescent in their niches but can be activated in response to injury and participate in endothelial repair and smooth muscle cell accumulation to form neointima. However, delineation of the characteristics and of the migration and differentiation behaviors of these stem cells is an area of ongoing investigation. A set of genetic mouse models for cell lineage tracing has been developed to specifically address the nature of these cells and both migration and differentiation processes during physiological angiogenesis and in vascular diseases. This review summarizes the current knowledge on resident stem cells, which has become more defined and refined in vascular biology research, thus contributing to the development of new potential therapeutic strategies to promote endothelial regeneration and ameliorate vascular disease development.
Collapse
Affiliation(s)
- Li Zhang
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Shirin Issa Bhaloo
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| | - Ting Chen
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academic of Sciences (B.Z.)
| | - Qingbo Xu
- From the Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z., T.C., Q.X.)
- School of Cardiovascular Medicine and Sciences, King’s College London, BHF Centre, United Kingdom (S.I.B., Q.X.)
| |
Collapse
|
25
|
Ackers I, Szymanski C, Duckett KJ, Consitt LA, Silver MJ, Malgor R. Blocking Wnt5a signaling decreases CD36 expression and foam cell formation in atherosclerosis. Cardiovasc Pathol 2018; 34:1-8. [PMID: 29474941 DOI: 10.1016/j.carpath.2018.01.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/12/2018] [Accepted: 01/19/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND AIMS Wnt5a is a highly studied member of the Wnt family and recently has been implicated in the pathogenesis of atherosclerosis, but its precise role is unknown. Foam cell development is a critical process to atherosclerotic plaque formation. In the present study, we investigated the role of noncanonical Wnt5a signaling in the development of foam cells. METHODS Human carotid atherosclerotic tissue and THP-1-derived macrophages were used to investigate the contribution of Wnt5a signaling in the formation of foam cells. Immunohistochemistry was used to evaluate protein expression of scavenger receptors and noncanonical Wnt5a receptors [frizzled 5 (Fz5) and receptor tyrosine kinase-like orphan receptor 2 (Ror2)] in human atherosclerotic macrophages/foam cells. Changes in protein expression in response to Wnt5a stimulation/inhibition were determined by Western blot, and lipid accumulation was evaluated by fluorescent lipid droplet staining. RESULTS Wnt5a (P<.05), Fz5 (P<.01), and Ror2 (P<.01) were significantly expressed in advanced atherosclerotic lesions compared to less advanced lesions (N=10). Wnt5a, Fz5, and Ror2 were expressed in macrophages/foam cells within the plaque. In vitro studies revealed that Wnt5a significantly increased the expression of the lipid uptake receptor CD36 (P<.05) but not the lipid efflux receptor ATP-binding cassette transporter (P>.05). rWnt5a also significantly increased lipid accumulation in THP-1 macrophages (P<.05). Furthermore, inhibition of Wnt5a signaling with Box5 prevented lipid accumulation (P<.01) and prevented CD36 up-regulation (P<.01). CONCLUSIONS These results suggest a direct role for Wnt5a signaling in the pathogenesis of atherosclerosis, specifically the accumulation of lipid in macrophages and the formation of foam cells.
Collapse
Affiliation(s)
- Ian Ackers
- Heritage Fellow, Translational Biomedical Sciences Program, Ohio University, Athens, Ohio, USA
| | - Candice Szymanski
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - K Jordan Duckett
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Leslie A Consitt
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA; The Diabetes Institute, Ohio University, Athens, Ohio, USA
| | | | - Ramiro Malgor
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA; The Diabetes Institute, Ohio University, Athens, Ohio, USA; Molecular and Cellular Biology Graduate Program, Ohio University, Athens, Ohio, USA.
| |
Collapse
|
26
|
Abstract
Chronic diseases account for approximately 45% of all deaths in developed countries and are particularly prevalent in countries with the most sophisticated and robust public health systems. Chronic metabolic diseases, specifically lifestyle-related diseases pertaining to diet and exercise, continue to be difficult to treat clinically. The most prevalent of these chronic metabolic diseases include obesity, diabetes, non-alcoholic fatty liver disease, chronic kidney disease and cardiovascular disease and will be the focus of this review. Wnt proteins are highly conserved glycoproteins best known for their role in development and homeostasis of tissues. Given the importance of Wnt signalling in homeostasis, aberrant Wnt signalling likely regulates metabolic processes and may contribute to the development of chronic metabolic diseases. Expression of Wnt proteins and dysfunctional Wnt signalling has been reported in multiple chronic diseases. It is interesting to speculate about an interrelationship between the Wnt signalling pathways as a potential pathological mechanism in chronic metabolic diseases. The aim of this review is to summarize reported findings on the contrasting roles of Wnt signalling in lifestyle-related chronic metabolic diseases; specifically, the contribution of Wnt signalling to lipid accumulation, fibrosis and chronic low-grade inflammation.
Collapse
Affiliation(s)
- Ian Ackers
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- OHF Fellow, Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, USA
| | - Ramiro Malgor
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Ramiro Malgor, Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, 202b Academic & Research Center, Athens, OH, 45701-2979 USA.
| |
Collapse
|
27
|
Karamariti E, Zhai C, Yu B, Qiao L, Wang Z, Potter CMF, Wong MM, Simpson RML, Zhang Z, Wang X, Del Barco Barrantes I, Niehrs C, Kong D, Zhao Q, Zhang Y, Hu Y, Zhang C, Xu Q. DKK3 (Dickkopf 3) Alters Atherosclerotic Plaque Phenotype Involving Vascular Progenitor and Fibroblast Differentiation Into Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2017; 38:425-437. [PMID: 29284609 DOI: 10.1161/atvbaha.117.310079] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 12/13/2017] [Indexed: 01/31/2023]
Abstract
OBJECTIVE DKK3 (dickkopf 3), a 36-kD secreted glycoprotein, has been shown to be involved in the differentiation of partially reprogrammed cells and embryonic stem cells to smooth muscle cells (SMCs), but little is known about its involvement in vascular disease. This study aims to assess the effects of DKK3 on atherosclerotic plaque composition. APPROACH AND RESULTS In the present study, we used a murine model of atherosclerosis (ApoE-/-) in conjunction with DKK3-/- and performed tandem stenosis of the carotid artery to evaluate atherosclerotic plaque development. We found that the absence of DKK3 leads to vulnerable atherosclerotic plaques, because of a reduced number of SMCs and reduced matrix protein deposition, as well as increased hemorrhage and macrophage infiltration. Further in vitro studies revealed that DKK3 can induce differentiation of Sca1+ (stem cells antigen 1) vascular progenitors and fibroblasts into SMCs via activation of the TGF-β (transforming growth factor-β)/ATF6 (activating transcription factor 6) and Wnt signaling pathways. Finally, we assessed the therapeutic potential of DKK3 in mouse and rabbit models and found that DKK3 altered the atherosclerotic plaque content via increasing SMC numbers and reducing vascular inflammation. CONCLUSIONS Cumulatively, we provide the first evidence that DKK3 is a potent SMC differentiation factor, which might have a therapeutic effect in reducing intraplaque hemorrhage related to atherosclerotic plaque phenotype.
Collapse
Affiliation(s)
- Eirini Karamariti
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Chungang Zhai
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Baoqi Yu
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Lei Qiao
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Zhihong Wang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Claire M F Potter
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Mei Mei Wong
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Russell M L Simpson
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Zhongyi Zhang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Xiaocong Wang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Ivan Del Barco Barrantes
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Christof Niehrs
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Deling Kong
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Qiang Zhao
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Yun Zhang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Yanhua Hu
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Cheng Zhang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.).
| | - Qingbo Xu
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.).
| |
Collapse
|