1
|
Singh D, Mehghini P, Rodriguez-Palacios A, Di Martino L, Cominelli F, Basson AR. Anti-Inflammatory Effect of Dietary Pentadecanoic Fatty Acid Supplementation on Inflammatory Bowel Disease in SAMP1/YitFc Mice. Nutrients 2024; 16:3031. [PMID: 39275347 PMCID: PMC11397537 DOI: 10.3390/nu16173031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Dietary fats have been linked to the increasing incidence of chronic diseases, including inflammatory bowel diseases (IBD), namely, Crohn's disease (CD). METHODS This study investigated the impact of pentadecanoic acid (C15:0), a type of an odd-numbered chain saturated fatty acid, for its potential anti-inflammatory properties in different mouse models of experimental IBD using the SAMP1/YitFc (SAMP) mouse line (14- or 24-week-old), including chronic ileitis and DSS-induced colitis. To quantitively assess the effect of C:15, we tested two dosages of C:15 in selected experiments in comparison to control mice. Intestinal inflammation and intestinal permeability were used as primary outcomes. RESULTS In ileitis, C:15 supplementation showed an anti-inflammatory effect in SAMP mice (e.g., a reduction in ileitis severity vs. control p < 0.0043), which was reproducible when mice were tested in the DSS model of colitis (e.g., reduced permeability vs. control p < 0.0006). Of relevance, even the short-term C:15 therapy prevented colitis in mice by maintaining body weight, decreasing inflammation, preserving gut integrity, and alleviating colitis signs. CONCLUSIONS Collectively, the findings from both ileitis and colitis in SAMP mice indicate that C:15 may have therapeutic effects in the treatment of IBD (colitis in the short term). This promising effect has major translational potential for the alleviation of IBD in humans.
Collapse
Affiliation(s)
- Drishtant Singh
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
| | - Paola Mehghini
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Alexander Rodriguez-Palacios
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Mouse Models Core, Silvio O’Conte Cleveland Digestive Diseases Research Core Center, Cleveland, OH 44106, USA
- Germ-Free and Gut Microbiome Core, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Luca Di Martino
- Case Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Fabio Cominelli
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Mouse Models Core, Silvio O’Conte Cleveland Digestive Diseases Research Core Center, Cleveland, OH 44106, USA
- Germ-Free and Gut Microbiome Core, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Abigail Raffner Basson
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.); (A.R.-P.); (F.C.)
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
2
|
Mori M, Sakamoto A, Kawakami R, Guo L, Slenders L, Mosquera JV, Ghosh SKB, Wesseling M, Shiraki T, Bellissard A, Shah P, Weinkauf CC, Konishi T, Sato Y, Cornelissen A, Kawai K, Jinnouchi H, Xu W, Vozenilek AE, Williams D, Tanaka T, Sekimoto T, Kelly MC, Fernandez R, Grogan A, Coslet AJ, Fedotova A, Kurse A, Mokry M, Romero ME, Kolodgie FD, Pasterkamp G, Miller CL, Virmani R, Finn AV. CD163 + Macrophages Induce Endothelial-to-Mesenchymal Transition in Atheroma. Circ Res 2024; 135:e4-e23. [PMID: 38860377 DOI: 10.1161/circresaha.123.324082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Cell phenotype switching is increasingly being recognized in atherosclerosis. However, our understanding of the exact stimuli for such cellular transformations and their significance for human atherosclerosis is still evolving. Intraplaque hemorrhage is thought to be a major contributor to plaque progression in part by stimulating the influx of CD163+ macrophages. Here, we explored the hypothesis that CD163+ macrophages cause plaque progression through the induction of proapoptotic endothelial-to-mesenchymal transition (EndMT) within the fibrous cap. METHODS Human coronary artery sections from CVPath's autopsy registry were selected for pathological analysis. Athero-prone ApoE-/- and ApoE-/-/CD163-/- mice were used for in vivo studies. Human peripheral blood mononuclear cell-induced macrophages and human aortic endothelial cells were used for in vitro experiments. RESULTS In 107 lesions with acute coronary plaque rupture, 55% had pathological evidence of intraplaque hemorrhage in nonculprit vessels/lesions. Thinner fibrous cap, greater CD163+ macrophage accumulation, and a larger number of CD31/FSP-1 (fibroblast specific protein-1) double-positive cells and TUNEL (terminal deoxynucleotidyl transferase-dUTP nick end labeling) positive cells in the fibrous cap were observed in nonculprit intraplaque hemorrhage lesions, as well as in culprit rupture sections versus nonculprit fibroatheroma sections. Human aortic endothelial cells cultured with supernatants from hemoglobin/haptoglobin-exposed macrophages showed that increased mesenchymal marker proteins (transgelin and FSP-1) while endothelial markers (VE-cadherin and CD31) were reduced, suggesting EndMT induction. Activation of NF-κB (nuclear factor kappa β) signaling by proinflammatory cytokines released from CD163+ macrophages directly regulated the expression of Snail, a critical transcription factor during EndMT induction. Western blot analysis for cleaved caspase-3 and microarray analysis of human aortic endothelial cells indicated that apoptosis was stimulated during CD163+ macrophage-induced EndMT. Additionally, CD163 deletion in athero-prone mice suggested that CD163 is required for EndMT and plaque progression. Using single-cell RNA sequencing from human carotid endarterectomy lesions, a population of EndMT was detected, which demonstrated significant upregulation of apoptosis-related genes. CONCLUSIONS CD163+ macrophages provoke EndMT, which may promote plaque progression through fibrous cap thinning.
Collapse
MESH Headings
- Humans
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Animals
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Macrophages/metabolism
- Macrophages/pathology
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/metabolism
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Mice
- Cells, Cultured
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Male
- Mice, Knockout, ApoE
- Mice, Inbred C57BL
- Apoptosis
- Female
- Epithelial-Mesenchymal Transition
- Coronary Vessels/pathology
- Coronary Vessels/metabolism
Collapse
Affiliation(s)
- Masayuki Mori
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Atsushi Sakamoto
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
- Hamamatsu University School of Medicine, Shizuoka, Japan (A.S.)
| | - Rika Kawakami
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Liang Guo
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Lotte Slenders
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Jose Verdezoto Mosquera
- Department of Public Health Sciences, Department of Biochemistry and Molecular Genetics, Center for Public Health Genomics, University of Virginia, Charlottesville (J.V.M., C.L.M.)
| | - Saikat Kumar B Ghosh
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Marian Wesseling
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Tatsuya Shiraki
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Arielle Bellissard
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Palak Shah
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | | | - Takao Konishi
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Yu Sato
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Anne Cornelissen
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Kenji Kawai
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Hiroyuki Jinnouchi
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Weili Xu
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Aimee E Vozenilek
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Desiree Williams
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Takamasa Tanaka
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Teruo Sekimoto
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Michael C Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD (M.C.K.)
| | - Raquel Fernandez
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Alyssa Grogan
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - A J Coslet
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Alisa Fedotova
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Anjali Kurse
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Michal Mokry
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Maria E Romero
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Frank D Kolodgie
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Gerard Pasterkamp
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
- University Medical Center Utrecht, the Netherlands (L.S., M.W., M. Mokry, G.P.)
| | - Clint L Miller
- Department of Public Health Sciences, Department of Biochemistry and Molecular Genetics, Center for Public Health Genomics, University of Virginia, Charlottesville (J.V.M., C.L.M.)
| | - Renu Virmani
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
| | - Aloke V Finn
- Department of Pathology, CVPath Institute, Inc, Gaithersburg, MD (M. Mori, A.S., R.K., L.G., S.K.B.G., T. Shiraki, A.B., P.S., T.K., Y.S., A.C., K.K., H.J., W.X., A.E.V., D.W., T.T., T. Sekimoto, R.F., A.G., A.J.C., A.F., A.K., M.E.R., F.D.K., R.V., A.V.F.)
- University of Maryland School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
3
|
Li B, Shaikh F, Zamzam A, Abdin R, Qadura M. Inflammatory Biomarkers to Predict Major Adverse Cardiovascular Events in Patients with Carotid Artery Stenosis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:997. [PMID: 38929614 PMCID: PMC11205582 DOI: 10.3390/medicina60060997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: Inflammatory proteins and their prognostic value in patients with carotid artery stenosis (CAS) have not been adequately studied. Herein, we identified CAS-specific biomarkers from a large pool of inflammatory proteins and assessed the ability of these biomarkers to predict adverse events in individuals with CAS. Materials and Methods: Samples of blood were prospectively obtained from 336 individuals (290 with CAS and 46 without CAS). Plasma concentrations of 29 inflammatory proteins were determined at recruitment, and the patients were followed for 24 months. The outcome of interest was a major adverse cardiovascular event (MACE; composite of stroke, myocardial infarction, or death). The differences in plasma protein concentrations between patients with vs. without a 2-year MACE were determined using the independent t-test or Mann-Whitney U test to identify CAS-specific prognostic biomarkers. Kaplan-Meier and Cox proportional hazards analyses with adjustment for baseline demographic and clinical characteristics were performed to assess the prognostic value of differentially expressed inflammatory proteins in predicting a 2-year MACE in patients with CAS. Results: The mean age of the cohort was 68.8 (SD 10.2) years and 39% were female. The plasma concentrations of two inflammatory proteins were significantly higher in individuals with a 2-year MACE relative to those without a 2-year MACE: IL-6 (5.07 (SD 4.66) vs. 3.36 (SD 4.04) pg/mL, p = 0.03) and CD163 (233.825 (SD 230.306) vs. 159.673 (SD 175.669) pg/mL, p = 0.033). Over a follow-up period of 2 years, individuals with elevated levels of IL-6 were more likely to develop MACE (HR 1.269 (95% CI 1.122-1.639), p = 0.042). Similarly, over a 2-year period, patients with high levels of CD163 were more likely to develop MACE (HR 1.413 (95% CI 1.022-1.954), p = 0.036). Conclusions: The plasma levels of inflammatory proteins IL-6 and CD163 are independently associated with adverse outcomes in individuals with CAS. These CAS-specific prognostic biomarkers may assist in the risk stratification of patients at an elevated risk of a MACE and subsequently guide further vascular evaluation, specialist referrals, and aggressive medical/surgical management, thereby improving outcomes for patients with CAS.
Collapse
Affiliation(s)
- Ben Li
- Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A1, Canada
- Temerty Centre for Artificial Intelligence Research and Education in Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Farah Shaikh
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
| | - Abdelrahman Zamzam
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
| | - Rawand Abdin
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada;
| | - Mohammad Qadura
- Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada;
- Division of Vascular Surgery, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada; (F.S.); (A.Z.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A1, Canada
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
4
|
Ghinea N, Liehn EA, Grommes J, Delattre DD, Olesen TK. Follicle-stimulating hormone receptor expression in advanced atherosclerotic plaques. Sci Rep 2024; 14:10176. [PMID: 38702476 PMCID: PMC11068877 DOI: 10.1038/s41598-024-60962-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
Experimental evidence indicates that follicle-stimulating hormone (FSH), an essential hormone for reproduction, can act directly on endothelial cells inducing atherosclerosis activation and development. However, it remains unknown whether the FSH-receptor (FSHR) is expressed in human atherosclerosis plaques. To demonstrate the FSHR presence, we used immunohistochemical and immunoelectron microscopy involving a specific monoclonal antibody FSHR1A02 that recognizes an epitope present in the FSHR-ectodomain. In all 55 patients with atherosclerotic plaques located in carotid, coronary, femoral arteries, and iliac aneurysm, FSHR was selectively expressed in arterial endothelium covering atherosclerotic plaques and endothelia lining intraplaque neovessels. Lymphatic neovessels were negative for FSHR. M1-macrophages, foam cells, and giant multinucleated cells were also FSHR-positive. FSHR was not detected in normal internal thoracic artery. Immunoelectron microscopy performed in ApoEKO/hFSHRKI mice with atherosclerotic plaques, after injection in vivo with mouse anti-hFSHR monoclonal antibody FSHR1A02 coupled to colloidal gold, showed FSHR presence on the luminal surface of arterial endothelial cells covering atherosclerotic plaques. Therefore, FSHR can bind, internalize, and deliver into the plaque circulating ligands to FSHR-positive cells. In conclusion, we report FSHR expression in endothelial cells, M1-macrophages, M1-derived foam cells, giant multinucleated macrophages, and osteoclasts associated with human atherosclerotic plaques.
Collapse
Affiliation(s)
- Nicolae Ghinea
- Département Recherche Translationnelle, Centre de Recherche, Institut Curie, 26 rue d'Ulm, 75005, Paris, France.
- FSHR Theranostics SAS, 11 Rue de Rungis, 75013, Paris, France.
| | - Elisa Anamaria Liehn
- Institute of Molecular Medicine, University of Southern Denmark, 25 J.B. Winslow Vej, 5230, Odense, Denmark
- National Institute of Pathology "Victor Babes", Splaiul Independentei 99-101, 050096, Bucharest, Romania
- National Heart Centre Singapore, 5 Hospital Dr, Singapore, 169609, Singapore
| | | | | | | |
Collapse
|
5
|
Zhang X, Heo GS, Li A, Lahad D, Detering L, Tao J, Gao X, Zhang X, Luehmann H, Sultan D, Lou L, Venkatesan R, Li R, Zheng J, Amrute J, Lin CY, Kopecky BJ, Gropler RJ, Bredemeyer A, Lavine K, Liu Y. Development of a CD163-Targeted PET Radiotracer That Images Resident Macrophages in Atherosclerosis. J Nucl Med 2024; 65:775-780. [PMID: 38548349 PMCID: PMC11064833 DOI: 10.2967/jnumed.123.266910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/26/2024] [Indexed: 05/03/2024] Open
Abstract
Tissue-resident macrophages are complementary to proinflammatory macrophages to promote the progression of atherosclerosis. The noninvasive detection of their presence and dynamic variation will be important to the understanding of their role in the pathogenesis of atherosclerosis. The goal of this study was to develop a targeted PET radiotracer for imaging CD163-positive (CD163+) macrophages in multiple mouse atherosclerosis models and assess the potential of CD163 as a biomarker for atherosclerosis in humans. Methods: CD163-binding peptide was identified using phage display and conjugated with a NODAGA chelator for 64Cu radiolabeling ([64Cu]Cu-ICT-01). CD163-overexpressing U87 cells were used to measure the binding affinity of [64Cu]Cu-ICT-01. Biodistribution studies were performed on wild-type C57BL/6 mice at multiple time points after tail vein injection. The sensitivity and specificity of [64Cu]Cu-ICT-01 in imaging CD163+ macrophages upregulated on the surface of atherosclerotic plaques were assessed in multiple mouse atherosclerosis models. Immunostaining, flow cytometry, and single-cell RNA sequencing were performed to characterize the expression of CD163 on tissue-resident macrophages. Human carotid atherosclerotic plaques were used to measure the expression of CD163+ resident macrophages and test the binding specificity of [64Cu]Cu-ICT-01. Results: [64Cu]Cu-ICT-01 showed high binding affinity to U87 cells. The biodistribution study showed rapid blood and renal clearance with low retention in all major organs at 1, 2, and 4 h after injection. In an ApoE-/- mouse model, [64Cu]Cu-ICT-01 demonstrated sensitive and specific detection of CD163+ macrophages and capability for tracking the progression of atherosclerotic lesions; these findings were further confirmed in Ldlr-/- and PCSK9 mouse models. Immunostaining showed elevated expression of CD163+ macrophages across the plaques. Flow cytometry and single-cell RNA sequencing confirmed the specific expression of CD163 on tissue-resident macrophages. Human tissue characterization demonstrated high expression of CD163+ macrophages on atherosclerotic lesions, and ex vivo autoradiography revealed specific binding of [64Cu]Cu-ICT-01 to human CD163. Conclusion: This work reported the development of a PET radiotracer binding CD163+ macrophages. The elevated expression of CD163+ resident macrophages on human plaques indicated the potential of CD163 as a biomarker for vulnerable plaques. The sensitivity and specificity of [64Cu]Cu-ICT-01 in imaging CD163+ macrophages warrant further investigation in translational settings.
Collapse
Affiliation(s)
- Xiuli Zhang
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Alexandria Li
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Divangana Lahad
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Joan Tao
- Department of Medicine, University of Missouri, Columbia, Missouri
| | - Xuefeng Gao
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Xiaohui Zhang
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Lanlan Lou
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Rajiu Venkatesan
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Ran Li
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Jie Zheng
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Junedh Amrute
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri
| | - Benjamin J Kopecky
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri
| | - Andrea Bredemeyer
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Kory Lavine
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri; and
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, University of Missouri, Columbia, Missouri;
| |
Collapse
|
6
|
Jansen I, Cahalane R, Hengst R, Akyildiz A, Farrell E, Gijsen F, Aikawa E, van der Heiden K, Wissing T. The interplay of collagen, macrophages, and microcalcification in atherosclerotic plaque cap rupture mechanics. Basic Res Cardiol 2024; 119:193-213. [PMID: 38329498 PMCID: PMC11008085 DOI: 10.1007/s00395-024-01033-5] [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: 08/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
The rupture of an atherosclerotic plaque cap overlying a lipid pool and/or necrotic core can lead to thrombotic cardiovascular events. In essence, the rupture of the plaque cap is a mechanical event, which occurs when the local stress exceeds the local tissue strength. However, due to inter- and intra-cap heterogeneity, the resulting ultimate cap strength varies, causing proper assessment of the plaque at risk of rupture to be lacking. Important players involved in tissue strength include the load-bearing collagenous matrix, macrophages, as major promoters of extracellular matrix degradation, and microcalcifications, deposits that can exacerbate local stress, increasing tissue propensity for rupture. This review summarizes the role of these components individually in tissue mechanics, along with the interplay between them. We argue that to be able to improve risk assessment, a better understanding of the effect of these individual components, as well as their reciprocal relationships on cap mechanics, is required. Finally, we discuss potential future steps, including a holistic multidisciplinary approach, multifactorial 3D in vitro model systems, and advancements in imaging techniques. The obtained knowledge will ultimately serve as input to help diagnose, prevent, and treat atherosclerotic cap rupture.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Cahalane
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ranmadusha Hengst
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ali Akyildiz
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank Gijsen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Jiang Y, Sun Z, Ge Z, Tao Z, Liu M, Zhong W, Dong N, Xu L, Wang H, Xu Y, Shen X. Differential expression of Semaphorin-7A /CD163-positive macrophages in large artery and cardiogenic stroke. BMC Neurol 2024; 24:70. [PMID: 38373967 PMCID: PMC10875813 DOI: 10.1186/s12883-024-03559-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Identification of the causes of stroke of undetermined etiology, specifically cardioembolism (CE) and non-CE causes, can inform treatment planning and prognosis prediction. The objective of this study was to analyze the disparities in thrombus composition, particularly Semaphorin-7A (Sema7A) and CD163, between patients diagnosed with large-artery atherosclerosis (LAA) and those with CE, and to investigate their potential association with prognosis. METHODS Thrombi were collected from patients who underwent mechanical thrombectomy at two hospitals. The patients were categorized into two groups: LAA and CE. We compared the levels of Sema7A and CD163 between these groups and analyzed their relationships with stroke severity, hemorrhagic transformation and prognosis. RESULTS The study involved a total of 67 patients. Sema7A expression was found to be significantly higher in the CE group compared to LAA (p < 0.001). Conversely, no statistically significant differences were observed for CD163 between the groups. The presence of Sema7A/CD163 did not show any associations with stroke severity or hemorrhagic transformation (all p > 0.05). However, both Sema7A (OR, 2.017; 95% CI, 1.301-3.518; p = 0.005) and CD163 (OR, 2.283; 95% CI, 1.252-5.724; p = 0.03) were associated with the poor prognosis for stroke, after adjusting for stroke severity. CONCLUSION This study highlights that CE thrombi exhibited higher levels of Sema7A expression compared to LAA thrombi. Moreover, we found a positive correlation between Sema7A/CD163 levels and the poor prognosis of patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Geriatrics, Bengbu Medical College Clinical College of Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Zhichao Sun
- Department of Pathology, Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Zhonglin Ge
- Department of Neurology, Lianyungang Second People's Hospital, Lianyungang, 222000, China.
| | - Zhonghai Tao
- Department of Neurology, Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Mengqian Liu
- Department of Geriatrics, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, 222000, China
| | - Wen Zhong
- Department of Geriatrics, Lianyungang Hospital Affiliated to Jiangsu University, Lianyungang, 222000, China
| | - Nan Dong
- Department of Neurology, Shaoxing Central Hospital, Shaoxing, China
| | - Lei Xu
- Department of Pathology, Lianyungang Second People's Hospital, Lianyungang, 222000, China
| | - Hui Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiwen Xu
- Department of Infectious Disease, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xiaozhu Shen
- Department of Geriatrics, Bengbu Medical College Clinical College of Lianyungang Second People's Hospital, Lianyungang, 222000, China.
| |
Collapse
|
8
|
Gianopoulos I, Daskalopoulou SS. Macrophage profiling in atherosclerosis: understanding the unstable plaque. Basic Res Cardiol 2024; 119:35-56. [PMID: 38244055 DOI: 10.1007/s00395-023-01023-z] [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: 08/09/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 01/22/2024]
Abstract
The development and rupture of atherosclerotic plaques is a major contributor to myocardial infarctions and ischemic strokes. The dynamic evolution of the plaque is largely attributed to monocyte/macrophage functions, which respond to various stimuli in the plaque microenvironment. To this end, macrophages play a central role in atherosclerotic lesions through the uptake of oxidized low-density lipoprotein that gets trapped in the artery wall, and the induction of an inflammatory response that can differentially affect the stability of the plaque in men and women. In this environment, macrophages can polarize towards pro-inflammatory M1 or anti-inflammatory M2 phenotypes, which represent the extremes of the polarization spectrum that include Mhem, M(Hb), Mox, and M4 populations. However, this traditional macrophage model paradigm has been redefined to include numerous immune and nonimmune cell clusters based on in-depth unbiased single-cell approaches. The goal of this review is to highlight (1) the phenotypic and functional properties of monocyte subsets in the circulation, and macrophage populations in atherosclerotic plaques, as well as their contribution towards stable or unstable phenotypes in men and women, and (2) single-cell RNA sequencing studies that have advanced our knowledge of immune, particularly macrophage signatures present in the atherosclerotic niche. We discuss the importance of performing high-dimensional approaches to facilitate the development of novel sex-specific immunotherapies that aim to reduce the risk of cardiovascular events.
Collapse
Affiliation(s)
- Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada.
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine and Health Sciences, McGill University Health Centre, McGill University, Montreal, Canada.
- Department of Medicine, Research Institute of the McGill University Health Centre, Glen Site, 1001 Decarie Boulevard, EM1.2210, Montreal, Quebec, H4A 3J1, Canada.
| |
Collapse
|
9
|
Wieland EB, Kempen LJ, Donners MM, Biessen EA, Goossens P. Macrophage heterogeneity in atherosclerosis: A matter of context. Eur J Immunol 2024; 54:e2350464. [PMID: 37943053 DOI: 10.1002/eji.202350464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
During atherogenesis, plaque macrophages take up and process deposited lipids, trigger inflammation, and form necrotic cores. The traditional inflammatory/anti-inflammatory paradigm has proven insufficient in explaining their complex disease-driving mechanisms. Instead, we now appreciate that macrophages exhibit remarkable heterogeneity and functional specialization in various pathological contexts, including atherosclerosis. Technical advances for studying individual cells, especially single-cell RNA sequencing, indeed allowed to identify novel macrophage subsets in both murine and human atherosclerosis, highlighting the existence of diverse macrophage activation states throughout pathogenesis. In addition, recent studies highlighted the role of the local microenvironment in shaping the macrophages' phenotype and function. However, this remains largely undescribed in the context of atherosclerosis. In this review we explore the origins of macrophages and their functional specialization, shedding light on the diverse sources of macrophage accumulation in the atherosclerotic plaque. Next, we discuss the phenotypic diversity observed in both murine and human atherosclerosis, elucidating their distinct functions and spatial distribution within plaques. Finally, we highlight the importance of the local microenvironment in both phenotypic and functional specialization of macrophages in atherosclerosis and elaborate on the need for spatial multiomics approaches to provide a better understanding of the different macrophage subsets' roles in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Elias B Wieland
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Laura Jap Kempen
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège, Belgium
- Laboratory of Immunophysiology, GIGA Institute, Liege University, Liège, Belgium
| | - Marjo Mpc Donners
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Erik Al Biessen
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Pieter Goossens
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| |
Collapse
|
10
|
Cohen CD, De Blasio MJ, Farrugia GE, Dona MS, Hsu I, Prakoso D, Kiriazis H, Krstevski C, Nash DM, Li M, Gaynor TL, Deo M, Drummond GR, Ritchie RH, Pinto AR. Mapping the cellular and molecular landscape of cardiac non-myocytes in murine diabetic cardiomyopathy. iScience 2023; 26:107759. [PMID: 37736052 PMCID: PMC10509303 DOI: 10.1016/j.isci.2023.107759] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/01/2023] [Accepted: 08/25/2023] [Indexed: 09/23/2023] Open
Abstract
Diabetes is associated with a significantly elevated risk of heart failure. However, despite extensive efforts to characterize the phenotype of the diabetic heart, the molecular and cellular protagonists that underpin cardiac pathological remodeling in diabetes remain unclear, with a notable paucity of data regarding the impact of diabetes on non-myocytes within the heart. Here we aimed to define key differences in cardiac non-myocytes between spontaneously type-2 diabetic (db/db) and healthy control (db/h) mouse hearts. Single-cell transcriptomic analysis revealed a concerted diabetes-induced cellular response contributing to cardiac remodeling. These included cell-specific activation of gene programs relating to fibroblast hyperplasia and cell migration, and dysregulation of pathways involving vascular homeostasis and protein folding. This work offers a new perspective for understanding the cellular mediators of diabetes-induced cardiac pathology, and pathways that may be targeted to address the cardiac complications associated with diabetes.
Collapse
Affiliation(s)
- Charles D. Cohen
- Cardiac Cellular Systems, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, VIC, Australia
| | - Miles J. De Blasio
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Gabriella E. Farrugia
- Cardiac Cellular Systems, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Baker Department of Cardiovascular Research and Implementation, La Trobe University, Melbourne, VIC, Australia
| | - Malathi S.I. Dona
- Cardiac Cellular Systems, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
| | - Ian Hsu
- Cardiac Cellular Systems, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
| | - Darnel Prakoso
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
| | - Helen Kiriazis
- Preclinical Cardiology, Microsurgery and Imaging Platform, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
| | - Crisdion Krstevski
- Cardiac Cellular Systems, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, VIC, Australia
| | - David M. Nash
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
| | - Mandy Li
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
| | - Taylah L. Gaynor
- Cardiac Cellular Systems, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, VIC, Australia
| | - Minh Deo
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, VIC, Australia
| | - Rebecca H. Ritchie
- Heart Failure Pharmacology, Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, VIC, Australia
| | - Alexander R. Pinto
- Cardiac Cellular Systems, Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, VIC, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
11
|
Nienhaus F, Walz M, Rothe M, Jahn A, Pfeiler S, Busch L, Stern M, Heiss C, Vornholz L, Cames S, Cramer M, Schrauwen-Hinderling V, Gerdes N, Temme S, Roden M, Flögel U, Kelm M, Bönner F. Quantitative assessment of angioplasty-induced vascular inflammation with 19F cardiovascular magnetic resonance imaging. J Cardiovasc Magn Reson 2023; 25:54. [PMID: 37784080 PMCID: PMC10546783 DOI: 10.1186/s12968-023-00964-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Macrophages play a pivotal role in vascular inflammation and predict cardiovascular complications. Fluorine-19 magnetic resonance imaging (19F MRI) with intravenously applied perfluorocarbon allows a background-free direct quantification of macrophage abundance in experimental vascular disease models in mice. Recently, perfluorooctyl bromide-nanoemulsion (PFOB-NE) was applied to effectively image macrophage infiltration in a pig model of myocardial infarction using clinical MRI scanners. In the present proof-of-concept approach, we aimed to non-invasively image monocyte/macrophage infiltration in response to carotid artery angioplasty in pigs using 19F MRI to assess early inflammatory response to mechanical injury. METHODS In eight minipigs, two different types of vascular injury were conducted: a mild injury employing balloon oversize angioplasty only (BA, n = 4) and a severe injury provoked by BA in combination with endothelial denudation (BA + ECDN, n = 4). PFOB-NE was administered intravenously three days after injury followed by 1H and 19F MRI to assess vascular inflammatory burden at day six. Vascular response to mechanical injury was validated using X-ray angiography, intravascular ultrasound and immunohistology in at least 10 segments per carotid artery. RESULTS Angioplasty was successfully induced in all eight pigs. Response to injury was characterized by positive remodeling with predominantly adventitial wall thickening and concomitant infiltration of monocytes/macrophages. No severe adverse reactions were observed following PFOB-NE administration. In vivo 19F signals were only detected in the four pigs following BA + ECDN with a robust signal-to-noise ratio (SNR) of 14.7 ± 4.8. Ex vivo analysis revealed a linear correlation of 19F SNR to local monocyte/macrophage cell density. Minimum detection limit of infiltrated monocytes/macrophages was estimated at approximately 410 cells/mm2. CONCLUSIONS In this proof-of-concept study, 19F MRI enabled quantification of monocyte/macrophage infiltration after vascular injury with sufficient sensitivity. This may provide the opportunity to non-invasively monitor vascular inflammation with MRI in patients after angioplasty or even in atherosclerotic plaques.
Collapse
Affiliation(s)
- Fabian Nienhaus
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Moritz Walz
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Maik Rothe
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Annika Jahn
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
- Central Animal Research Facility, Heinrich Heine University, Düsseldorf, Germany
| | - Susanne Pfeiler
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Lucas Busch
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Manuel Stern
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Christian Heiss
- Department of Clinical and Experimental Medicine, University of Surrey, Faculty of Health and Medical Sciences, Guildford, UK
- Department of Vascular Medicine, Surrey and Sussex Healthcare NHS Trust, Redhill, UK
| | - Lilian Vornholz
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Sandra Cames
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Mareike Cramer
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Vera Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sebastian Temme
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Experimental Anesthesiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Florian Bönner
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital and Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Düsseldorf, Germany.
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
12
|
Ravi S, Duraisamy P, Krishnan M, Martin LC, Manikandan B, Ramar M. Sitosterol-rich Digera muricata against 7-ketocholesterol and lipopolysaccharide-mediated atherogenic responses by modulating NF-ΚB/iNOS signalling pathway in macrophages. 3 Biotech 2023; 13:331. [PMID: 37670802 PMCID: PMC10475456 DOI: 10.1007/s13205-023-03741-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023] Open
Abstract
Digera muricata L., commonly known as Tartara, is an edible herb used as traditional medicine in many countries of Africa and Asia. This study aimed to elucidate the effect of a phytosterol-rich extract of D. muricata on 7-ketocholesterol-mediated atherosclerosis in macrophages. The extract was examined by phytochemical analyses, GC-MS, TLC, DPPH scavenging and hRBC membrane stabilization assays. Macrophage polarization was studied with experimental groups framed based on alamar blue cell viability and griess assays. Regulations of arginase enzyme activity, ROS generation, mitochondrial membrane potential, cell membrane integrity, pinocytosis, lipid uptake and peroxidation, as well as, intracellular calcium deposition were determined. In addition, expressions of atherogenic mediators were analysed using PCR, ELISA and immunocytochemistry techniques. Diverse phytochemicals with higher free radical scavenging activity and anti-inflammatory potential have been detected in the D. muricata. Co-treatment with D. muricata markedly reduced the atherogenic responses induced by 7KCh in the presence of LPS such as ROS, especially, NO and O2- along with lipid peroxidation. Furthermore, D. muricata significantly normalized mitochondrial membrane potential, cell membrane integrity, pinocytic activity, intracellular lipid accumulation and calcium deposition. These results provided us with the potentiality of D. muricata in ameliorating atherogenesis. Additionally, it decreased the expression of pro-atherogenic mediators (iNOS, COX-2, MMP9, IL-6, IL-1β, CD36, CD163 and TGFβ1) and increased anti-atherogenic mediators (MRC1 and PPARγ) with high cellular expressions of NF-κB and iNOS. Results showed the potential of sitosterol-rich D. muricata as a versatile biomedical therapeutic agent against abnormal macrophage polarization and its associated pathologies.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| | | | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni’s College for Women, Chennai, 600 015 India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai, 600 025 India
| |
Collapse
|
13
|
Sakamoto A, Suwa K, Kawakami R, Finn AV, Maekawa Y, Virmani R, Finn AV. Significance of Intra-plaque Hemorrhage for the Development of High-Risk Vulnerable Plaque: Current Understanding from Basic to Clinical Points of View. Int J Mol Sci 2023; 24:13298. [PMID: 37686106 PMCID: PMC10487895 DOI: 10.3390/ijms241713298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Acute coronary syndromes due to atherosclerotic coronary artery disease are a leading cause of morbidity and mortality worldwide. Intra-plaque hemorrhage (IPH), caused by disruption of intra-plaque leaky microvessels, is one of the major contributors of plaque progression, causing a sudden increase in plaque volume and eventually plaque destabilization. IPH and its healing processes are highly complex biological events that involve interactions between multiple types of cells in the plaque, including erythrocyte, macrophages, vascular endothelial cells and vascular smooth muscle cells. Recent investigations have unveiled detailed molecular mechanisms by which IPH leads the development of high-risk "vulnerable" plaque. Current advances in clinical diagnostic imaging modalities, such as magnetic resonance image and intra-coronary optical coherence tomography, increasingly allow us to identify IPH in vivo. To date, retrospective and prospective clinical trials have revealed the significance of IPH as detected by various imaging modalities as a reliable prognostic indicator of high-risk plaque. In this review article, we discuss recent advances in our understanding for the significance of IPH on the development of high-risk plaque from basic to clinical points of view.
Collapse
Affiliation(s)
- Atsushi Sakamoto
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Kenichiro Suwa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Rika Kawakami
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Alexandra V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Yuichiro Maekawa
- Division of Cardiology, Internal Medicine III, Hamamatsu University School of Medicine, Hamamatsu 431-3125, Japan; (K.S.); (Y.M.)
| | - Renu Virmani
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| | - Aloke V. Finn
- CVPath Institute, Inc., Gaithersburg, MD 20878, USA; (A.S.); (R.K.); (A.V.F.); (R.V.)
| |
Collapse
|
14
|
Strizova Z, Benesova I, Bartolini R, Novysedlak R, Cecrdlova E, Foley L, Striz I. M1/M2 macrophages and their overlaps - myth or reality? Clin Sci (Lond) 2023; 137:1067-1093. [PMID: 37530555 PMCID: PMC10407193 DOI: 10.1042/cs20220531] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 08/03/2023]
Abstract
Macrophages represent heterogeneous cell population with important roles in defence mechanisms and in homoeostasis. Tissue macrophages from diverse anatomical locations adopt distinct activation states. M1 and M2 macrophages are two polarized forms of mononuclear phagocyte in vitro differentiation with distinct phenotypic patterns and functional properties, but in vivo, there is a wide range of different macrophage phenotypes in between depending on the microenvironment and natural signals they receive. In human infections, pathogens use different strategies to combat macrophages and these strategies include shaping the macrophage polarization towards one or another phenotype. Macrophages infiltrating the tumours can affect the patient's prognosis. M2 macrophages have been shown to promote tumour growth, while M1 macrophages provide both tumour-promoting and anti-tumour properties. In autoimmune diseases, both prolonged M1 activation, as well as altered M2 function can contribute to their onset and activity. In human atherosclerotic lesions, macrophages expressing both M1 and M2 profiles have been detected as one of the potential factors affecting occurrence of cardiovascular diseases. In allergic inflammation, T2 cytokines drive macrophage polarization towards M2 profiles, which promote airway inflammation and remodelling. M1 macrophages in transplantations seem to contribute to acute rejection, while M2 macrophages promote the fibrosis of the graft. The view of pro-inflammatory M1 macrophages and M2 macrophages suppressing inflammation seems to be an oversimplification because these cells exploit very high level of plasticity and represent a large scale of different immunophenotypes with overlapping properties. In this respect, it would be more precise to describe macrophages as M1-like and M2-like.
Collapse
Affiliation(s)
- Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Robin Bartolini
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Rene Novysedlak
- Third Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Eva Cecrdlova
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lily Koumbas Foley
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
15
|
van Dijk RA, Kleemann R, Schaapherder AF, van den Bogaerdt A, Hedin U, Matic L, Lindeman JH. Validating human and mouse tissues commonly used in atherosclerosis research with coronary and aortic reference tissue: similarities but profound differences in disease initiation and plaque stability. JVS Vasc Sci 2023; 4:100118. [PMID: 37810738 PMCID: PMC10551657 DOI: 10.1016/j.jvssci.2023.100118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 10/10/2023] Open
Abstract
Objective Characterization of the atherosclerotic process fully relies on histological evaluation and staging through a consensus grading system. So far, a head-to-head comparison of atherosclerotic process in experimental models and tissue resources commonly applied in atherosclerosis research with the actual human atherosclerotic process is missing. Material and Methods Aspects of the atherosclerotic process present in established murine atherosclerosis models and human carotid endarterectomy specimen were systematically graded using the modified American Heart Association histological classification (Virmani classification). Aspects were aligned with the atherosclerotic process observed in human coronary artery and aortic atherosclerosis reference tissues that were available through biobanks based on human tissue/organ donor material. Results Apart from absent intraplaque hemorrhages in aortic lesions, the histological characteristics of the different stages of human coronary and aortic atherosclerosis are similar. Carotid endarterectomy samples all represent end-stage "fibrous calcified plaque" lesions, although secondary, progressive, and vulnerable lesions with gross morphologies similar to coronary/aortic lesions occasionally present along the primary lesions. For the murine lesions, clear histological parallels were observed for the intermediate lesion types ("pathological intimal thickening," and "early fibroatheroma"). However, none of the murine lesions studied progressed to an equivalent of late fibroatheroma or beyond. Notable contrasts were observed for disease initiation: whereas disease initiation in humans is characterized by a mesenchymal cell influx in the intima, the earliest murine lesions are exclusively intimal, with subendothelial accumulation foam cells. A mesenchymal (and medial) response are absent. In fact, it is concluded that the stage of "adaptive intimal thickening" is absent in all mouse models included in this study. Conclusions The Virmani classification for coronary atherosclerosis can be applied for systematically grading experimental and clinical atherosclerosis. Application of this histological grading tool shows clear parallels for intermediate human and murine atherosclerotic lesions. However, clear contrasts are observed for disease initiation, and late stage atherosclerotic lesions. Carotid endarterectomy all represent end-stage fibrous calcified plaque lesions, although secondary earlier lesions may present in a subset of samples.
Collapse
Affiliation(s)
- Rogier A. van Dijk
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Kleemann
- The Netherlands Organization for Applied Scientific Research (TNO), Department of Metabolic Health Research, TNO Metabolic Health Research, Leiden, The Netherlands
| | | | | | - Ulf Hedin
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Ljubica Matic
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Jan H.N. Lindeman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
16
|
Sun J, Singh P, Shami A, Kluza E, Pan M, Djordjevic D, Michaelsen NB, Kennbäck C, van der Wel NN, Orho-Melander M, Nilsson J, Formentini I, Conde-Knape K, Lutgens E, Edsfeldt A, Gonçalves I. Spatial Transcriptional Mapping Reveals Site-Specific Pathways Underlying Human Atherosclerotic Plaque Rupture. J Am Coll Cardiol 2023; 81:2213-2227. [PMID: 37286250 DOI: 10.1016/j.jacc.2023.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/20/2023] [Accepted: 04/03/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Atherosclerotic plaque ruptures, triggered by blood flow-associated biomechanical forces, cause most myocardial infarctions and strokes. OBJECTIVES This study aims to investigate the exact location and underlying mechanisms of atherosclerotic plaque ruptures, identifying therapeutic targets against cardiovascular events. METHODS Histology, electron microscopy, bulk and spatial RNA sequencing on human carotid plaques were studied in proximal, most stenotic, and distal regions along the longitudinal blood flow direction. Genome-wide association studies were used to examine heritability enrichment and causal relationships of atherosclerosis and stroke. Associations between top differentially expressed genes (DEGs) and preoperative and postoperative cardiovascular events were examined in a validation cohort. RESULTS In human carotid atherosclerotic plaques, ruptures predominantly occurred in the proximal and most stenotic regions but not in the distal region. Histologic and electron microscopic examination showed that proximal and most stenotic regions exhibited features of plaque vulnerability and thrombosis. RNA sequencing identified DEGs distinguishing the proximal and most stenotic regions from the distal region which were deemed as most relevant to atherosclerosis-associated diseases as shown by heritability enrichment analyses. The identified pathways associated with the proximal rupture-prone regions were validated by spatial transcriptomics, firstly in human atherosclerosis. Of the 3 top DEGs, matrix metallopeptidase 9 emerged particularly because Mendelian randomization suggested that its high circulating levels were causally associated with atherosclerosis risk. CONCLUSIONS Our findings show plaque site-specific transcriptional signatures associated with proximal rupture-prone regions of carotid atherosclerotic plaques. This led to the geographical mapping of novel therapeutic targets, such as matrix metallopeptidase 9, against plaque rupture.
Collapse
Affiliation(s)
- Jiangming Sun
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Pratibha Singh
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Annelie Shami
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Ewelina Kluza
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Center, Amsterdam, the Netherlands; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mengyu Pan
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | | | - Natasha Barascuk Michaelsen
- Type 2 Diabetes and Cardiovascular Disease Research, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Cecilia Kennbäck
- Clinical Research Unit, Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Nicole N van der Wel
- Electron Microscopy Center Amsterdam, Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | | | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | | | | | - Esther Lutgens
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Center, Amsterdam, the Netherlands; Cardiovascular Medicine, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Department of Cardiology, Skåne University Hospital, Malmö, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Department of Cardiology, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
17
|
Hamann B, Klimova A, Klotz F, Frank F, Jänichen C, Kapalla M, Sabarstinski P, Wolk S, Morawietz H, Poitz DM, Hofmann A, Reeps C. Regulation of CD163 Receptor in Patients with Abdominal Aortic Aneurysm and Associations with Antioxidant Enzymes HO-1 and NQO1. Antioxidants (Basel) 2023; 12:antiox12040947. [PMID: 37107322 PMCID: PMC10135987 DOI: 10.3390/antiox12040947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Red blood cells are found within the abdominal aortic aneurysm (AAA), in the intraluminal thrombus (ILT), and in neovessels. Hemolysis promotes aortic degeneration, e.g., by heme-induced reactive oxygen species formation. To reduce its toxicity, hemoglobin is endocytosed by the CD163 receptor and heme is degraded by heme oxygenase-1 (HO-1). A soluble form (sCD163) is discussed as an inflammatory biomarker representing the activation of monocytes and macrophages. HO-1 and NAD(P)H quinone dehydrogenase 1 (NQO1) are antioxidant genes that are induced by the Nrf2 transcription factor, but their regulation in AAA is only poorly understood. The aim of the present study was to analyze linkages between CD163, Nrf2, HO-1, and NQO1 and to clarify if plasma sCD163 has diagnostic and risk stratification potential. Soluble CD163 was 1.3-fold (p = 0.015) higher in AAA compared to patients without arterial disease. The difference remained significant after adjusting for age and sex. sCD163 correlated with the thickness of the ILT (rs = 0.26; p = 0.02) but not with the AAA diameter or volume. A high aneurysmal CD163 mRNA was connected to increases in NQO1, HMOX1, and Nrf2 mRNA. Further studies are needed to analyze the modulation of the CD163/HO-1/NQO1 pathway with the overall goal of minimizing the detrimental effects of hemolysis.
Collapse
Affiliation(s)
- Bianca Hamann
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Anna Klimova
- Core Unit Data Management and Analytics, National Center for Tumor Diseases Dresden (NCT/UCC), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Felicia Klotz
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Frieda Frank
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Christian Jänichen
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Marvin Kapalla
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Pamela Sabarstinski
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Steffen Wolk
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Anja Hofmann
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Christian Reeps
- Division of Vascular and Endovascular Surgery, Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, D-01307 Dresden, Germany
| |
Collapse
|
18
|
Belhoul-Fakir H, Wu J, Yeow YL, Musk GC, Kershaw H, Ingley E, Zhao BS, Reid CM, Lagat C, Evans B, Thompson PL, Brown ML, Hamzah J, Jansen S. Injury to the tunica media initiates atherogenesis in the presence of hyperlipidemia. Front Cardiovasc Med 2023; 10:1152124. [PMID: 37063951 PMCID: PMC10098105 DOI: 10.3389/fcvm.2023.1152124] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/10/2023] [Indexed: 03/31/2023] Open
Abstract
Background and aims Fatty streaks initiating the formation of atheromatous plaque appear in the tunica intima. The tunica media is not known to be a nidus for lipid accumulation initiating atherogenesis. We assessed changes to the tunica media in response to a micro-injury produced in the pig aorta. In addition, we assessed human carotid endarterectomy plaques for indication of atheroma initiation in the tunica media. Methods Three healthy landrace female pigs underwent laparotomy to inject autologous blood and create micro-hematomas at 6 sites within the tunica media of the infrarenal abdominal aorta. These pigs were fed a high-fat diet (HFD) for 4-12 weeks. Post-mortem aortas from all pigs, including a control group of healthy pigs, were serially stained to detect lipid deposits, vasa vasora (VV), immune cell infiltration and inflammatory markers, as well as changes to the vascular smooth muscle cell (vSMC) compartment. Moreover, 25 human carotid endarterectomy (CEA) specimens were evaluated for their lipid composition in the tunica media and intima. Results High lipid clusters, VV density, and immune cell infiltrates were consistently observed at 5 out of 6 injection sites under prolonged hyperlipidemia. The hyperlipidemic diet also affected the vSMC compartment in the tunica media adjacent to the tunica adventitia, which correlated with VV invasion and immune cell infiltration. Analysis of human carotid specimens post-CEA indicated that 32% of patients had significantly greater atheroma in the tunica media than in the arterial intima. Conclusion The arterial intima is not the only site for atherosclerosis initiation. We show that injury to the media can trigger atherogenesis.
Collapse
Affiliation(s)
- Hanane Belhoul-Fakir
- Curtin Medical School, Curtin University, Bentley, Perth, WA, Australia
- Laboratory of Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII MedicalCentre, Nedlands, WA, Australia
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
| | - Jiansha Wu
- Laboratory of Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII MedicalCentre, Nedlands, WA, Australia
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
| | - Yen L. Yeow
- Laboratory of Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII MedicalCentre, Nedlands, WA, Australia
| | - Gabrielle C. Musk
- Animal Care Services, The University of Western Australia, Crawley, Perth, WA, Australia
| | - Helen Kershaw
- Animal Care Services, The University of Western Australia, Crawley, Perth, WA, Australia
| | - Evan Ingley
- Discipline of Medical, Molecular, and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
- School of Biomedical Sciences, Pharmacology, and Toxicology, The University of Western Australia, Perth, WA, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Bichen Sophie Zhao
- Curtin Medical School, Curtin University, Bentley, Perth, WA, Australia
- Laboratory of Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII MedicalCentre, Nedlands, WA, Australia
| | - Christopher M. Reid
- School of Public Health and Preventive Medicine, Monash University, Clayton, VIC, Australia
- School of Population Health, Curtin University, Bentley, Perth, WA, Australia
| | - Christopher Lagat
- Western Australia School of Mine: Minerals, Energy and Chemical Engineering, Curtin University, Kensington, Perth, WA, Australia
| | - Brian Evans
- Western Australia School of Mine: Minerals, Energy and Chemical Engineering, Curtin University, Kensington, Perth, WA, Australia
| | - Peter L. Thompson
- Laboratory of Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII MedicalCentre, Nedlands, WA, Australia
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
| | - Michael L. Brown
- School of Population Health, Curtin University, Bentley, Perth, WA, Australia
| | - Juliana Hamzah
- Curtin Medical School, Curtin University, Bentley, Perth, WA, Australia
- Laboratory of Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII MedicalCentre, Nedlands, WA, Australia
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
| | - Shirley Jansen
- Curtin Medical School, Curtin University, Bentley, Perth, WA, Australia
- Laboratory of Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII MedicalCentre, Nedlands, WA, Australia
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
- Department of Vascular and Endovascular Surgery, Sir Charles Gairdner Hospital, Nedlands, Perth, WA, Australia
| |
Collapse
|
19
|
Duan R, Liu Y, Tang D, Xiao S, Lin R, Zhao M. Single-cell RNA-Seq reveals CVI-mAb-induced Lyve1 + M2-like macrophages reduce atherosclerotic plaque area in Apoe -/- mice. Int Immunopharmacol 2023; 116:109794. [PMID: 36736225 DOI: 10.1016/j.intimp.2023.109794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Atherosclerosis is a lipid imbalance-induced autoimmune disease. Macrophages participate in the development and progression of atherosclerosis. Although numerous studies have utilized single-cell RNA sequencing to identify the role of various macrophage phenotypes in atherosclerosis, the macrophage subpopulations that have therapeutic benefits against atherosclerosis are not fully understood. METHODS In this study, a single-cell RNA sequencing analysis was performed on the F4/80+ macrophages of apolipoprotein E-deficient (Apoe-/-) mice on a normal diet (ND), a high-fat diet (HFD), and a high-fat diet (HFD) with collagen VI monoclonal antibodies (CVI-mAb) treatment. A population of M2-like macrophages expressing the hyaluronan receptor Lyve1 was almost exclusively detectable in Apoe-/- mice on an HFD with CVI-mAb treatment, compared with other groups. Differential gene expression and gene ontology enrichment analyses revealed specific gene expression patterns that distinguished this macrophage subset and uncovered its functions. RESULTS Lyve1+ M2 macrophages appear to have specialized functions in lipid metabolism. Lyve1+ M2-like macrophages were sorted via fluorescence- activated cell sorting (FACS) and adoptively transferred to Apoe-/- mice fed an HFD. CONCLUSION Our result showed that Lyve1+ M2 macrophages could reduce the plaque areas in Apoe-/- mice.
Collapse
Affiliation(s)
- Rui Duan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Liu
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Dongmei Tang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sujun Xiao
- The Affiliated Nanhua Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Run Lin
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ming Zhao
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
20
|
Chiorescu RM, Mocan M, Inceu AI, Buda AP, Blendea D, Vlaicu SI. Vulnerable Atherosclerotic Plaque: Is There a Molecular Signature? Int J Mol Sci 2022; 23:13638. [PMID: 36362423 PMCID: PMC9656166 DOI: 10.3390/ijms232113638] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis and its clinical manifestations, coronary and cerebral artery diseases, are the most common cause of death worldwide. The main pathophysiological mechanism for these complications is the rupture of vulnerable atherosclerotic plaques and subsequent thrombosis. Pathological studies of the vulnerable lesions showed that more frequently, plaques rich in lipids and with a high level of inflammation, responsible for mild or moderate stenosis, are more prone to rupture, leading to acute events. Identifying the vulnerable plaques helps to stratify patients at risk of developing acute vascular events. Traditional imaging methods based on plaque appearance and size are not reliable in prediction the risk of rupture. Intravascular imaging is a novel technique able to identify vulnerable lesions, but it is invasive and an operator-dependent technique. This review aims to summarize the current data from literature regarding the main biomarkers involved in the attempt to diagnose vulnerable atherosclerotic lesions. These biomarkers could be the base for risk stratification and development of the new therapeutic drugs in the treatment of patients with vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Roxana Mihaela Chiorescu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Mihaela Mocan
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine, 400349 Cluj-Napoca, Romania
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Andreea Paula Buda
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Dan Blendea
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
- Department of Cardiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400437 Cluj-Napoca, Romania
| | - Sonia Irina Vlaicu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| |
Collapse
|
21
|
Zhang L, Li X, Lyu Q, Shi G. Imaging diagnosis and research progress of carotid plaque vulnerability. JOURNAL OF CLINICAL ULTRASOUND : JCU 2022; 50:905-912. [PMID: 35801515 DOI: 10.1002/jcu.23266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 05/26/2022] [Accepted: 06/16/2022] [Indexed: 06/15/2023]
Abstract
Ischemic stroke (IS) exhibits a high disability rate, mortality, and recurrence rate, imposing a serious threat to human survival and health. Its occurrence is affected by various factors. Although the previous research has demonstrated that the occurrence of IS is mainly associated with lumen stenosis caused by carotid atherosclerotic plaque (AP), recent studies have revealed that many patients will still suffer from IS even with mild carotid artery lumen stenosis. Blood supply disturbance causes 10% of IS to the corresponding cerebral blood supply area caused by carotid vulnerable plaque. Thrombus blockage of distal branch vessels caused by rupture of vulnerable carotid plaque is the main cause of ischemic stroke. Therefore, how to accurately evaluate vulnerable plaque and intervene as soon as possible is a problem that needs to be solved in clinic. The vulnerability of plaque is determined by its internal components, including thin and incomplete fibrous cap, necrotic lipid core, intra-plaque hemorrhage, intra-plaque neovascularization, and ulcerative plaque formation. The development of imaging technology enables the routine detection of AP vulnerability. By analyzing the pathological changes, characteristics, and formation mechanism of carotid plaque vulnerability, this article aims to explore the modern imaging methods which can be used to identify plaque composition and plaque vulnerability to provide a reference basis for disease diagnosis and differential diagnosis.
Collapse
Affiliation(s)
- Lianlian Zhang
- Yancheng Clinical College of Xuzhou Medical University, The First peolie's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Xia Li
- Affiliated Hospital of Jiangsu medical vocational college, The Third People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Qi Lyu
- Taizhou People's Hospital, Taizhou, China
| | - Guofu Shi
- Affiliated Hospital of Jiangsu medical vocational college, The Third People's Hospital of Yancheng, Yancheng, Jiangsu, China
| |
Collapse
|
22
|
Identification Markers of Carotid Vulnerable Plaques: An Update. Biomolecules 2022; 12:biom12091192. [PMID: 36139031 PMCID: PMC9496377 DOI: 10.3390/biom12091192] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 12/02/2022] Open
Abstract
Vulnerable plaques have been a hot topic in the field of stroke and carotid atherosclerosis. Currently, risk stratification and intervention of carotid plaques are guided by the degree of luminal stenosis. Recently, it has been recognized that the vulnerability of plaques may contribute to the risk of stroke. Some classical interventions, such as carotid endarterectomy, significantly reduce the risk of stroke in symptomatic patients with severe carotid stenosis, while for asymptomatic patients, clinically silent plaques with rupture tendency may expose them to the risk of cerebrovascular events. Early identification of vulnerable plaques contributes to lowering the risk of cerebrovascular events. Previously, the identification of vulnerable plaques was commonly based on imaging technologies at the macroscopic level. Recently, some microscopic molecules pertaining to vulnerable plaques have emerged, and could be potential biomarkers or therapeutic targets. This review aimed to update the previous summarization of vulnerable plaques and identify vulnerable plaques at the microscopic and macroscopic levels.
Collapse
|
23
|
Mutengo KH, Masenga SK, Mwesigwa N, Patel KP, Kirabo A. Hypertension and human immunodeficiency virus: A paradigm for epithelial sodium channels? Front Cardiovasc Med 2022; 9:968184. [PMID: 36093171 PMCID: PMC9452753 DOI: 10.3389/fcvm.2022.968184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/10/2022] [Indexed: 02/03/2023] Open
Abstract
Hypertension is a risk factor for end organ damage and death and is more common in persons with HIV compared to the general population. Several mechanisms have been studied in the pathogenesis of hypertension. Current evidence suggests that the epithelial sodium channel (ENaC) plays a key role in regulating blood pressure through the transport of sodium and water across membranes in the kidney tubules, resulting in retention of sodium and water and an altered fluid balance. However, there is scarcity of information that elucidates the role of ENaC in HIV as it relates to increasing the risk for development or pathogenesis of hypertension. This review summarized the evidence to date implicating a potential role for altered ENaC activity in contributing to hypertension in patients with HIV.
Collapse
Affiliation(s)
- Katongo H. Mutengo
- School of Medicine and Health Sciences, HAND Research Group, Mulungushi University, Livingstone Campus, Livingstone, Zambia,School of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Sepiso K. Masenga
- School of Medicine and Health Sciences, HAND Research Group, Mulungushi University, Livingstone Campus, Livingstone, Zambia,School of Public Health and Medicine, University of Zambia, Lusaka, Zambia
| | - Naome Mwesigwa
- Department of Medicine and Dentistry, Kampala International University, Kampala, Uganda
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States,*Correspondence: Annet Kirabo,
| |
Collapse
|
24
|
Khare HA, Døssing KBV, Ringgaard L, Christensen E, Urbak L, Sillesen H, Ripa RS, Binderup T, Pedersen SF, Kjaer A. In vivo detection of urokinase-type plasminogen activator receptor (uPAR) expression in arterial atherogenesis using [64Cu]Cu-DOTA-AE105 positron emission tomography (PET). Atherosclerosis 2022; 352:103-111. [DOI: 10.1016/j.atherosclerosis.2022.03.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 12/21/2022]
|
25
|
Hyperglycemia Induces Inflammatory Response of Human Macrophages to CD163-Mediated Scavenging of Hemoglobin-Haptoglobin Complexes. Int J Mol Sci 2022; 23:ijms23031385. [PMID: 35163309 PMCID: PMC8836198 DOI: 10.3390/ijms23031385] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
Hyperglycemia, a hallmark of diabetes, can induce inflammatory programming of macrophages. The macrophage scavenger receptor CD163 internalizes and degrades hemoglobin-haptoglobin (Hb-Hp) complexes built due to intravascular hemolysis. Clinical studies have demonstrated a correlation between impaired scavenging of Hb-Hp complexes via CD163 and diabetic vascular complications. Our aim was to identify whether hyperglycemia is able to amplify inflammation via Hb-Hp complex interactions with the immune system. M(IFNγ), M(IL-4), and control M0 macrophages were differentiated out of primary human monocytes in normo- (5 mM) and hyperglycemic (25 mM) conditions. CD163 gene expression was decreased 5.53 times in M(IFNγ) with a further decrease of 1.99 times in hyperglycemia. Hyperglycemia suppressed CD163 surface expression in M(IFNγ) (1.43 times). Flow cytometry demonstrated no impairment of Hb-Hp uptake in hyperglycemia. However, hyperglycemia induced an inflammatory response of M(IFNγ) to Hb-Hp1-1 and Hb-Hp2-2 uptake with different dynamics. Hb-Hp1-1 uptake stimulated IL-6 release (3.03 times) after 6 h but suppressed secretion (5.78 times) after 24 h. Contrarily, Hb-Hp2-2 uptake did not affect IL-6 release after 6h but increased secretion after 24 h (3.06 times). Our data show that hyperglycemia induces an inflammatory response of innate immune cells to Hb-Hp1-1 and Hb-Hp2-2 uptake, converting the silent Hb-Hp complex clearance that prevents vascular damage into an inflammatory process, hereby increasing the susceptibility of diabetic patients to vascular complications.
Collapse
|
26
|
Single-Cell RNA-Seq Reveals a Crosstalk between Hyaluronan Receptor LYVE-1-Expressing Macrophages and Vascular Smooth Muscle Cells. Cells 2022; 11:cells11030411. [PMID: 35159221 PMCID: PMC8834524 DOI: 10.3390/cells11030411] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Atherosclerosis is a chronic inflammatory disease where macrophages participate in the progression of the disease. However, the role of resident-like macrophages (res-like) in the atherosclerotic aorta is not completely understood. Methods: A single-cell RNA sequencing analysis of CD45+ leukocytes in the atherosclerotic aorta of apolipoprotein E–deficient (Apoe−/−) mice on a normal cholesterol diet (NCD) or a high cholesterol diet (HCD), respecting the side-to-specific predisposition to atherosclerosis, was performed. A population of res-like macrophages expressing hyaluronan receptor LYVE-1 was investigated via flow cytometry, co-culture experiments, and immunofluorescence in human atherosclerotic plaques from carotid artery disease patients (CAD). Results: We identified 12 principal leukocyte clusters with distinct atherosclerosis disease-relevant gene expression signatures. LYVE-1+ res-like macrophages, expressing a high level of CC motif chemokine ligand 24 (CCL24, eotaxin-2), expanded under hypercholesteremia in Apoe−/− mice and promoted VSMC phenotypic modulation to osteoblast/chondrocyte-like cells, ex vivo, in a CCL24-dependent manner. Moreover, the abundance of LYVE-1+CCL24+ macrophages and elevated systemic levels of CCL24 were associated with vascular calcification and CAD events. Conclusions: LYVE-1 res-like macrophages, via the secretion of CCL24, promote the transdifferentiation of VSMC to osteogenic-like cells with a possible role in vascular calcification and likely a detrimental role in atherosclerotic plaque destabilization.
Collapse
|
27
|
Qian C, Jing Y, Xia M, Ye Q. Comprehensive analysis of dysregulated genes associated with atherosclerotic plaque destabilization. Exp Biol Med (Maywood) 2021; 246:2487-2494. [PMID: 34308657 DOI: 10.1177/15353702211033247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Atherosclerotic plaque destabilization is a dominating cause of acute cardiovascular events such as myocardial infarction and stroke. This study aims to identify genetic biomarkers related to atherosclerotic plaque destabilization using bioinformatics. Three transcriptome datasets of human carotid atherosclerotic plaque samples were downloaded from ArrayExpress and Gene Expression Omnibus databases, including E-MATB-2055, E-TABM-190, and GSE120521. With Robust Rank Aggregation analysis, we documented 46 differentially expressed genes between stable and unstable/ruptured plaques. Functional enrichment analysis using DAVID tool demonstrated that these genes were mainly related to biological functions such as extracellular matrix disassembly, collagen catabolic process, response to mechanical stimulus, and PPAR signaling pathway. A protein-protein interaction network for the differentially expressed genes was constructed, and eight pivotal genes (ITGAM, MMP9, PLAUR, CCR1, CD163, CD36, ADAM8, and IL1RN) were obtained from the network with a connective degree > 5. The expression patterns of these hub differentially expressed genes could be verified in atherosclerotic plaque samples with intraplaque hemorrhage. Using gene set variation analysis, the eight genes were integrated to generate an atherosclerotic plaque destabilization score, which showed a high performance in not only discriminating individuals with myocardial infarction from those with stable coronary illness, but also in predicting future acute cardiovascular events in atherosclerotic patients. In conclusion, the findings of this study will enhance our knowledge on the pathological mechanisms involved in atherosclerotic plaque destabilization, and provide potential gene biomarkers for risk stratification of patients with atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yuling Jing
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Meng Xia
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|