1
|
Machiraju P, Srinivas R, Kannan R, George R, Heymans S, Mukhopadhyay R, Ghosh A. Paired Transcriptomic Analyses of Atheromatous and Control Vessels Reveal Novel Autophagy and Immunoregulatory Genes in Peripheral Artery Disease. Cells 2024; 13:1269. [PMID: 39120300 PMCID: PMC11312159 DOI: 10.3390/cells13151269] [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: 06/15/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Peripheral artery disease (PAD), a significant health burden worldwide, affects lower extremities due to atherosclerosis in peripheral vessels. Although the mechanisms of PAD have been well studied, the molecular milieu of the plaques localized within peripheral arteries are not well understood. Thus, to identify PAD-lesion-specific gene expression profiles precluding genetic, environmental, and dietary biases, we studied the transcriptomic profile of nine plaque tissues normalized to non-plaque tissues from the same donors. A total of 296 upregulated genes, 274 downregulated genes, and 186 non-coding RNAs were identified. STAG1, SPCC3, FOXQ1, and E2F3 were key downregulated genes, and CD93 was the top upregulated gene. Autophagosome assembly, cellular response to UV, cytoskeletal organization, TCR signaling, and phosphatase activity were the key dysregulated pathways identified. Telomerase regulation and autophagy were identified as novel interacting pathways using network analysis. The plaque tissue was predominantly composed of immune cells and dedifferentiated cell populations indicated by cell-specific marker-imputed gene expression analysis. This study identifies novel genes, non-coding RNAs, associated regulatory pathways, and the cell composition of the plaque tissue in PAD patients. The autophagy and immunoregulatory genes may drive novel mechanisms, resulting in atheroma. These novel interacting networks and genes have potential for PAD-specific therapeutic applications.
Collapse
Affiliation(s)
- Praveen Machiraju
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore 560099, India; (P.M.); (R.K.)
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Rajesh Srinivas
- Department of Vascular and Endovascular Surgery, Narayana Health, Bangalore 560099, India; (R.S.); (R.G.)
| | - Ramaraj Kannan
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore 560099, India; (P.M.); (R.K.)
| | - Robbie George
- Department of Vascular and Endovascular Surgery, Narayana Health, Bangalore 560099, India; (R.S.); (R.G.)
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, bus911, 3000 Leuven, Belgium
| | - Rupak Mukhopadhyay
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore 560099, India; (P.M.); (R.K.)
| |
Collapse
|
2
|
Veeckmans G, Van San E, Vanden Berghe T. A guide to ferroptosis, the biological rust of cellular membranes. FEBS J 2024; 291:2767-2783. [PMID: 37935445 DOI: 10.1111/febs.16993] [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: 07/21/2023] [Revised: 10/19/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Unprotected iron can rust due to oxygen exposure. Similarly, in our body, oxidative stress can kill cells in an iron-dependent manner, which can give rise to devastating diseases. This type of cell death is referred to as ferroptosis. Generally, ferroptosis is defined as an iron-catalyzed form of regulated necrosis that occurs through excessive peroxidation of polyunsaturated fatty acids within cellular membranes. This review summarizes how ferroptosis is executed by a rather primitive biochemical process, under tight regulation of lipid, iron, and redox metabolic processes. An overview is given of major classes of ferroptosis inducers and inhibitors, and how to detect ferroptosis. Finally, its detrimental role in disease is briefly discussed.
Collapse
Affiliation(s)
| | - Emily Van San
- Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Tom Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Belgium
- VIB-UGent Center for Inflammation Research, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| |
Collapse
|
3
|
Ma C, Lu T, He Y, Guo D, Duan L, Jia R, Cai D, Gao T, Chen Z, Xue B, Li T, He Y. Comprehensive analysis of autophagy-related gene expression profiles identified five gene biomarkers associated with immune infiltration and advanced plaques in carotid atherosclerosis. Orphanet J Rare Dis 2023; 18:66. [PMID: 36959587 PMCID: PMC10037854 DOI: 10.1186/s13023-023-02660-2] [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: 10/20/2022] [Accepted: 03/11/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Autophagy plays an important role in the progression of carotid atherosclerosis (CAS). This study aimed to identify hub autophagy-related genes (ATGs) associated with CAS. METHODS GSE43292 and GSE28829 datasets of early and advanced CAS plaques were enrolled from the Gene Expression Omnibus (GEO) database. A comprehensive analysis of differentially expressed ATGs (DE-ATGs) was conducted. Functional enrichment assay was used to explore biological functions of DE-ATGs. The hub ATGs were identified by protein-protein interaction (PPI) network. Immunohistochemistry (IHC) and Real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to validate hub ATGs at the protein level and mRNA level. Correlation analysis of hub ATGs with immune cells was also conducted. In addition, a competitive endogenous RNA (ceRNA) network was constructed, and diagnostic value of hub ATGs was evaluated. RESULTS A total of 19 DE-ATGs were identified in early and advanced CAS plaques. Functional enrichment analysis of DE-ATGs suggested that they were closely correlated to autophagy, apoptosis, and lipid regulation. Moreover, 5 hub ATGs, including TNFSF10, ITGA6, CTSD, CCL2, and CASP1, were identified and further verified by IHC. The area under the curve (AUC) values of the 5 hub ATGs were 0.818, 0.732, 0.792, 0.814, and 0.812, respectively. Competing endogenous RNA (ceRNA) networks targeting the hub ATGs were also constructed. In addition, the 5 hub ATGs were found to be closely associated with immune cell infiltration in CAS. CONCLUSION In this study, we identified 5 hub ATGs including CASP1, CCL2, CTSD, ITGA6 and TNFSF10, which could serve as candidate diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Chi Ma
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China
| | - Taoyuan Lu
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China
| | - Yanyan He
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China
- Department of Cerebrovascular Disease, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Dehua Guo
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China
| | - Lin Duan
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China
- Department of Cerebrovascular Disease, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Rufeng Jia
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China
| | - Dongyang Cai
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Tao Gao
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Zhongcan Chen
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Binghua Xue
- Department of Endocrinology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Tianxiao Li
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China.
- Department of Cerebrovascular Disease, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.
| | - Yingkun He
- Department of Cerebrovascular Disease, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China.
- Henan Provincial NeuroInterventional Engineering Research Center, Henan International Joint Laboratory of Cerebrovascular Disease, and Henan Engineering Research Center of Cerebrovascular Intervention Innovation, Zhengzhou, Henan, 450003, China.
| |
Collapse
|
4
|
Puylaert P, Zurek M, Rayner KJ, De Meyer GRY, Martinet W. Regulated Necrosis in Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:1283-1306. [PMID: 36134566 DOI: 10.1161/atvbaha.122.318177] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During atherosclerosis, lipid-rich plaques are formed in large- and medium-sized arteries, which can reduce blood flow to tissues. This situation becomes particularly precarious when a plaque develops an unstable phenotype and becomes prone to rupture. Despite advances in identifying and treating vulnerable plaques, the mortality rate and disability caused by such lesions remains the number one health threat in developed countries. Vulnerable, unstable plaques are characterized by a large necrotic core, implying a prominent role for necrotic cell death in atherosclerosis and plaque destabilization. Necrosis can occur accidentally or can be induced by tightly regulated pathways. Over the past decades, different forms of regulated necrosis, including necroptosis, ferroptosis, pyroptosis, and secondary necrosis, have been identified, and these may play an important role during atherogenesis. In this review, we describe several forms of necrosis that may occur in atherosclerosis and how pharmacological modulation of these pathways can stabilize vulnerable plaques. Moreover, some challenges of targeting necrosis in atherosclerosis such as the presence of multiple death-inducing stimuli in plaques and extensive cross-talk between necrosis pathways are discussed. A better understanding of the role of (regulated) necrosis in atherosclerosis and the mechanisms contributing to plaque destabilization may open doors to novel pharmacological strategies and will enable clinicians to tackle the residual cardiovascular risk that remains in many atherosclerosis patients.
Collapse
Affiliation(s)
- Pauline Puylaert
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Michelle Zurek
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology and Immunology and Centre for Infection, Immunity and Inflammation, Faculty of Medicine, University of Ottawa, ON, Canada (K.J.R.).,University of Ottawa Heart Institute, ON, Canada (K.J.R.)
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| | - Wim Martinet
- Laboratory of Physiopharmacology and Infla-Med Centre of Excellence, University of Antwerp, Belgium (P.P., M.Z., G.R.Y.D.M., W.M.)
| |
Collapse
|
5
|
Durda P, Raffield LM, Lange EM, Olson NC, Jenny NS, Cushman M, Deichgraeber P, Grarup N, Jonsson A, Hansen T, Mychaleckyj JC, Psaty BM, Reiner AP, Tracy RP, Lange LA. Circulating Soluble CD163, Associations With Cardiovascular Outcomes and Mortality, and Identification of Genetic Variants in Older Individuals: The Cardiovascular Health Study. J Am Heart Assoc 2022; 11:e024374. [PMID: 36314488 PMCID: PMC9673628 DOI: 10.1161/jaha.121.024374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Monocytes/macrophages participate in cardiovascular disease. CD163 (cluster of differentiation 163) is a monocyte/macrophage receptor, and the shed sCD163 (soluble CD163) reflects monocyte/macrophage activation. We examined the association of sCD163 with incident cardiovascular disease events and performed a genome-wide association study to identify sCD163-associated variants. Methods and Results We measured plasma sCD163 in 5214 adults (aged ≥65 years, 58.7% women, 16.2% Black) of the CHS (Cardiovascular Health Study). We used Cox regression models (associations of sCD163 with incident events and mortality); median follow-up was 26 years. Genome-wide association study analyses were stratified on race. Adjusted for age, sex, and race and ethnicity, sCD163 levels were associated with all-cause mortality (hazard ratio [HR], 1.08 [95% CI, 1.04-1.12] per SD increase), cardiovascular disease mortality (HR, 1.15 [95% CI, 1.09-1.21]), incident coronary heart disease (HR, 1.10 [95% CI, 1.04-1.16]), and incident heart failure (HR, 1.18 [95% CI, 1.12-1.25]). When further adjusted (eg, cardiovascular disease risk factors), only incident coronary heart disease lost significance. In European American individuals, genome-wide association studies identified 38 variants on chromosome 2 near MGAT5 (top result rs62165726, P=3.3×10-18),19 variants near chromosome 17 gene ASGR1 (rs55714927, P=1.5×10-14), and 18 variants near chromosome 11 gene ST3GAL4. These regions replicated in the European ancestry ADDITION-PRO cohort, a longitudinal cohort study nested in the Danish arm of the Anglo-Danish-Dutch study of Intensive Treatment Intensive Treatment In peOple with screeNdetcted Diabetes in Primary Care. In Black individuals, we identified 9 variants on chromosome 6 (rs3129781 P=7.1×10-9) in the HLA region, and 3 variants (rs115391969 P=4.3×10-8) near the chromosome 16 gene MYLK3. Conclusions Monocyte function, as measured by sCD163, may be predictive of overall and cardiovascular-specific mortality and incident heart failure.
Collapse
Affiliation(s)
- Peter Durda
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | | | - Ethan M. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of MedicineUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Nels C. Olson
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | - Nancy Swords Jenny
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | - Mary Cushman
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT,Department of MedicineLarner College of Medicine, University of VermontBurlingtonVT
| | - Pia Deichgraeber
- Steno Diabetes CenterAarhus University HospitalAarhusDenmark,Department of Endocrinology and Internal MedicineAarhus University HospitalAarhusDenmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic ResearchCopenhagenDenmark
| | - Anna Jonsson
- Novo Nordisk Foundation Center for Basic Metabolic ResearchCopenhagenDenmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic ResearchCopenhagenDenmark
| | | | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health ServicesUniversity of WashingtonSeattleWA
| | - Alex P. Reiner
- Department of EpidemiologyUniversity of WashingtonSeattleWA
| | - Russell P. Tracy
- Department of Pathology and Laboratory MedicineLarner College of Medicine, University of VermontBurlingtonVT,Department of BiochemistryLarner College of Medicine, University of VermontBurlingtonVT
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of MedicineUniversity of Colorado Anschutz Medical CampusAuroraCO
| |
Collapse
|
6
|
Sorto P, Mäyränpää MI, Saksi J, Nuotio K, Ijäs P, Tuimala J, Vikatmaa P, Soinne L, Kovanen PT, Lindsberg PJ. Glutamine synthetase in human carotid plaque macrophages associates with features of plaque vulnerability: An immunohistological study. Atherosclerosis 2022; 352:18-26. [DOI: 10.1016/j.atherosclerosis.2022.05.008] [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: 05/18/2021] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
|
7
|
Nakagawa K, Tanaka M, Hahm TH, Nguyen HN, Matsui T, Chen YX, Nakashima Y. Accumulation of Plasma-Derived Lipids in the Lipid Core and Necrotic Core of Human Atheroma: Imaging Mass Spectrometry and Histopathological Analyses. Arterioscler Thromb Vasc Biol 2021; 41:e498-e511. [PMID: 34470476 DOI: 10.1161/atvbaha.121.316154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kazunori Nakagawa
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan
| | - Mitsuru Tanaka
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Tae-Hun Hahm
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Huu-Nghi Nguyen
- Department of Science and International Collaboration, Institute for Research and Development of Organic Products, Hanoi, Vietnam (H.-N.N.)
| | - Toshiro Matsui
- Laboratory of Food Analysis, Department of Bioscience and Biotechnology, Faculty of Agriculture (M.T., T.-H.H., T.M.), Kyushu University, Fukuoka, Japan
| | - Yong-Xiang Chen
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan.,Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Alberta, Canada (Y.-X.C.)
| | - Yutaka Nakashima
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences (K.N., Y.-X.C., Y.N.), Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
István L, Czakó C, Élő Á, Mihály Z, Sótonyi P, Varga A, Ungvári Z, Csiszár A, Yabluchanskiy A, Conley S, Csipő T, Lipecz Á, Kovács I, Nagy ZZ. Imaging retinal microvascular manifestations of carotid artery disease in older adults: from diagnosis of ocular complications to understanding microvascular contributions to cognitive impairment. GeroScience 2021; 43:1703-1723. [PMID: 34100219 PMCID: PMC8492863 DOI: 10.1007/s11357-021-00392-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Carotid artery stenosis (CAS) is a consequence of systemic atherosclerotic disease affecting the aging populations of the Western world. CAS is frequently associated with cognitive impairment. However, the mechanisms contributing to the development of vascular cognitive impairment (VCI) associated with CAS are multifaceted and not fully understood. In addition to embolization and decreased blood flow due to the atherosclerotic lesion in the carotid artery, microcirculatory dysfunction in the cerebral circulation also plays a critical role in CAS-related VCI. To better understand the microvascular contributions to cognitive decline associated with CAS and evaluate microvascular protective effects of therapeutic interventions, it is essential to examine the structural and functional changes of the microvessels in the central nervous system (CNS). However, there are some limitations of in vivo brain vascular imaging modalities. The retinal microvasculature provides a unique opportunity to study pathogenesis of cerebral small vessel disease and VCI, because the cerebral circulation and the retinal circulation share similar anatomy, physiology and embryology. Similar microvascular pathologies may manifest in the brain and the retina, thus ocular examination can be used as a noninvasive screening tool to investigate pathological changes in the CNS associated with CAS. In this review, ocular signs of CAS and the retinal manifestations of CAS-associated microvascular dysfunction are discussed. The advantages and limitation of methods that are capable of imaging the ocular circulation (including funduscopy, fluorescein angiography, Doppler sonography, optical coherence tomography [OCT] and optical coherence tomography angiography [OCTA]) are discussed. The potential use of dynamic retinal vessel analysis (DVA), which allows for direct visualization of neurovascular coupling responses in the CNS, for understanding microvascular contributions to cognitive decline in CAS patients is also considered.
Collapse
Affiliation(s)
- Lilla István
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Cecilia Czakó
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Ágnes Élő
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| | - Zsuzsanna Mihály
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Péter Sótonyi
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Andrea Varga
- Department of Vascular & Endovascular Surgery, Semmelweis University, Budapest, Hungary
| | - Zoltán Ungvári
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Anna Csiszár
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamás Csipő
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Ágnes Lipecz
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
- Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Josa Andras Hospital, Nyiregyhaza, Hungary
| | - Illés Kovács
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary.
- Department of Ophthalmology, Weill Cornell Medical College, New York City, NY, USA.
- Department of Clinical Ophtalmology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary.
| | - Zoltán Zsolt Nagy
- Department of Ophthalmology, Semmelweis University, 39 Mária Street, 1085, Budapest, Hungary
| |
Collapse
|
9
|
Search for Reliable Circulating Biomarkers to Predict Carotid Plaque Vulnerability. Int J Mol Sci 2020; 21:ijms21218236. [PMID: 33153204 PMCID: PMC7662861 DOI: 10.3390/ijms21218236] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/30/2020] [Accepted: 11/01/2020] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is responsible for 20% of ischemic strokes, and the plaques from the internal carotid artery the most frequently involved. Lipoproteins play a key role in carotid atherosclerosis since lipid accumulation contributes to plaque progression and chronic inflammation, both factors leading to plaque vulnerability. Carotid revascularization to prevent future vascular events is reasonable in some patients with high-grade carotid stenosis. However, the degree of stenosis alone is not sufficient to decide upon the best clinical management in some situations. In this context, it is essential to further characterize plaque vulnerability, according to specific characteristics (lipid-rich core, fibrous cap thinning, intraplaque hemorrhage). Although these features can be partly detected by imaging techniques, identifying carotid plaque vulnerability is still challenging. Therefore, the study of circulating biomarkers could provide adjunctive criteria to predict the risk of atherothrombotic stroke. In this regard, several molecules have been found altered, but reliable biomarkers have not been clearly established yet. The current review discusses the concept of vulnerable carotid plaque, and collects existing information about putative circulating biomarkers, being particularly focused on lipid-related and inflammatory molecules.
Collapse
|
10
|
CD163+ macrophages are associated with a vulnerable plaque phenotype in human carotid plaques. Sci Rep 2020; 10:14362. [PMID: 32873809 PMCID: PMC7463157 DOI: 10.1038/s41598-020-71110-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 07/27/2020] [Indexed: 01/15/2023] Open
Abstract
Macrophages are a functionally heterogeneous group of immune cells abundant in atherosclerotic plaques. Macrophages expressing CD163 are associated with intraplaque hemorrhage and have previously been considered atheroprotective. However, in a recent study CD163-deficient atherosclerotic ApoE-/- mice exhibited smaller and less complex plaques, suggesting a proatherogenic role of CD163. Previous smaller studies on CD163+ macrophages and plaque stability in humans have yielded diverging results. Here we assessed the association of CD163+ cells to plaque vulnerability in a large cohort of human carotid plaques. CD163 protein expression was analyzed by immunohistochemistry in 200 human carotid plaques removed by endarterectomy from 103 patients with and 93 patients without cerebrovascular symptoms. Furthermore, CD163 mRNA expression was analyzed in 66 of the plaques. Both protein and mRNA expression of CD163 was higher in plaques from symptomatic patients and in plaques with high vulnerability index. CD163+ macrophages were primarily found in shoulder regions and in the center of the plaques. The present data show that CD163 is associated with increased plaque vulnerability in human carotid plaques, supporting the notion that CD163+ macrophages could contribute to clinical events.
Collapse
|
11
|
The Protective Role of Heme Oxygenase-1 in Atherosclerotic Diseases. Int J Mol Sci 2019; 20:ijms20153628. [PMID: 31344980 PMCID: PMC6695885 DOI: 10.3390/ijms20153628] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. These products have anti-inflammatory, anti-oxidant, anti-apoptotic, and anti-thrombotic properties. Although HO-1 is expressed at low levels in most tissues under basal conditions, it is highly inducible in response to various pathophysiological stresses/stimuli. HO-1 induction is thus thought to be an adaptive defense system that functions to protect cells and tissues against injury in many disease settings. In atherosclerosis, HO-1 may play a protective role against the progression of atherosclerosis, mainly due to the degradation of pro-oxidant heme, the generation of anti-oxidants biliverdin and bilirubin and the production of vasodilator CO. In animal models, a lack of HO-1 was shown to accelerate atherosclerosis, whereas HO-1 induction reduced atherosclerosis. It was also reported that HO-1 induction improved the cardiac function and postinfarction survival in animal models of heart failure or myocardial infarction. Recently, we and others examined blood HO-1 levels in patients with atherosclerotic diseases, e.g., coronary artery disease (CAD) and peripheral artery disease (PAD). Taken together, these findings to date support the notion that HO-1 plays a protective role against the progression of atherosclerotic diseases. This review summarizes the roles of HO-1 in atherosclerosis and focuses on the clinical studies that examined the relationships between HO-1 levels and atherosclerotic diseases.
Collapse
|
12
|
Synergistic protective effects of a statin and an angiotensin receptor blocker for initiation and progression of atherosclerosis. PLoS One 2019; 14:e0215604. [PMID: 31050669 PMCID: PMC6499436 DOI: 10.1371/journal.pone.0215604] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022] Open
Abstract
Aim Although the atheroprotective effects of statins and angiotensin II receptor blockers (ARBs) are well-established, little is known about their additive effects, especially during the early period of atherosclerosis. The aim of this study was to investigate whether combination of a statin and an ARB exerts synergistic anti-atherosclerotic effects, and to elucidate the mechanisms of combined effects. Methods Atherosclerotic plaques were developed in arteries of 23 rabbits using a high-cholesterol diet (HCD) and intra-arterial balloon inflation. Rabbits received one of five different treatment strategies for 4 weeks: positive control [n = 5, HCD]; negative control [n = 3, regular chow diet]; statin [n = 5, HCD and rosuvastatin 10 mg]; ARB [n = 5, HCD and olmesartan 20 mg]; and combination [n = 5, HCD and statin+ARB]. Results Histological analysis demonstrated that development of atherosclerotic plaques was inhibited more in combination group than in statin group (P = 0.001). Although macrophage infiltration identified by RAM11 staining was not significantly different between combination and individual treatment groups (31.76±4.84% [combination] vs. 38.11±6.53% [statin; P = 0.35] or 35.14±2.87% [ARB; P = 0.62]), the relative proportion of pro-inflammatory M1-macrophages was significantly lower in combination group than in ARB group (3.20±0.47% vs. 5.20±0.78%, P = 0.02). Furthermore, M2-macrophage polarization was higher in combination group than in statin group (17.70±3.04% vs. 7.86±0.68%, P = 0.001). Conclusion Combination treatment with a statin and an ARB produced synergistic protective effects for atherosclerosis initiation and progression, which may be attributed to modulation of macrophage characteristics in the early period of atherosclerosis.
Collapse
|
13
|
Nuotio K, Ijäs P, Heikkilä HM, Koskinen SM, Saksi J, Vikatmaa P, Sorto P, Mäkitie L, Eriksson H, Kasari S, Silvennoinen H, Valanne L, Mäyränpää MI, Kovanen PT, Soinne L, Lindsberg PJ. Morphology and histology of silent and symptom-causing atherosclerotic carotid plaques - Rationale and design of the Helsinki Carotid Endarterectomy Study 2 (the HeCES2). Ann Med 2018; 50:501-510. [PMID: 30010425 DOI: 10.1080/07853890.2018.1494851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
INTRODUCTION Every fifth ischemic stroke is caused by thromboembolism originating from an atherosclerotic carotid artery plaque. While prevention is the most cost-effective stroke therapy, antiplatelet and cholesterol-lowering drugs have a ceiling effect in their efficacy. Therefore, discovery of novel pathophysiologic targets are needed to improve the primary and secondary prevention of stroke. This article provides a detailed study design and protocol of HeCES2, an observational prospective cohort study with the objective to investigate the pathophysiology of carotid atherosclerosis. MATERIALS AND METHODS Recruitment and carotid endarterectomies of the study patients with carotid atherosclerosis were performed from October 2012 to September 2015. After brain and carotid artery imaging, endarterectomised carotid plaques (CPs) and blood samples were collected from 500 patients for detailed biochemical and molecular analyses. Findings to date: We developed a morphological grading for macroscopic characteristics within CPs. The dominant macroscopic CP characteristics were: smoothness 62%, ulceration 61%, intraplaque hemorrhage 60%, atheromatous gruel 59%, luminal coral-type calcification 34%, abundant (44%) and moderate (39%) intramural calcification, and symptom-causing "hot spot" area 53%. Future plans: By combining clinically oriented and basic biomedical research, this large-scale study attempts to untangle the pathophysiological perplexities of human carotid atherosclerosis. Key Messages This article is a rationale and design of the HeCES2 study that is an observational prospective cohort study with the objective to investigate the pathophysiology of carotid atherosclerosis. The HeCES2 study strives to develop diagnostic algorithms including radiologic imaging to identify carotid atherosclerosis patients who warrant surgical treatment. In addition, the study aims at finding out new tools for clinical risk stratification as well as novel molecular targets for drug development.
Collapse
Affiliation(s)
- Krista Nuotio
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland.,b Department of Neurology , Helsinki University Hospital and Clinical Neurosciences, University of Helsinki , Helsinki , Finland
| | - Petra Ijäs
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland.,b Department of Neurology , Helsinki University Hospital and Clinical Neurosciences, University of Helsinki , Helsinki , Finland
| | - Hanna M Heikkilä
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland
| | - Suvi M Koskinen
- c Clinicum, Department of Neurosciences , University of Helsinki , Helsinki , Finland.,d Department of Radiology, Hospital District of Helsinki and Uusimaa Medical Imaging Center , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Jani Saksi
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland
| | - Pirkka Vikatmaa
- e Abdominal Center, Vascular Surgery, Helsinki University Hospital , Helsinki , Finland
| | - Pia Sorto
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland
| | - Laura Mäkitie
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland.,b Department of Neurology , Helsinki University Hospital and Clinical Neurosciences, University of Helsinki , Helsinki , Finland
| | - Henrietta Eriksson
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland.,b Department of Neurology , Helsinki University Hospital and Clinical Neurosciences, University of Helsinki , Helsinki , Finland
| | - Sonja Kasari
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland
| | - Heli Silvennoinen
- d Department of Radiology, Hospital District of Helsinki and Uusimaa Medical Imaging Center , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Leena Valanne
- d Department of Radiology, Hospital District of Helsinki and Uusimaa Medical Imaging Center , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Mikko I Mäyränpää
- f Department of Pathology , University of Helsinki and HUSLAB, Helsinki University Hospital , Helsinki , Finland
| | | | - Lauri Soinne
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland.,b Department of Neurology , Helsinki University Hospital and Clinical Neurosciences, University of Helsinki , Helsinki , Finland
| | - Perttu J Lindsberg
- a Molecular Neurology, Research Programs Unit, Biomedicum Helsinki , University of Helsinki , Helsinki , Finland.,b Department of Neurology , Helsinki University Hospital and Clinical Neurosciences, University of Helsinki , Helsinki , Finland
| |
Collapse
|
14
|
Lin P, Chang CC, Yuan KC, Yeh HJ, Fang SU, Cheng T, Teng KT, Chao KC, Tang JH, Kao WY, Lin PY, Liu JS, Chang JS. Red Blood Cell Aggregation-Associated Dietary Pattern Predicts Hyperlipidemia and Metabolic Syndrome. Nutrients 2018; 10:E1127. [PMID: 30127325 PMCID: PMC6115951 DOI: 10.3390/nu10081127] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/05/2018] [Accepted: 08/15/2018] [Indexed: 12/26/2022] Open
Abstract
Red blood cell (RBC) aggregation and iron status are interrelated and strongly influenced by dietary factors, and their alterations pose a great risk of dyslipidemia and metabolic syndrome (MetS). Currently, RBC aggregation-related dietary patterns remain unclear. This study investigated the dietary patterns that were associated with RBC aggregation and their predictive effects on hyperlipidemia and MetS. Anthropometric and blood biochemical data and food frequency questionnaires were collected from 212 adults. Dietary patterns were derived using reduced rank regression from 32 food groups. Adjusted linear regression showed that hepcidin, soluble CD163, and serum transferrin saturation (%TS) independently predicted RBC aggregation (all p < 0.01). Age-, sex-, and log-transformed body mass index (BMI)-adjusted prevalence rate ratio (PRR) showed a significant positive correlation between RBC aggregation and hyperlipidemia (p-trend < 0.05). RBC aggregation and iron-related dietary pattern scores (high consumption of noodles and deep-fried foods and low intake of steamed, boiled, and raw food, dairy products, orange, red, and purple vegetables, white and light-green vegetables, seafood, and rice) were also significantly associated with hyperlipidemia (p-trend < 0.05) and MetS (p-trend = 0.01) after adjusting for age, sex, and log-transformed BMI. Our results may help dieticians develop dietary strategies for preventing dyslipidemia and MetS.
Collapse
Affiliation(s)
- Pei Lin
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan.
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Kuo-Ching Yuan
- Department of Emergency and Critical Care Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Hsing-Jung Yeh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Sheng-Uei Fang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Tiong Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Kai-Tse Teng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Kuo-Ching Chao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Jui-Hsiang Tang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Wei-Yu Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Pao-Ying Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Ju-Shian Liu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Jung-Su Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan.
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Chinese Taipei Society for the Study of Obesity, CTSSO, Taipei 11031, Taiwan.
| |
Collapse
|
15
|
Upregulation of arylsulfatase B in carotid atherosclerosis is associated with symptoms of cerebral embolization. Sci Rep 2017; 7:4338. [PMID: 28659610 PMCID: PMC5489491 DOI: 10.1038/s41598-017-04497-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/16/2017] [Indexed: 12/30/2022] Open
Abstract
The aim of this study was to identify genes for which the expression within carotid atherosclerosis was reproducibly associated with the symptoms of cerebral embolization. Two publically available microarray datasets E-MEXP-2257 and GSE21545 were analysed using GeneSpring 11.5. The two datasets utilized a total of 22 and 126 carotid atherosclerosis samples, obtained from patients with and without symptoms of cerebral embolization, respectively. To assess whether the findings were reproducible we analysed carotid atherosclerosis samples from another 8 patients with and 7 patients without symptoms of cerebral embolization using real-time PCR. In vitro studies using VSMC were performed to assess the functional relevance of one of the validated genes. We identified 1624 and 135 differentially expressed genes within carotid atherosclerosis samples of symptomatic compared to asymptomatic patients using the E-MEXP-2257 and GSE21545 datasets, respectively (≥1.15-absolute fold-change, P < 0.05). Only 7 differentially expressed genes or 0.4% (7/1,752) were consistent between the datasets. We validated the differential expression of ARSB which was upregulated 1.15-fold (P = 0.029) in atherosclerosis from symptomatic patients. In vitro incubation of VSMCs with the ARSB inhibitor L-ascorbic acid resulted in marked upregulation of SIRT1 and AMPK. This study suggests that ARSB may represent a novel target to limit carotid embolization.
Collapse
|
16
|
Dong J, Song C, Zhang L, Feng X, Feng R, Lu Q, Zhao Z, Bao J, Zhou J, Jing Z. Identified key genes related to carotid atheroma plaque from gene expression chip. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:1-6. [PMID: 27684644 DOI: 10.1080/21691401.2016.1216858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
As one of the leading reason in morbidity and death in the world, atherosclerosis is usually associated with vessel stenosis, ulceration, and inflammatory cell infiltration. However, the formation mechanism of atheroma plaque is unknown. In this research, we have used bioinformatics tools to identify 118 differential expression genes from a GEO dataset. Besides, we also revealed KYNU as a crucial gene in atheroma plaque development.
Collapse
Affiliation(s)
- Jian Dong
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Chao Song
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Lei Zhang
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Xiang Feng
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Rui Feng
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Qingsheng Lu
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Zhiqing Zhao
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China.,b Department of Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Junmin Bao
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Jian Zhou
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| | - Zaiping Jing
- a Department of Vascular Surgery , Changhai Hospital, Second Military Medical University , Shanghai , China
| |
Collapse
|
17
|
Perisic L, Aldi S, Sun Y, Folkersen L, Razuvaev A, Roy J, Lengquist M, Åkesson S, Wheelock CE, Maegdefessel L, Gabrielsen A, Odeberg J, Hansson GK, Paulsson-Berne G, Hedin U. Gene expression signatures, pathways and networks in carotid atherosclerosis. J Intern Med 2016; 279:293-308. [PMID: 26620734 DOI: 10.1111/joim.12448] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Embolism from unstable atheromas in the carotid bifurcation is a major cause of stroke. Here, we analysed gene expression in endarterectomies from patients with symptomatic (S) and asymptomatic (AS) carotid stenosis to identify pathways linked to plaque instability. METHODS Microarrays were prepared from plaques (n = 127) and peripheral blood samples (n = 96) of S and AS patients. Gene set enrichment, pathway mapping and network analyses of differentially expressed genes were performed. RESULTS These studies revealed upregulation of haemoglobin metabolism (P = 2.20E-05) and bone resorption (P = 9.63E-04) in S patients. Analysis of subgroups of patients indicated enrichment of calcification and osteoblast differentiation in S patients on statins, as well as inflammation and apoptosis in plaques removed >1 month compared to <2 weeks after symptom. By prediction profiling, a panel of 30 genes, mostly transcription factors, discriminated between plaques from S versus AS patients with 78% accuracy. By meta-analysis, common gene networks associated with atherosclerosis mapped to hypoxia, chemokines, calcification, actin cytoskeleton and extracellular matrix. A set of dysregulated genes (LMOD1, SYNPO2, PLIN2 and PPBP) previously not described in atherosclerosis were identified from microarrays and validated by quantitative PCR and immunohistochemistry. CONCLUSIONS Our findings confirmed a central role for inflammation and proteases in plaque instability, and highlighted haemoglobin metabolism and bone resorption as important pathways. Subgroup analysis suggested prolonged inflammation following the symptoms of plaque instability and calcification as a possible stabilizing mechanism by statins. In addition, transcriptional regulation may play an important role in the determination of plaque phenotype. The results from this study will serve as a basis for further exploration of molecular signatures in carotid atherosclerosis.
Collapse
Affiliation(s)
- L Perisic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - S Aldi
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Y Sun
- Translational Science Center, Personalized Healthcare and Biomarkers, R&D, Astra Zeneca, Stockholm, Sweden
| | - L Folkersen
- Department of Molecular Genetics, Novo Nordisk, Copenhagen, Denmark.,Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - A Razuvaev
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - J Roy
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - M Lengquist
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - S Åkesson
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - C E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - L Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - A Gabrielsen
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - J Odeberg
- Department of Medicine, Karolinska Institute, Stockholm, Sweden.,Science for Life Laboratory, Department of Proteomics, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - G K Hansson
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - U Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
18
|
Swaminathan B, Goikuria H, Vega R, Rodríguez-Antigüedad A, López Medina A, Freijo MDM, Vandenbroeck K, Alloza I. Autophagic marker MAP1LC3B expression levels are associated with carotid atherosclerosis symptomatology. PLoS One 2014; 9:e115176. [PMID: 25503069 PMCID: PMC4264866 DOI: 10.1371/journal.pone.0115176] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 11/19/2014] [Indexed: 12/31/2022] Open
Abstract
Objectives The mechanism by which atheroma plaque becomes unstable is not completely understood to date but analysis of differentially expressed genes in stable versus unstable plaques may provide clues. This will be crucial toward disclosing the mechanistic basis of plaque instability, and may help to identify prognostic biomarkers for ischaemic events. The objective of our study was to identify differences in expression levels of 59 selected genes between symptomatic patients (unstable plaques) and asymptomatic patients (stable plaques). Methods 80 carotid plaques obtained by carotid endarterectomy and classified as symptomatic (>70% stenosis) or asymptomatic (>80% stenosis) were used in this study. The expression levels of 59 genes were quantified by qPCR on RNA extracted from the carotid plaques obtained by endarterectomy and analyzed by means of various bioinformatic tools. Results Several genes associated with autophagy pathways displayed differential expression levels between asymptomatic and symptomatic (i.e. MAP1LC3B, RAB24, EVA1A). In particular, mRNA levels of MAP1LC3B, an autophagic marker, showed a 5−fold decrease in symptomatic samples, which was confirmed in protein blots. Immune system−related factors and endoplasmic reticulum-associated markers (i.e. ERP27, ITPR1, ERO1LB, TIMP1, IL12B) emerged as differently expressed genes between asymptomatic and symptomatic patients. Conclusions Carotid atherosclerotic plaques in which MAP1LC3B is underexpressed would not be able to benefit from MAP1LC3B−associated autophagy. This may lead to accumulation of dead cells at lesion site with subsequent plaque destabilization leading to cerebrovascular events. Identified biomarkers and network interactions may represent novel targets for development of treatments against plaque destabilization and thus for the prevention of cerebrovascular events.
Collapse
Affiliation(s)
- Bhairavi Swaminathan
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
| | - Haize Goikuria
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
- Achucarro Basque Center for Neurosciences, Zamudio, Spain
| | - Reyes Vega
- Department of Neurology, Basurto Hospital, Bilbao, Spain
| | | | | | | | - Koen Vandenbroeck
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
- IKERBASQUE, Basque Foundation for Sciences, Bilbao, Spain
- Achucarro Basque Center for Neurosciences, Zamudio, Spain
| | - Iraide Alloza
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
- IKERBASQUE, Basque Foundation for Sciences, Bilbao, Spain
- Achucarro Basque Center for Neurosciences, Zamudio, Spain
- * E-mail:
| |
Collapse
|
19
|
Valanti E, Tsompanidis A, Sanoudou D. Pharmacogenomics in the development and characterization of atheroprotective drugs. Methods Mol Biol 2014; 1175:259-300. [PMID: 25150873 DOI: 10.1007/978-1-4939-0956-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Atherosclerosis is the main cause of cardiovascular disease (CVD) and can lead to stroke, myocardial infarction, and death. The clinically available atheroprotective drugs aim mainly at reducing the levels of circulating low-density lipoprotein (LDL), increasing high-density lipoprotein (HDL), and attenuating inflammation. However, the cardiovascular risk remains high, along with morbidity, mortality, and incidence of adverse drug events. Pharmacogenomics is increasingly contributing towards the characterization of existing atheroprotective drugs, the evaluation of novel ones, and the identification of promising, unexplored therapeutic targets, at the global molecular pathway level. This chapter presents highlights of pharmacogenomics investigations and discoveries that have contributed towards the elucidation of pharmacological atheroprotection, while opening the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Efi Valanti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens, 115 27, Greece
| | | | | |
Collapse
|
20
|
Ayari H, Bricca G. Identification of two genes potentially associated in iron-heme homeostasis in human carotid plaque using microarray analysis. J Biosci 2013; 38:311-5. [PMID: 23660665 DOI: 10.1007/s12038-013-9310-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Classic characteristics are poor predictors of the risk of thromboembolism. Thus, better markers for the carotid atheroma plaque formation and symptom causing are needed. Our objective was to study by microarray analysis gene expression of genes involved in homeostasis of iron and heme in carotid atheroma plaque from the same patient. mRNA gene expression was measured by an Affymetrix GeneChip Human Gene 1.0 ST arrays (Affymetrix, Santa Clara, CA, USA) using RNA prepared from 68 specimens of endarteriectomy from 34 patients. Two genes involved in iron-heme homeostasis, CD163 and heme oxygenase (HO-1), were analysed in 34 plaques. CD163 (2.18, p01.45E-08) and HO-1 (fold-change 2.67, p02.07E-09) mRNAs were induced. We suggest that atheroma plaques show a more pronounced induction of CD163 and HO-1. Although further evidence is needed, our results support previous data. To our knowledge, this is the first report comparing gene expression between intact arterial tissue and carotid plaque using microarray analysis.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Carotid Artery Diseases/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Female
- Heme/metabolism
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Homeostasis
- Humans
- Hypertension/metabolism
- Iron/metabolism
- Male
- Oligonucleotide Array Sequence Analysis
- Plaque, Atherosclerotic/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Transcriptional Activation
- Transcriptome
Collapse
Affiliation(s)
- Hanène Ayari
- Universite Lyon1, INSERM, ERI22, EA 4173, Lyon, France.
| | | |
Collapse
|
21
|
Perisic L, Hedin E, Razuvaev A, Lengquist M, Osterholm C, Folkersen L, Gillgren P, Paulsson-Berne G, Ponten F, Odeberg J, Hedin U. Profiling of atherosclerotic lesions by gene and tissue microarrays reveals PCSK6 as a novel protease in unstable carotid atherosclerosis. Arterioscler Thromb Vasc Biol 2013; 33:2432-43. [PMID: 23908247 DOI: 10.1161/atvbaha.113.301743] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Carotid plaque instability is a major cause of ischemic stroke, but detailed knowledge about underlying molecular pathways is still lacking. Here, we evaluated large-scale transcriptomic and protein expression profiling in a biobank of carotid endarterectomies followed by characterization of identified candidates, as a platform for discovery of novel proteins differentially regulated in unstable carotid lesions. APPROACH AND RESULTS Genes highly upregulated in symptomatic versus asymptomatic plaques were selected from Affymetrix microarray analyses (n=127 plaques), and tissue microarrays constructed from 34 lesions were assayed for 21 corresponding proteins by immunohistochemistry. Quantification of stainings demonstrated differential expression of CD36, CD137, and DOCK7 (P<0.05) in unstable versus stable lesions and the most significant upregulation of a proprotein convertase, PCSK6 (P<0.0001). Increased expression of PCSK6 in symptomatic lesions was verified by quantitative real-time polymerase chain reaction (n=233), and the protein was localized to smooth muscle α-actin positive cells and extracellular matrix of the fibrous cap by immunohistochemistry. PCSK6 expression positively correlated to genes associated with inflammation, matrix degradation, and mitogens in microarrays. Stimulation of human carotid smooth muscle cells in vitro with cytokines caused rapid induction of PCSK6 mRNA. CONCLUSIONS Using a combination of transcriptomic and tissue microarray profiling, we demonstrate a novel approach to identify proteins differentially expressed in unstable carotid atherosclerosis. The proprotein convertase PCSK6 was detected at increased levels in the fibrous cap of symptomatic carotid plaques, possibly associated with key processes in plaque rupture such as inflammation and extracellular matrix remodeling. Further studies are needed to clarify the role of PCSK6 in atherosclerosis.
Collapse
Affiliation(s)
- Ljubica Perisic
- From the Department of Molecular Medicine and Surgery (L.P., E.H., A.R., M.L., C.O., U.H.), and Department of Medicine (G.P.-B., J.O.), Karolinska Institute, Stockholm, Sweden; Department of Molecular Genetics, Novo Nordisk, Copenhagen, Denmark (L.F.); Department of Surgery, Södersjukhuset, Stockholm, Sweden (P.G.); Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden (F.P.); and Department of Proteomics, Royal Institute of Technology, Stockholm, Sweden (J.O.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ijäs P, Saksi J, Soinne L, Tuimala J, Jauhiainen M, Jula A, Kähönen M, Kesäniemi YA, Kovanen PT, Kaste M, Lindsberg PJ. Haptoglobin 2 allele associates with unstable carotid plaque and major cardiovascular events. Atherosclerosis 2013; 230:228-34. [PMID: 24075749 DOI: 10.1016/j.atherosclerosis.2013.07.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 07/15/2013] [Accepted: 07/15/2013] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Intraplaque hemorrhages (IPH) may predispose to unstable atherosclerotic disease and its atherothrombotic complications, ischemic stroke and coronary syndromes. However, the discriminative value of IPH has been limited in histological and imaging studies suggesting that confounding factors modulate the response to IPH. We studied whether common variants of haptoglobin (Hp), which facilitates the removal of free hemoglobin and protects tissues from heme-iron induced oxidative damage, would modify the inflammatory response to IPH and the risk of unstable carotid stenosis (CS) and major cardiovascular diseases. METHODS We genotyped Hp polymorphism in 91 patients with a high-grade CS from Helsinki Carotid Endarterectomy Study (HeCES) and in 1417 individuals from Health 2000, a Finnish epidemiological cross-sectional health survey, and determined heme oxygenase-1 (HO1) expression in relation to Hp genotypes in carotid plaques. RESULTS In the Health 2000 cohort, Hp genotype frequencies were 0.143 (hp1-1), 0.486 (hp1-2) and 0.371 (hp2-2) consistent with Hardy-Weinberg equilibrium and those reported from other Caucasian populations. Among patients with unstable CS, the frequency of hp2-2 genotype was higher than in the control population (0.516 vs. 0.371, P = 0.025). Hp genotypes correlated with HO1 expression in the plaque (r = 0.47, P = 0.027). In the Health 2000 cohort, hp2 allele was associated with an increased risk of major cardiovascular diseases (ischemic stroke, TIA, myocardial infarction, coronary heart disease) with an adjusted OR of 1.46 (95% CI 1.03-2.06). CONCLUSION Common haptoglobin variants modulate the inflammatory response to IPH and associate with the risk of unstable carotid stenosis and major ischemic cardiovascular events.
Collapse
Affiliation(s)
- Petra Ijäs
- Department of Neurology, Helsinki University Central Hospital, Finland; Research Programs Unit, Molecular Neurology, Biomedicum-Helsinki and Department of Clinical Neurosciences, University of Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yunoki K, Inoue T, Sugioka K, Nakagawa M, Inaba M, Wada S, Ohsawa M, Komatsu R, Itoh A, Haze K, Yoshiyama M, Becker AE, Ueda M, Naruko T. Association between hemoglobin scavenger receptor and heme oxygenase-1-related anti-inflammatory mediators in human coronary stable and unstable plaques. Hum Pathol 2013; 44:2256-65. [PMID: 23850497 DOI: 10.1016/j.humpath.2013.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 04/05/2013] [Accepted: 05/08/2013] [Indexed: 10/26/2022]
Abstract
Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that is induced by intraplaque hemorrhage and degrades free heme and releases ferrous iron, which is rapidly sequestered by ferritin. In vitro studies have shown that binding of hemoglobin to hemoglobin scavenger receptor (CD163) induces HO-1 and the anti-inflammatory mediator interleukin (IL)-10. We immunohistochemically examined the relationship between CD163 expression in macrophages and intraplaque hemorrhage, HO-1, IL-10, and ferritin using coronary atherectomy specimens from patients with stable (SAP) or unstable angina pectoris (UAP). A total of 67 patients underwent atherectomy for SAP (n = 33) or UAP (n = 34). Samples were stained with antibodies against smooth muscle cells, macrophages, glycophorin-A (a protein specific to erythrocyte membranes), CD163, HO-1, IL-10, and ferritin. To identify cell types of HO-1-positive cells, double immunostaining was also performed. Double immunostaining for HO-1 and macrophages revealed that the vast majority of HO-1-positive cells were macrophages. Morphometric analysis demonstrated that CD163-positive macrophage score and the percentage of glycophorin-A-, HO-1-, IL-10-, and ferritin-positive areas were significantly higher in UAP than in SAP patients (CD163, P < .005; glycophorin-A, P < .0001; HO-1, P < .0001; IL-10, P < .005; ferritin, P = .0001). Moreover, CD163-positive macrophage score was positively associated with the percentage of glycophorin-A-, HO-1-, IL-10-, and ferritin-positive areas (glycophorin-A, r = 0.60, P < .0001; HO-1, r = 0.67, P < .0001; IL-10, r = 0.45, P < .0005; ferritin, r = 0.61, P < .0001). These findings suggest that enhanced expression of HO-1 and HO-1-related atheroprotective molecules plays an important role in exerting anti-inflammatory, antioxidant, and scavenging functions, which could contribute to plaque stabilization.
Collapse
Affiliation(s)
- Kei Yunoki
- Department of Cardiology, Osaka City General Hospital, Osaka 534-0021, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Histologically unstable asymptomatic carotid plaques have altered expression of genes involved in chemokine signalling leading to localised plaque inflammation and rupture. Eur J Vasc Endovasc Surg 2012; 45:121-7. [PMID: 23265682 DOI: 10.1016/j.ejvs.2012.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 11/07/2012] [Indexed: 11/23/2022]
Abstract
BACKGROUND Many studies have evaluated histological and gene expression profiles in TIA/stroke patients after onset of symptoms, but there is limited understanding as to how these plaque related features interact before symptom onset. In particular, no studies have evaluated differential gene expression in histologically unstable (vs stable plaques) in neurologically asymptomatic patients. METHODS Nine asymptomatic patients had their plaques scored blindly by two independent Histopathologists using the AHA plaque scoring system. RNA extracted from the plaques was hybridised onto a whole genome microarray. Analysis was performed using GenomeStudio (v1.0) and the DAVID bioinformatics resource (v6.7). RESULTS Three plaques were histologically unstable (Grade 2/3), while six were stable (Grade 0/1). 346 differentially expressed genes (>1.3 fold, P < 0.05) were identified (293 down-regulated and 53 up-regulated) between stable and unstable plaques. Genes related to chemokine and protein signalling (pro-inflammatory/pro-apoptotic) were identified to have high enrichment scores (>1.3) and were significantly up-regulated in unstable (asymptomatic) plaques. CONCLUSION The findings confirm the intuitively held belief that changes in chemokine and protein signalling may be associated with acute plaque disruption and precede the onset of symptoms. Once validated, these genes could therefore become targets for innovative medical treatments in the future or could help identify asymptomatic patients with histologically unstable plaques that would benefit from surgical intervention.
Collapse
|
25
|
Lee K, Santibanez-Koref M, Polvikoski T, Birchall D, Mendelow AD, Keavney B. Increased expression of fatty acid binding protein 4 and leptin in resident macrophages characterises atherosclerotic plaque rupture. Atherosclerosis 2012; 226:74-81. [PMID: 23122912 PMCID: PMC3566542 DOI: 10.1016/j.atherosclerosis.2012.09.037] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 09/23/2012] [Accepted: 09/25/2012] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Resident macrophages play an important role in atheromatous plaque rupture. The macrophage gene expression signature associated with plaque rupture is incompletely defined due to the complex cellular heterogeneity in the plaque. We aimed to characterise differential gene expression in resident plaque macrophages from ruptured and stable human atheromatous lesions. METHODS AND RESULTS We performed genome-wide expression analyses of isolated macrophage-rich regions of stable and ruptured human atherosclerotic plaques. Plaques present in carotid endarterectomy specimens were designated as stable or ruptured using clinical, radiological and histopathological criteria. Macrophage-rich regions were excised from 5 ruptured and 6 stable plaques by laser micro-dissection. Transcriptional profiling was performed using Affymetrix microarrays. The profiles were characteristic of activated macrophages. At a false discovery rate of 10%, 914 genes were differentially expressed between stable and ruptured plaques. The findings were confirmed in fourteen further stable and ruptured samples for a subset of eleven genes with the highest expression differences (p < 0.05). Pathway analysis revealed that components of the PPAR/Adipocytokine signaling pathway were the most significantly upregulated in ruptured compared to stable plaques (p = 5.4 × 10(-7)). Two key components of the pathway, fatty-acid binding-protein 4 (FABP4) and leptin, showed nine-fold (p = 0.0086) and five-fold (p = 0.0012) greater expression respectively in macrophages from ruptured plaques. CONCLUSIONS We found differences in gene expression signatures between macrophages isolated from stable and ruptured human atheromatous plaques. Our findings indicate the involvement of FABP4 and leptin in the progression of atherosclerosis and plaque rupture, and suggest that down-regulation of PPAR/adipocytokine signaling within plaques may have therapeutic potential.
Collapse
Affiliation(s)
- K Lee
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, NE1 3BZ Newcastle-upon-Tyne, UK.
| | | | | | | | | | | |
Collapse
|
26
|
Folkersen L, Persson J, Ekstrand J, Agardh HE, Hansson GK, Gabrielsen A, Hedin U, Paulsson-Berne G. Prediction of ischemic events on the basis of transcriptomic and genomic profiling in patients undergoing carotid endarterectomy. Mol Med 2012; 18:669-75. [PMID: 22371308 DOI: 10.2119/molmed.2011.00479] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 02/23/2012] [Indexed: 12/16/2022] Open
Abstract
Classic risk factors, including age, smoking, serum cholesterol, diabetes and blood pressure, constitute the basis of present risk prediction models but fail to identify all individuals at risk. The objective of this study was to investigate if genomic and transcriptional patterns improve prediction of ischemic events in patients with established carotid artery disease. Genotype and gene expression profiles were obtained from carotid plaque tissue (n = 126) and peripheral blood mononuclear cells (n = 97) of patients undergoing carotid endarterectomy. Patients were followed for an average of 44 months, and 25 ischemic events occurred (18 ischemic strokes and 7 myocardial infarctions). Blinded leave-one-out cross-validation on Cox regression coefficients was used to assign gene expression-based risk scores to each patient. When compared with classic risk factors, addition of carotid plaque gene expression-based risk score improved the prediction of future ischemic events from an area under the curve (AUC) of 0.66 to an AUC of 0.79. The inclusion of gene expression risk score from peripheral blood mononuclear cells or from 25 established myocardial infarction risk single nucleotide polymorphisms only exhibited marginal effects on the prediction of ischemic events. Prediction of ischemic events is improved by inclusion of gene expression profiling from carotid endarterectomy tissue compared with prediction on the basis of classic risk markers alone in patients with atherosclerosis. The method may be developed to identify subjects at very high risk of ischemic events.
Collapse
Affiliation(s)
- Lasse Folkersen
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Møller HJ. Soluble CD163. Scandinavian Journal of Clinical and Laboratory Investigation 2011; 72:1-13. [DOI: 10.3109/00365513.2011.626868] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
28
|
Hermus L, Schuitemaker JHN, Tio RA, Breek JC, Slart RHJA, de Boef E, Zeebregts CJ. Novel serum biomarkers in carotid artery stenosis: useful to identify the vulnerable plaque? Clin Biochem 2011; 44:1292-8. [PMID: 21939648 DOI: 10.1016/j.clinbiochem.2011.08.1141] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 08/22/2011] [Accepted: 08/24/2011] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Serum biomarkers representing inflammatory activity in vulnerable carotid plaques may be used to identify high-risk patients for cerebral ischemic events. We aimed to analyze the relationship between concentrations of four novel biomarkers and neurological symptoms: Neopterin, PTX3, sCD163, and sTREM-1. In addition, we analyzed the relationship between these markers and the presence of coronary (CAD) and peripheral (PAD) artery disease. DESIGN AND METHODS Serum biomarker levels were determined in 100 patients undergoing carotid endarterectomy; 33 for stroke, 32 for transient ischemic attack, and 23 for amaurosis fugax. 12 Patients were asymptomatic. Risk factors for atherosclerotic disease and history of CAD and PAD were also assessed. RESULTS Symptomatic patients did not show significantly elevated biomarker levels compared to asymptomatic patients and levels did not differ among symptomatic subgroups. Neopterin levels were elevated in patients with concomitant coronary and peripheral artery disease (CAD (32%) 10.2 ± 6.6 vs no CAD (68%) 7.6 ± 2.9 nmol/L, PAD (20%) 12.3 ± 7.4 vs no PAD (80%) 7.5 ± 3.0 nmol/L, p<0.05). sTREM-1 was elevated in patients with CAD (50.8 ± 53.2 vs 28.0 ± 31.6 ng/L, p<0.05). PTX3 and sCD163 were not significantly elevated in CAD nor PAD. CONCLUSION Our findings suggest that serum neopterin and sTREM-1 levels may be related to the presence of atherosclerotic disease, but not to carotid plaque vulnerability.
Collapse
Affiliation(s)
- Linda Hermus
- Department of Surgery (Division of Vascular Surgery), University Medical Center Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
29
|
Razuvaev A, Ekstrand J, Folkersen L, Agardh H, Markus D, Swedenborg J, Hansson GK, Gabrielsen A, Paulsson-Berne G, Roy J, Hedin U. Correlations between clinical variables and gene-expression profiles in carotid plaque instability. Eur J Vasc Endovasc Surg 2011; 42:722-30. [PMID: 21741279 DOI: 10.1016/j.ejvs.2011.05.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 05/30/2011] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Strokes, a major cause of disability, are often caused by embolism from unstable carotid plaques. The aim of this study was to validate a biobank of human carotid endarterectomies as a platform for further exploration of pathways for plaque instability. For this purpose, we investigated the relationship between clinical parameters of plaque instability and expression of genes previously shown to be associated with either plaque instability or healing processes in the vessel wall. METHODS A database of clinical information and gene-expression microarray data from 106 carotid endarterectomies were used. RESULTS Expression of matrix metalloproteinase (MMP)-9 and MMP-7 was 100-fold higher in plaques than in normal artery. In general, genes associated with inflammation (such as RANKL and CD68) were overexpressed in symptomatic compared with asymptomatic plaques. Plaques obtained from patients undergoing surgery within 2 weeks after an embolic event showed up-regulation of genes involved in healing reactions in the vessel wall (including elastin and collagen). Statin treatment, as well as echodense lesions, were associated with a more stable phenotype. CONCLUSION Here, we demonstrate that gene-expression profiles reflect clinical parameters. Our results suggest that microarray technology and clinical variables can be used for the future identification of central molecular pathways in plaque instability.
Collapse
Affiliation(s)
- A Razuvaev
- Center for Molecular Medicine and Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska University Hospital Solna and Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Saksi J, Ijäs P, Nuotio K, Sonninen R, Soinne L, Salonen O, Saimanen E, Tuimala J, Lehtonen-Smeds EM, Kaste M, Kovanen PT, Lindsberg PJ. Gene expression differences between stroke-associated and asymptomatic carotid plaques. J Mol Med (Berl) 2011; 89:1015-26. [PMID: 21607540 PMCID: PMC3170468 DOI: 10.1007/s00109-011-0773-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 05/06/2011] [Accepted: 05/11/2011] [Indexed: 02/02/2023]
Abstract
Atherosclerotic carotid stenosis is an important risk factor for stroke. Carotid plaques (CPs) causing stroke may present a distinct type of molecular pathology compared with transient ischemic attack (TIA)-associated or asymptomatic plaques. We compared the gene expression profiles of CPs from stroke patients (n = 12) and asymptomatic patients (n = 9), both with similar risk factors and severity of carotid stenosis (>70%). Sixty probes showed over 1.5-fold expression difference at 5% false discovery rate. Functional clustering showed enrichment of genes in 51 GO categories and seven pathways, the most significant of which relate to extracellular-matrix interaction, PPAR gamma signaling, scavanger receptor activity, and lysosomal activity. Differential expression of ten genes was confirmed in an extended replication group (n = 43), where the most significant expression differences were found in CD36 (2.1-fold change, p = 0.005), CD163 (1.7-fold change, p = 0.007) and FABP4 (2.2-fold change, p = 0.015). These include four genes not previously linked to plaque destabilization: GLUL (2.2-fold change, p = 0.016), FUCA1 (2.2-fold change, p = 0.025), IL1RN (1.6-fold change, p = 0.034), and S100A8 (2.5-fold change, p = 0.047). Strong correlations were found to plaque ulceration, plaque hemorrhage, and markers of apoptosis and proliferation (activated caspase 3, TUNEL, and Ki67). Protein expression of these genes was confirmed by immunohistochemistry and was found in the atheromatous areas of CPs critical for plaque destabilization. This study presents a comprehensive transcriptional analysis of stroke-associated CPs and demonstrates a significant transcriptome difference between stroke-associated and asymptomatic CPs. Follow-up studies on the identified genes are needed to define whether they could be used as biomarkers of symptomatic CPs or have a role in plaque destabilization.
Collapse
Affiliation(s)
- Jani Saksi
- Research Programs Unit, Molecular Neurology, Biomedicum Helsinki, University of Helsinki, P.O. Box700, Haartmaninkatu 8, FI-00290, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Agardh HE, Folkersen L, Ekstrand J, Marcus D, Swedenborg J, Hedin U, Gabrielsen A, Paulsson-Berne G. Expression of fatty acid-binding protein 4/aP2 is correlated with plaque instability in carotid atherosclerosis. J Intern Med 2011; 269:200-10. [PMID: 21073559 DOI: 10.1111/j.1365-2796.2010.02304.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE the molecular basis for atherosclerotic plaque vulnerability with high risk of plaque rupture and thromboembolism is complex. We investigated whether clinical estimates of plaque stability correlate with differentially expressed mRNA transcripts within the lesion. METHODS AND RESULTS endarterectomy samples from patients undergoing surgery for symptomatic and asymptomatic carotid stenosis were prospectively collected and clinical parameters recorded in the Biobank of Karolinska Carotid Endarterectomies. mRNA expression profiling (n = 40) and quantitative RT-PCR (n = 105) revealed increased levels of fatty acid-binding protein 4 (FABP4/aP2) in lesions from patients with recent symptoms of plaque instability compared to asymptomatic patients (array: FC = 2, P < 0.05; RT-PCR: P < 0.05). At the mRNA level, FABP4/aP2 correlated with the cell markers CD36, CD68 and CD163 of monocyte/macrophage lineage as well as with CD4-positive T cells. FABP4/aP2 mRNA expression was also correlated with enzymes of the leukotriene pathway, 5-lipoxygenase and leukotriene A4 hydrolase. In addition, analysis of transcript profiles identified CD52 and adipophilin as the mRNAs with the highest correlation with FABP4/aP2. Expression of FABP4/aP2 by macrophages and CD52 by T cells in the lesion was confirmed by immunohistochemistry. CONCLUSIONS expression of FABP4/aP2 is increased at the mRNA level in unstable carotid plaques. Immunohistochemical analyses showed localization of FABP4/aP2 to macrophage populations. These FABP4/aP2-positive macrophages constitute an important and prevalent phenotype and could provide a new link between scavenging-mediated lipid uptake and cellular metabolic stress in plaque. In addition FABP4/aP2 correlates with other important signs of inflammation and plaque instability, such as T cells and leukotriene enzymes. Taken together, these results indicate that FABP4/aP2 is a key factor connecting vascular and cellular lipid accumulation to inflammation.
Collapse
Affiliation(s)
- H E Agardh
- Department of Medicine, Experimental Cardiovascular Research, Karolinska Institutet Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Lin HL, Zhang L, Liu CX, Xu XS, Tang MX, Lv HX, Li CJ, Sun HW, Zhang M, Hong J, Zhang Y. Haemin-enhanced expression of haem oxygenase-1 stabilizes erythrocyte-induced vulnerable atherosclerotic plaques. Br J Pharmacol 2010; 160:1484-95. [PMID: 20590638 DOI: 10.1111/j.1476-5381.2010.00799.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Previous studies demonstrated that intraplaque haemorrhage increased the contents of cholesterol and oxidants in atherosclerotic plaques. The present study was aimed to test the hypothesis that enhanced expression of haem oxygenase-1 (HO-1) may stabilize vulnerable plaques. EXPERIMENTAL APPROACH Intravascular ultrasound (IVUS) was performed to identify three similar abdominal aortic plaques in each of 58 fat-fed New Zealand rabbits after aortic balloon injury. With the guidance of IVUS, 50 microL autologous erythrocytes (RBC) or normal saline (NS) were injected from adventitia into two of the pre-selected plaques, respectively, whereas the third plaque served as a blank control. All rabbits were randomly divided into two groups, receiving intraperitoneal injection of haemin and saline respectively. KEY RESULTS Compared with NS or control plaques, RBC plaques had more macrophage infiltration and lipid content, thinner plaque fibrous cap, and higher expression of inflammatory factors and incidence of plaque rupture. RBC plaques in the haemin group had about a 50% lower incidence of plaque rupture than those in the control group. CONCLUSIONS AND IMPLICATIONS Haem oxygenase-1 may eliminate haem or other oxidants, exert unexpected anti-oxidative and anti-inflammatory effects and serve as a promising approach to the direct inhibition of erythrocyte-induced plaque instability.
Collapse
Affiliation(s)
- Hui Li Lin
- Shandong University Qilu Hospital, Jinan, Shandong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kadl A, Meher AK, Sharma PR, Lee MY, Doran AC, Johnstone SR, Elliott MR, Gruber F, Han J, Chen W, Kensler T, Ravichandran KS, Isakson BE, Wamhoff BR, Leitinger N. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res 2010; 107:737-46. [PMID: 20651288 DOI: 10.1161/circresaha.109.215715] [Citation(s) in RCA: 419] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Macrophages change their phenotype and biological functions depending on the microenvironment. In atherosclerosis, oxidative tissue damage accompanies chronic inflammation; however, macrophage phenotypic changes in response to oxidatively modified molecules are not known. OBJECTIVE To examine macrophage phenotypic changes in response to oxidized phospholipids that are present in atherosclerotic lesions. METHODS AND RESULTS We show that oxidized phospholipid-treated murine macrophages develop into a novel phenotype (Mox) that is strikingly different from the conventional M1 and M2 macrophage phenotypes. Compared to M1 and M2, Mox macrophages show a different gene expression pattern, as well as decreased phagocytotic and chemotactic capacity. Treatment with oxidized phospholipids induces both M1 and M2 macrophages to switch to the Mox phenotype. Whole-genome expression array analysis and subsequent gene ontology clustering revealed that the Mox phenotype was characterized by abundant overrepresentation of Nrf2-mediated expression of redox-regulatory genes. In macrophages isolated from Nrf2(-/-) mice, oxidized phospholipid-induced gene expression and regulation of redox status were compromised. Moreover, we found that Mox macrophages comprise 30% of all macrophages in advanced atherosclerotic lesions of low-density lipoprotein receptor knockout (LDLR(-/-)) mice. CONCLUSIONS Together, we identify Nrf2 as a key regulator in the formation of a novel macrophage phenotype (Mox) that develops in response to oxidative tissue damage. The unique biological properties of Mox macrophages suggest this phenotype may play an important role in atherosclerotic lesion development as well as in other settings of chronic inflammation.
Collapse
Affiliation(s)
- Alexandra Kadl
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Isoviita PM, Nuotio K, Saksi J, Turunen R, Ijäs P, Pitkäniemi J, Soinne L, Kaste M, Kovanen PT, Lindsberg PJ. An Imbalance Between CD36 and ABCA1 Protein Expression Favors Lipid Accumulation in Stroke-Prone Ulcerated Carotid Plaques. Stroke 2010; 41:389-93. [DOI: 10.1161/strokeaha.109.567636] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Pia M. Isoviita
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Krista Nuotio
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Jani Saksi
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Riitta Turunen
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Petra Ijäs
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Janne Pitkäniemi
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Lauri Soinne
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Markku Kaste
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Petri T. Kovanen
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| | - Perttu J. Lindsberg
- From Research Program of Molecular Neurology (P.M.I., K.N., J.S., R.T., P.I., L.S., P.J.L.), Biomedicum Helsinki, Finland; Department of Neurology (K.N., P.I., L.S., M.K., P.J.L.), Helsinki University Central Hospital, Finland; Department of Public Health (J.P.), University of Helsinki, Finland; Wihuri Research Institute (P.T.K.), Helsinki, Finland
| |
Collapse
|
35
|
Gleissner CA, Shaked I, Erbel C, Böckler D, Katus HA, Ley K. CXCL4 downregulates the atheroprotective hemoglobin receptor CD163 in human macrophages. Circ Res 2009; 106:203-11. [PMID: 19910578 DOI: 10.1161/circresaha.109.199505] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
RATIONALE CXCL4 is a platelet-derived chemokine that promotes macrophage differentiation from monocytes. Deletion of the PF4 gene that encodes CXCL4 reduces atherosclerotic lesions in ApoE(-/-) mice. OBJECTIVE We sought to study effects of CXCL4 on macrophage differentiation with possible relevance for atherogenesis. METHODS AND RESULTS Flow cytometry for expression of surface markers in macrophage colony-stimulating factor (M-CSF)- and CXCL4-induced macrophages demonstrated virtually complete absence of the hemoglobin scavenger receptor CD163 in CXCL4-induced macrophages. mRNA for CD163 was downregulated as early as 2 hours after CXCL4. CD163 protein reached a minimum after 3 days, which was not reversed by treatment of cells with M-CSF. The CXCL4 effect was entirely neutralized by heparin, which bound CXCL4 and prevented CXCL4 surface binding to monocytes. Pretreatment of cells with chlorate, which inhibits glycosaminoglycan synthesis, strongly inhibited CXCL4-dependent downregulation of CD163. Similar to recombinant CXCL4, releasate from human platelets also reduced CD163 expression. CXCL4-differentiated macrophages were unable to upregulate the atheroprotective enzyme heme oxygenase-1 at the RNA and protein level in response to hemoglobin-haptoglobin complexes. Immunofluorescence of human atherosclerotic plaques demonstrated presence of both CD68+CD163+ and CD68+CD163- macrophages. PF4 and CD163 gene expression within human atherosclerotic lesions were inversely correlated, supporting the in vivo relevance of CXCL4-induced downregulation of CD163. CONCLUSIONS CXCL4 may promote atherogenesis by suppressing CD163 in macrophages, which are then unable to upregulate the atheroprotective enzyme heme oxygenase-1 in response to hemoglobin.
Collapse
Affiliation(s)
- Christian A Gleissner
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Sinnaeve PR, Donahue MP, Grass P, Seo D, Vonderscher J, Chibout SD, Kraus WE, Sketch M, Nelson C, Ginsburg GS, Goldschmidt-Clermont PJ, Granger CB. Gene expression patterns in peripheral blood correlate with the extent of coronary artery disease. PLoS One 2009; 4:e7037. [PMID: 19750006 PMCID: PMC2736586 DOI: 10.1371/journal.pone.0007037] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 08/09/2009] [Indexed: 11/19/2022] Open
Abstract
Systemic and local inflammation plays a prominent role in the pathogenesis of atherosclerotic coronary artery disease, but the relationship of whole blood gene expression changes with coronary disease remains unclear. We have investigated whether gene expression patterns in peripheral blood correlate with the severity of coronary disease and whether these patterns correlate with the extent of atherosclerosis in the vascular wall. Patients were selected according to their coronary artery disease index (CADi), a validated angiographical measure of the extent of coronary atherosclerosis that correlates with outcome. RNA was extracted from blood of 120 patients with at least a stenosis greater than 50% (CADi≥23) and from 121 controls without evidence of coronary stenosis (CADi = 0). 160 individual genes were found to correlate with CADi (rho>0.2, P<0.003). Prominent differential expression was observed especially in genes involved in cell growth, apoptosis and inflammation. Using these 160 genes, a partial least squares multivariate regression model resulted in a highly predictive model (r2 = 0.776, P<0.0001). The expression pattern of these 160 genes in aortic tissue also predicted the severity of atherosclerosis in human aortas, showing that peripheral blood gene expression associated with coronary atherosclerosis mirrors gene expression changes in atherosclerotic arteries. In conclusion, the simultaneous expression pattern of 160 genes in whole blood correlates with the severity of coronary artery disease and mirrors expression changes in the atherosclerotic vascular wall.
Collapse
Affiliation(s)
- Peter R Sinnaeve
- Duke University Medical Center and Duke Clinical Research Institute, Duke University, Durham, North Carolina, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hägg DA, Olson FJ, Kjelldahl J, Jernås M, Thelle DS, Carlsson LM, Fagerberg B, Svensson PA. Expression of chemokine (C–C motif) ligand 18 in human macrophages and atherosclerotic plaques. Atherosclerosis 2009; 204:e15-20. [DOI: 10.1016/j.atherosclerosis.2008.10.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 09/24/2008] [Accepted: 10/07/2008] [Indexed: 10/21/2022]
|
38
|
Cagnin S, Biscuola M, Patuzzo C, Trabetti E, Pasquali A, Laveder P, Faggian G, Iafrancesco M, Mazzucco A, Pignatti PF, Lanfranchi G. Reconstruction and functional analysis of altered molecular pathways in human atherosclerotic arteries. BMC Genomics 2009; 10:13. [PMID: 19134193 PMCID: PMC2654039 DOI: 10.1186/1471-2164-10-13] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 01/09/2009] [Indexed: 12/24/2022] Open
Abstract
Background Atherosclerosis affects aorta, coronary, carotid, and iliac arteries most frequently than any other body vessel. There may be common molecular pathways sustaining this process. Plaque presence and diffusion is revealed by circulating factors that can mediate systemic reaction leading to plaque rupture and thrombosis. Results We used DNA microarrays and meta-analysis to study how the presence of calcified plaque modifies human coronary and carotid gene expression. We identified a series of potential human atherogenic genes that are integrated in functional networks involved in atherosclerosis. Caveolae and JAK/STAT pathways, and S100A9/S100A8 interacting proteins are certainly involved in the development of vascular disease. We found that the system of caveolae is directly connected with genes that respond to hormone receptors, and indirectly with the apoptosis pathway. Cytokines, chemokines and growth factors released in the blood flux were investigated in parallel. High levels of RANTES, IL-1ra, MIP-1alpha, MIP-1beta, IL-2, IL-4, IL-5, IL-6, IL-7, IL-17, PDGF-BB, VEGF and IFN-gamma were found in plasma of atherosclerotic patients and might also be integrated in the molecular networks underlying atherosclerotic modifications of these vessels. Conclusion The pattern of cytokine and S100A9/S100A8 up-regulation characterizes atherosclerosis as a proinflammatory disorder. Activation of the JAK/STAT pathway is confirmed by the up-regulation of IL-6, STAT1, ISGF3G and IL10RA genes in coronary and carotid plaques. The functional network constructed in our research is an evidence of the central role of STAT protein and the caveolae system to contribute to preserve the plaque. Moreover, Cav-1 is involved in SMC differentiation and dyslipidemia confirming the importance of lipid homeostasis in the atherosclerotic phenotype.
Collapse
Affiliation(s)
- Stefano Cagnin
- CRIBI Biotechnology Centre, University of Padova, Padova, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Slevin M, Wang Q, Font MA, Luque A, Juan-Babot O, Gaffney J, Kumar P, Kumar S, Badimon L, Krupinski J. Atherothrombosis and plaque heterology: different location or a unique disease? Pathobiology 2008; 75:209-25. [PMID: 18580067 DOI: 10.1159/000132382] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 01/08/2008] [Indexed: 11/19/2022] Open
Abstract
Formation of unstable plaques frequently results in atherothrombosis, the major cause for ischaemic stroke, myocardial infarction and peripheral arterial disease. Patients who have symptomatic thrombosis in one vascular bed are at increased risk of disease in other beds. However, the development of the disease in carotid, coronary and peripheral arteries may have different pathophysiology suggesting that more complex treatment protocols may have to be designed to reduce plaque development at different locations. In this review we describe the known risk factors, compare the developmental features of coronary and carotid plaque development and determine their association with end-point ischaemic events. Differences are also seen in the genetic contribution to plaque development as well as in the deregulation of gene and protein expression and cellular signal transduction activity of active cells in regions susceptible to thrombosis. Differences between carotid and coronary artery plaque development might help to explain the differences in anatomopathological appearance and risk of rupture.
Collapse
Affiliation(s)
- M Slevin
- School of Biology, Chemistry and Health Science, Manchester Metropolitan University, Manchester, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Levy AP, Purushothaman KR, Levy NS, Purushothaman M, Strauss M, Asleh R, Marsh S, Cohen O, Moestrup SK, Moller HJ, Zias EA, Benhayon D, Fuster V, Moreno PR. Downregulation of the Hemoglobin Scavenger Receptor in Individuals With Diabetes and the Hp 2-2 Genotype. Circ Res 2007; 101:106-10. [PMID: 17525367 DOI: 10.1161/circresaha.107.149435] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In individuals with diabetes mellitus (DM), the haptoglobin (Hp) genotype is a major determinant of susceptibility to myocardial infarction. We have proposed that this is because of DM and Hp genotype–dependent differences in the response to intraplaque hemorrhage. The macrophage hemoglobin scavenging receptor CD163 plays an essential role in the clearance of hemoglobin released from lysed red blood cells after intraplaque hemorrhage. We sought to test the hypothesis that expression of CD163 is DM and Hp genotype–dependent. CD163 was quantified in plaques by immunohistochemistry, on peripheral blood monocytes (PBMs) by FACS, and as soluble CD163 (sCD163) in plasma by ELISA. In DM plaques, despite an increase in macrophage infiltration, CD163 immunoreactivity was lower, resulting in a dramatic reduction in the percentage of macrophages expressing CD163 (27±2% versus 70±2%,
P
=0.0001). In individuals with DM as compared with individuals without DM, the percentage of PBMs expressing CD163 was reduced (3.7±0.6% versus 7.1±0.9%,
P
<0.002) whereas soluble plasma CD163 was increased (2.6±1.1 μg/mL versus 1.6±0.8 μg/mL,
P
<0.0005). Among DM individuals, the Hp 2-2 genotype was associated with a decrease in the percentage of PBMs expressing CD163 (2.3±0.5% versus 5.6±1.3%,
P
=0.01) and an increase in plasma soluble CD163 (3.0±0.2 μg/mL versus 2.3±0.2 μg/mL,
P
=0.04). Taken together, these results demonstrate an impaired hemoglobin clearance capacity in Hp 2-2 DM individuals and may provide the key insight explaining the increased incidence of myocardial infarction in this population.
Collapse
MESH Headings
- Antigens, CD/biosynthesis
- Antigens, CD/blood
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/biosynthesis
- Antigens, Differentiation, Myelomonocytic/blood
- Antigens, Differentiation, Myelomonocytic/genetics
- Diabetes Mellitus/blood
- Diabetes Mellitus/genetics
- Diabetes Mellitus/pathology
- Down-Regulation/genetics
- Genetic Predisposition to Disease/epidemiology
- Genotype
- Haptoglobins/genetics
- Haptoglobins/metabolism
- Hemoglobins/genetics
- Hemoglobins/metabolism
- Hemorrhage/blood
- Hemorrhage/epidemiology
- Hemorrhage/genetics
- Humans
- Incidence
- Macrophages/metabolism
- Myocardial Infarction/blood
- Myocardial Infarction/epidemiology
- Myocardial Infarction/genetics
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/blood
- Receptors, Cell Surface/genetics
- Receptors, Scavenger/antagonists & inhibitors
- Receptors, Scavenger/blood
- Receptors, Scavenger/genetics
Collapse
Affiliation(s)
- Andrew P Levy
- Technion Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sluimer JC, Kisters N, Cleutjens KB, Volger OL, Horrevoets AJ, van den Akker LH, Bijnens APJ, Daemen MJ. Dead or alive: gene expression profiles of advanced atherosclerotic plaques from autopsy and surgery. Physiol Genomics 2007; 30:335-41. [PMID: 17519360 DOI: 10.1152/physiolgenomics.00076.2007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Since inclusion of atherosclerotic tissues from different sources is often indispensable to study the full atherogenic spectrum, we investigated to what extent the expression profiles of advanced, stable atherosclerotic lesions obtained during autopsy and surgery are comparable. The gene expression profiles of human carotids with advanced atherosclerosis obtained at autopsy and at vascular surgery were studied by microarray analysis. Expression analysis was performed both at the single gene (Rosetta, Gene Ontology) and at the pathway level using Ingenuity and Gene Set Enrichment Analysis. In addition, mRNA and protein expression levels were validated using quantitative (q) RT-PCR and immunohistochemistry on unrelated advanced carotid lesions from autopsy and surgery. Microarray analysis indicated that the 97.2% of genes showed similar expression levels in advanced atherosclerotic lesions from autopsy and surgery. While the expression data revealed no differences in common atherosclerotic related pathways such as lipid metabolism and inflammation, the differentially expressed genes were mainly involved in basal cell metabolism and hypoxia driven pathways. qRT-PCR confirmed the differential expression of hypoxia-driven genes VEGF-A (2.3-fold upward arrow), glucose transporter (GLUT)-1 (2.5-fold upward arrow), GLUT3 (8.3-fold upward arrow), and hexokinase 1 (2.4-fold upward arrow) in autopsy vs. surgical specimens. Immunohistochemistry revealed that the transcriptional differences in these hypoxia-related genes were not reflected at the protein level. The gene expression profiles of advanced atherosclerotic lesions from autopsy and surgery are largely similar. However, >500 genes, mostly involved in basal cell metabolism and hypoxia were differentially expressed at mRNA, but not at the protein level.
Collapse
Affiliation(s)
- Judith C Sluimer
- University of Maastricht, Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Nuotio K, Isoviita PM, Saksi J, Ijäs P, Pitkäniemi J, Sonninen R, Soinne L, Saimanen E, Salonen O, Kovanen PT, Kaste M, Lindsberg PJ. Adipophilin expression is increased in symptomatic carotid atherosclerosis: correlation with red blood cells and cholesterol crystals. Stroke 2007; 38:1791-8. [PMID: 17446422 DOI: 10.1161/strokeaha.106.478867] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Adipophilin is an adipose differentiation-related protein expressed in lipid-containing cells. Using DNA microarray analysis, we previously found the adipophilin gene (ADFP) to be overexpressed in symptomatic carotid plaques (CP). This led us to further examine the role of adipophilin in carotid atherosclerosis relative to symptom status. METHODS Ninety-eight high-grade (>70%) CPs were obtained in carotid endarterectomy. The relative expression of ADFP mRNA was measured by quantitative real-time RT-PCR, and the relative amount of adipophilin protein was quantified with Western blotting. Detailed topographical correlations with extravasated red blood cells and extracellular cholesterol crystals were obtained by means of immunohistochemistry. RESULTS The relative expression of ADFP mRNA was increased in symptomatic compared with asymptomatic CPs at both the mRNA level (1.82+/-0.19[SE] versus 1.25+/-0.15, P=0.012) and the protein level (1.04+/-0.23 versus 0.46+/-0.14, P=0.043). Adipophilin colocalized with macrophage foam cells, extravasated red blood cells (P<0.0001), and cholesterol crystals (P<0.0001), and its expression associated with macroscopic ulceration of CP (P<0.0001). CONCLUSIONS Intraplaque hemorrhages may contribute to intracellular lipid accumulation and consequent adipophilin expression. Because adipophilin blocks cholesterol efflux from lipid-laden cells, they may die and develop a necrotic lipid core, thereby destabilizing the plaque.
Collapse
Affiliation(s)
- Krista Nuotio
- Neuroscience Program, Biomedicum Helsinki, Haartmaninkatu, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|