1
|
Hou N, Zhou H, Li J, Xiong X, Deng H, Xiong S. Macrophage polarization and metabolic reprogramming in abdominal aortic aneurysm. Immun Inflamm Dis 2024; 12:e1268. [PMID: 39530309 PMCID: PMC11555488 DOI: 10.1002/iid3.1268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a macrovascular disease with high morbidity and mortality in the elderly. The limitation of the current management is that most patients can only be followed up until the AAA diameter increases to a threshold, and surgical intervention is recommended. The development of preventive and curative drugs for AAA is urgently needed. Macrophage-mediated immune inflammation is one of the key pathological links in the occurrence and development of AAA. AIMS This review article aims to evaluate the impact of immunometabolism on macrophage biology and its role in AAA. METHODS We analyze publications focusing on the polarization and metabolic reprogramming in macrophages as well as their potential impact on AAA, and summarize the potential interventions that are currently available to regulate these processes. RESULTS The phenotypic and functional changes in macrophages are accompanied by significant alterations in metabolic pathways. The interaction between macrophage polarization and metabolic pathways significantly influences the progression of AAA. CONCLUSION Macrophage polarization is a manifestation of the gross dichotomy of macrophage function into pro-inflammatory killing and tissue repair, that is, classically activated M1 macrophages and alternatively activated M2 macrophages. Macrophage functions are closely linked to metabolic changes, and the emerging field of immunometabolism is providing unique insights into the role of macrophages in AAA. It is essential to further investigate the precise metabolic changes and their functional consequences in AAA-associated macrophages.
Collapse
Affiliation(s)
- Ningxin Hou
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongmin Zhou
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jun Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hongping Deng
- Department of Vascular SurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Sizheng Xiong
- Department of Vascular SurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
2
|
Mackay CDA, Meechem MB, Patel VB. Macrophages in vascular disease: Roles of mitochondria and metabolic mechanisms. Vascul Pharmacol 2024; 156:107419. [PMID: 39181483 DOI: 10.1016/j.vph.2024.107419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Macrophages are a dynamic cell type of the immune system implicated in the pathophysiology of vascular diseases and are a major contributor to pathological inflammation. Excessive macrophage accumulation, activation, and polarization is observed in aortic aneurysm (AA), atherosclerosis, and pulmonary arterial hypertension. In general, macrophages become activated and polarized to a pro-inflammatory phenotype, which dramatically changes cell behavior to become pro-inflammatory and infiltrative. These cell types become cumbersome and fail to be cleared by normal mechanisms such as autophagy. The result is a hyper-inflammatory environment causing the recruitment of adjacent cells and circulating immune cells to further augment the inflammatory response. In AA, this leads to excessive ECM degradation and chemokine secretion, ultimately causing macrophages to dominate the immune cell landscape in the aortic wall. In atherosclerosis, monocytes are recruited to the vascular wall, where they polarize to the pro-inflammatory phenotype and induce inflammatory pathway activation. This leads to the development of foam cells, which significantly contribute to neointima and necrotic core formation in atherosclerotic plaques. Pro-inflammatory macrophages, which affect other vascular diseases, present with fragmented mitochondria and corresponding metabolic dysfunction. Targeting macrophage mitochondrial dynamics has proved to be an exciting potential therapeutic approach to combat vascular disease. This review will summarize mitochondrial and metabolic mechanisms of macrophage activation, polarization, and accumulation in vascular diseases.
Collapse
Affiliation(s)
- Cameron D A Mackay
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Megan B Meechem
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
3
|
Zhang Y, Li G. Predicting feature genes correlated with immune infiltration in patients with abdominal aortic aneurysm based on machine learning algorithms. Sci Rep 2024; 14:5157. [PMID: 38431726 PMCID: PMC10908806 DOI: 10.1038/s41598-024-55941-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 02/29/2024] [Indexed: 03/05/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a condition characterized by a pathological and progressive dilatation of the infrarenal abdominal aorta. The exploration of AAA feature genes is crucial for enhancing the prognosis of AAA patients. Microarray datasets of AAA were downloaded from the Gene Expression Omnibus database. A total of 43 upregulated differentially expressed genes (DEGs) and 32 downregulated DEGs were obtained. Function, pathway, disease, and gene set enrichment analyses were performed, in which enrichments were related to inflammation and immune response. AHR, APLNR, ITGA10 and NR2F6 were defined as feature genes via machine learning algorithms and a validation cohort, which indicated high diagnostic abilities by the receiver operating characteristic curves. The cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) method was used to quantify the proportions of immune infiltration in samples of AAA and normal tissues. We have predicted AHR, APLNR, ITGA10 and NR2F6 as feature genes of AAA. CD8 + T cells and M2 macrophages correlated with these genes may be involved in the development of AAA, which have the potential to be developed as risk predictors and immune interventions.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, 271000, Shandong, China
- Postdoctoral Workstation, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, China
- Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu, China
| | - Gang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, 271000, Shandong, China.
| |
Collapse
|
4
|
Elizondo-Benedetto S, Sastriques-Dunlop S, Detering L, Arif B, Heo GS, Sultan D, Luehmann H, Zhang X, Gao X, Harrison K, Thies D, McDonald L, Combadière C, Lin CY, Kang Y, Zheng J, Ippolito J, Laforest R, Gropler RJ, English SJ, Zayed MA, Liu Y. Chemokine Receptor 2 Is A Theranostic Biomarker for Abdominal Aortic Aneurysms. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.06.23298031. [PMID: 37986880 PMCID: PMC10659515 DOI: 10.1101/2023.11.06.23298031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative vascular disease impacting aging populations with a high mortality upon rupture. There are no effective medical therapies to prevent AAA expansion and rupture. We previously demonstrated the role of the monocyte chemoattractant protein-1 (MCP-1) / C-C chemokine receptor type 2 (CCR2) axis in rodent AAA pathogenesis via positron emission tomography/computed tomography (PET/CT) using CCR2 targeted radiotracer 64 Cu-DOTA-ECL1i. We have since translated this radiotracer into patients with AAA. CCR2 PET showed intense radiotracer uptake along the AAA wall in patients while little signal was observed in healthy volunteers. AAA tissues collected from individuals scanned with 64 Cu-DOTA-ECL1i and underwent open-repair later demonstrated more abundant CCR2+ cells compared to non-diseased aortas. We then used a CCR2 inhibitor (CCR2i) as targeted therapy in our established male and female rat AAA rupture models. We observed that CCR2i completely prevented AAA rupture in male rats and significantly decreased rupture rate in female AAA rats. PET/CT revealed substantial reduction of 64 Cu-DOTA-ECL1i uptake following CCR2i treatment in both rat models. Characterization of AAA tissues demonstrated decreased expression of CCR2+ cells and improved histopathological features. Taken together, our results indicate the potential of CCR2 as a theranostic biomarker for AAA management.
Collapse
|
5
|
Bharadhwaj RA, Kumarswamy R. Long noncoding RNA TUG1 regulates smooth muscle cell differentiation via KLF4-myocardin axis. Am J Physiol Cell Physiol 2023; 325:C940-C950. [PMID: 37642238 PMCID: PMC10635660 DOI: 10.1152/ajpcell.00275.2023] [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: 06/23/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Abdominal aortic aneurysms (AAAs) are asymptomatic vascular diseases that have life-threatening outcomes. Smooth muscle cell (SMC) dysfunction plays an important role in AAA development. The contribution of non-coding genome, specifically the role of long non-coding RNAs (lncRNAs) in SMC dysfunction, is relatively unexplored. We investigated the role of lncRNA TUG1 in SMC dysfunction. To identify potential lncRNAs relevant to SMC functionality, lncRNA profiling was performed in angiotensin-II-treated SMCs. AAA was induced by angiotensin-II treatment in mice. Transcriptional regulation of TUG1 was studied using promoter luciferase and chromatin-immuno-precipitation experiments. Gain-or-loss-of-function experiments were performed in vitro to investigate TUG1-mediated regulation of SMC function. Immunoprecipitation experiments were conducted to elucidate the mechanism underlying TUG1-mediated SMC dysfunction. TUG1 was upregulated in SMCs following angiotensin-II treatment. Similarly, TUG1 levels were elevated in abdominal aorta in a mouse model of angiotensin-II-induced AAA. Further investigations showed that angiotensin-II-induced TUG1 expression could be suppressed by inhibiting Notch-signaling pathway, both in vitro and in mouse AAA model and that TUG1 is a direct transcriptional target of the Notch pathway. In aneurysmal tissues, TUG1 expression was inversely correlated with the expression of SMC contractile genes. Overexpression of TUG1 repressed SMC differentiation in vitro, whereas siRNA/shRNA-mediated TUG1 knockdown showed an opposite effect. Mechanistically, TUG1 interacts with transcriptional repressor KLF4 and facilitates its recruitment to myocardin promoter ultimately leading to the repression of SMC differentiation. In summary, our study uncovers a novel role for the lncRNA TUG1 wherein it modulates SMC differentiation via the KLF4-myocardin axis, which may have potential implications in AAA development.NEW & NOTEWORTHY TUG1 is an angiotensin-II-induced long noncoding RNA that mediates smooth muscle cell (SMC) dysfunction through interaction with transcriptional repressor KLF4.
Collapse
Affiliation(s)
- Ravi Abishek Bharadhwaj
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Regalla Kumarswamy
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Mannion AJ, Holmgren L. Nuclear mechanosensing of the aortic endothelium in health and disease. Dis Model Mech 2023; 16:dmm050361. [PMID: 37909406 PMCID: PMC10629673 DOI: 10.1242/dmm.050361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
The endothelium, the monolayer of endothelial cells that line blood vessels, is exposed to a number of mechanical forces, including frictional shear flow, pulsatile stretching and changes in stiffness influenced by extracellular matrix composition. These forces are sensed by mechanosensors that facilitate their transduction to drive appropriate adaptation of the endothelium to maintain vascular homeostasis. In the aorta, the unique architecture of the vessel gives rise to changes in the fluid dynamics, which, in turn, shape cellular morphology, nuclear architecture, chromatin dynamics and gene regulation. In this Review, we discuss recent work focusing on how differential mechanical forces exerted on endothelial cells are sensed and transduced to influence their form and function in giving rise to spatial variation to the endothelium of the aorta. We will also discuss recent developments in understanding how nuclear mechanosensing is implicated in diseases of the aorta.
Collapse
Affiliation(s)
- Aarren J. Mannion
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| | - Lars Holmgren
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 64, Sweden
| |
Collapse
|
7
|
Ramadhiani R, Ikeda K, Miyagawa K, Ryanto GRT, Tamada N, Suzuki Y, Kirita Y, Matoba S, Hirata KI, Emoto N. Endothelial cell senescence exacerbates pulmonary hypertension by inducing juxtacrine Notch signaling in smooth muscle cells. iScience 2023; 26:106662. [PMID: 37192975 PMCID: PMC10182325 DOI: 10.1016/j.isci.2023.106662] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 03/17/2023] [Accepted: 04/06/2023] [Indexed: 05/18/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease characterized by a progressive increase in pulmonary artery pressure caused by pathological pulmonary artery remodeling. Here, we demonstrate that endothelial cell (EC) senescence plays a negative role in pulmonary hypertension via juxtacrine interaction with smooth muscle cells (SMCs). By using EC-specific progeroid mice, we discovered that EC progeria deteriorated vascular remodeling in the lungs, and exacerbated pulmonary hypertension in mice. Mechanistically, senescent ECs overexpressed Notch ligands, which resulted in increased Notch signaling and activated proliferation and migration capacities in neighboring SMCs. Pharmacological inhibition of Notch signaling reduced the effects of senescent ECs on SMCs functions in vitro, and improved the worsened pulmonary hypertension in EC-specific progeroid mice in vivo. Our findings show that EC senescence is a critical disease-modifying factor in PAH and that EC-mediated Notch signaling is a pharmacotherapeutic target for the treatment of PAH, particularly in the elderly.
Collapse
Affiliation(s)
- Risa Ramadhiani
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Kazuya Miyagawa
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Gusty Rizky Tough Ryanto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Naoki Tamada
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Yoko Suzuki
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
| | - Yuhei Kirita
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Satoaki Matoba
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| |
Collapse
|
8
|
Ling X, Jie W, Qin X, Zhang S, Shi K, Li T, Guo J. Gut microbiome sheds light on the development and treatment of abdominal aortic aneurysm. Front Cardiovasc Med 2022; 9:1063683. [PMID: 36505348 PMCID: PMC9732037 DOI: 10.3389/fcvm.2022.1063683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/03/2022] [Indexed: 11/27/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is an inflammatory vascular disease with high disability and mortality. Its susceptible risk factors include old age, being male, smoking, hypertension, and aortic atherosclerosis. With the improvement of screening techniques, AAA incidence and number of deaths caused by aneurysm rupture increase annually, attracting much clinical attention. Due to the lack of non-invasive treatment, early detection and development of novel treatment of AAA is an urgent clinical concern. The pathophysiology and progression of AAA are characterized by inflammatory destruction. The gut microbiota is an "invisible organ" that directly or indirectly affects the vascular wall inflammatory cell infiltration manifested with enhanced arterial wall gut microbiota and metabolites, which plays an important role in the formation and progression of AAA. As such, the gut microbiome may become an important risk factor for AAA. This review summarizes the direct and indirect effects of the gut microbiome on the pathogenesis of AAA and highlights the gut microbiome-mediated inflammatory responses and discoveries of relevant therapeutic targets that may help manage the development and rupture of AAA.
Collapse
Affiliation(s)
- Xuebin Ling
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Department of Cardiovascular Medicine of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Wei Jie
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Department of Cardiovascular Medicine of the First Affiliated Hospital, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Xue Qin
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Department of Cardiovascular Medicine of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Shuya Zhang
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Department of Cardiovascular Medicine of the First Affiliated Hospital, Hainan Medical University, Haikou, China
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Kaijia Shi
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Department of Cardiovascular Medicine of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Tianfa Li
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Department of Cardiovascular Medicine of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Junli Guo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Department of Cardiovascular Medicine of the First Affiliated Hospital, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| |
Collapse
|
9
|
Xiong T, Lv XS, Wu GJ, Guo YX, Liu C, Hou FX, Wang JK, Fu YF, Liu FQ. Single-Cell Sequencing Analysis and Multiple Machine Learning Methods Identified G0S2 and HPSE as Novel Biomarkers for Abdominal Aortic Aneurysm. Front Immunol 2022; 13:907309. [PMID: 35769488 PMCID: PMC9234288 DOI: 10.3389/fimmu.2022.907309] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/06/2022] [Indexed: 11/20/2022] Open
Abstract
Identifying biomarkers for abdominal aortic aneurysms (AAA) is key to understanding their pathogenesis, developing novel targeted therapeutics, and possibly improving patients outcomes and risk of rupture. Here, we identified AAA biomarkers from public databases using single-cell RNA-sequencing, weighted co-expression network (WGCNA), and differential expression analyses. Additionally, we used the multiple machine learning methods to identify biomarkers that differentiated large AAA from small AAA. Biomarkers were validated using GEO datasets. CIBERSORT was used to assess immune cell infiltration into AAA tissues and investigate the relationship between biomarkers and infiltrating immune cells. Therefore, 288 differentially expressed genes (DEGs) were screened for AAA and normal samples. The identified DEGs were mostly related to inflammatory responses, lipids, and atherosclerosis. For the large and small AAA samples, 17 DEGs, mostly related to necroptosis, were screened. As biomarkers for AAA, G0/G1 switch 2 (G0S2) (Area under the curve [AUC] = 0.861, 0.875, and 0.911, in GSE57691, GSE47472, and GSE7284, respectively) and for large AAA, heparinase (HPSE) (AUC = 0.669 and 0.754, in GSE57691 and GSE98278, respectively) were identified and further verified by qRT-PCR. Immune cell infiltration analysis revealed that the AAA process may be mediated by T follicular helper (Tfh) cells and the large AAA process may also be mediated by Tfh cells, M1, and M2 macrophages. Additionally, G0S2 expression was associated with neutrophils, activated and resting mast cells, M0 and M1 macrophages, regulatory T cells (Tregs), resting dendritic cells, and resting CD4 memory T cells. Moreover, HPSE expression was associated with M0 and M1 macrophages, activated and resting mast cells, Tregs, and resting CD4 memory T cells. Additional, G0S2 may be an effective diagnostic biomarker for AAA, whereas HPSE may be used to confer risk of rupture in large AAAs. Immune cells play a role in the onset and progression of AAA, which may improve its diagnosis and treatment.
Collapse
Affiliation(s)
- Tao Xiong
- Department of Cardiovascular, Shaanxi Provincial People’s Hospital, Xi’an, China
- Department of Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiao-Shuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Gu-Jie Wu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yao-Xing Guo
- Department of Pathology, College of Basic Medical Sciences China Medical University, Shenyang, China
| | - Chang Liu
- Department of Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fang-Xia Hou
- Department of Cardiovascular, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Jun-Kui Wang
- Department of Cardiovascular, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Yi-Fan Fu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Fu-Qiang Liu
- Department of Cardiovascular, Shaanxi Provincial People’s Hospital, Xi’an, China
- *Correspondence: Fu-Qiang Liu,
| |
Collapse
|
10
|
Jones B, Debski A, Hans CP, Go MR, Agarwal G. Structurally abnormal collagen fibrils in abdominal aortic aneurysm resist platelet adhesion. J Thromb Haemost 2022; 20:470-477. [PMID: 34714974 DOI: 10.1111/jth.15576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/16/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Platelet adhesion to the subendothelial collagen fibrils is one of the first steps in hemostasis. Understanding how structural perturbations in the collagen fibril affect platelet adhesion can provide novel insights into disruption of hemostasis in various diseases. We have recently identified the presence of abnormal collagen fibrils with compromised D-periodic banding in the extracellular matrix remodeling present in abdominal aortic aneurysms (AAA). OBJECTIVE In this study, we employed multimodal microscopy approaches to characterize how collagen fibril structure impacts platelet adhesion in clinical AAA tissues. METHODS Ultrastructural atomic force microscopy (AFM) analysis was performed on tissue sections after staining with fluorescently labeled collagen hybridizing peptide (CHP) to recognize degraded collagen. Second harmonic generation (SHG) microscopy was used on CHP-stained sections to identify regions of intact versus degraded collagen. Finally, platelet adhesion was identified via SHG and indirect immunofluorescence on the same tissue sections. RESULTS Our results indicate that ultrastructural features characterizing collagen fibril abnormalities coincide with CHP staining. SHG signal was absent from CHP-positive regions. Additionally, platelet binding was primarily localized to regions with SHG signal. Abnormal collagen fibrils present in AAA (in SHG negative regions) were thus found to inhibit platelet adhesion compared to normal fibrils. CONCLUSIONS Our investigations reveal how the collagen fibril structure in the vessel wall can serve as another regulator of platelet-collagen adhesion. These results can be broadly applied to understand the role of collagen fibril structure in regulating thrombosis or bleeding disorders.
Collapse
Affiliation(s)
- Blain Jones
- Biomedical Engineering Graduate Program, The Ohio State University, Columbus, Ohio, USA
| | - Anna Debski
- Department of Material Science and Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Chetan P Hans
- Department of Cardiovascular Medicine, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Michael R Go
- Division of Vascular Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Gunjan Agarwal
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
11
|
Abstract
Two vasculitides, giant cell arteritis (GCA) and Takayasu arteritis (TAK), are recognized as autoimmune and autoinflammatory diseases that manifest exclusively within the aorta and its large branches. In both entities, the age of the affected host is a critical risk factor. TAK manifests during the 2nd-4th decade of life, occurring while the immune system is at its height of performance. GCA is a disease of older individuals, with infrequent cases during the 6th decade and peak incidence during the 8th decade of life. In both vasculitides, macrophages and T cells infiltrate into the adventitia and media of affected vessels, induce granulomatous inflammation, cause vessel wall destruction, and reprogram vascular cells to drive adventitial and neointimal expansion. In GCA, abnormal immunity originates in an aged immune system and evolves within the aged vascular microenvironment. One hallmark of the aging immune system is the preferential loss of CD8+ T cell function. Accordingly, in GCA but not in TAK, CD8+ effector T cells play a negligible role and anti-inflammatory CD8+ T regulatory cells are selectively impaired. Here, we review current evidence of how the process of immunosenescence impacts the risk for GCA and how fundamental differences in the age of the immune system translate into differences in the granulomatous immunopathology of TAK versus GCA.
Collapse
|
12
|
Vinpocetine protects against the development of experimental abdominal aortic aneurysms. Clin Sci (Lond) 2021; 134:2959-2976. [PMID: 33111936 DOI: 10.1042/cs20201057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Abdominal aortic aneurysm (AAA), commonly occurring in the aged population, is a degenerative disease that dilate and weaken infrarenal aorta due to progressive degeneration of aortic wall integrity. Vinpocetine, a derivative of alkaloid vincamine, has long been used for cerebrovascular disorders and cognitive impairment in the aged population. Recent studies have indicated that vinpocetine antagonizes occlusive vascular disorders such as intimal hyperplasia and atherosclerosis. However, its role in vascular degenerative disease AAA remains unexplored. Herein, we determined the effect of vinpocetine on the formation of AAA as well as the intervention of pre-existing moderate AAA. AAA was induced by periaortic elastase application in C57BL/6J mice. Systemic vinpocetine treatment was applied daily via intraperitoneal injection. We showed that vinpocetine pre-treatment remarkably attenuated aneurysmal dilation assessed by diameter and volume. More importantly, vinpocetine also significantly suppressed the progression of pre-existing moderate AAA in a post-intervention model. Vinpocetine improved multiple cellular and molecular changes associated with AAA, such as elastin degradation, media smooth muscle cell depletion, collagen fibers remodeling and macrophage infiltration in aneurysmal tissues. Vinpocetine potently suppressed tumor necrosis factor-α-induced nuclear factor kappa-light-chain-enhancer of activated B cells activation and proinflammatory mediator expression in primary cultured macrophages in vitro, as well as in the aorta wall in vivo, suggesting vinpocetine conferred anti-AAA effect at least partially via the inhibition of inflammation. Taken together, our findings reveal a novel role of vinpocetine in AAA formation, development and progression. Given the excellent safety profile of vinpocetine, the present study suggests vinpocetine may be a novel therapeutic agent for AAA prevention and treatment.
Collapse
|
13
|
Phie J, Thanigaimani S, Golledge J. Systematic Review and Meta-Analysis of Interventions to Slow Progression of Abdominal Aortic Aneurysm in Mouse Models. Arterioscler Thromb Vasc Biol 2021; 41:1504-1517. [PMID: 33567871 DOI: 10.1161/atvbaha.121.315942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- James Phie
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry (J.P., S.T., J.G.), James Cook University, Townsville, Australia
| | - Shivshankar Thanigaimani
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry (J.P., S.T., J.G.), James Cook University, Townsville, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry (J.P., S.T., J.G.), James Cook University, Townsville, Australia.,Australian Institute of Tropical Health and Medicine (J.G.), James Cook University, Townsville, Australia.,Department of Vascular and Endovascular Surgery, Townsville University Hospital, Queensland, Australia (J.G.)
| |
Collapse
|
14
|
Yuan Z, Lu Y, Wei J, Wu J, Yang J, Cai Z. Abdominal Aortic Aneurysm: Roles of Inflammatory Cells. Front Immunol 2021; 11:609161. [PMID: 33613530 PMCID: PMC7886696 DOI: 10.3389/fimmu.2020.609161] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
Abdominal aortic aneurysms (AAAs) are local dilations of infrarenal segment of aortas. Molecular mechanisms underlying the pathogenesis of AAA remain not fully clear. However, inflammation has been considered as a central player in the development of AAA. In the past few decades, studies demonstrated a host of inflammatory cells, including T cells, macrophages, dendritic cells, neutrophils, B cells, and mast cells, etc. infiltrating into aortic walls, which implicated their crucial roles. In addition to direct cell contacts and cytokine or protease secretions, special structures like inflammasomes and neutrophil extracellular traps have been investigated to explore their functions in aneurysm formation. The above-mentioned inflammatory cells and associated structures may initiate and promote AAA expansion. Understanding their impacts and interaction networks formation is meaningful to develop new strategies of screening and pharmacological interventions for AAA. In this review, we aim to discuss the roles and mechanisms of these inflammatory cells in AAA pathogenesis.
Collapse
Affiliation(s)
- Zhen Yuan
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Lu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jia Wei
- Department of Urology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Wu
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jin Yang
- Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.,Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Zhejun Cai
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Jiaxing Key Laboratory of Cardiac Rehabilitation, Jiaxing, China
| |
Collapse
|
15
|
Ni XQ, Zhang YR, Jia LX, Lu WW, Zhu Q, Ren JL, Chen Y, Zhang LS, Liu X, Yu YR, Jia MZ, Ning ZP, Du J, Tang CS, Qi YF. Inhibition of Notch1-mediated inflammation by intermedin protects against abdominal aortic aneurysm via PI3K/Akt signaling pathway. Aging (Albany NY) 2021; 13:5164-5184. [PMID: 33535178 PMCID: PMC7950288 DOI: 10.18632/aging.202436] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022]
Abstract
The Notch1-mediated inflammatory response participates in the development of abdominal aortic aneurysm (AAA). The vascular endogenous bioactive peptide intermedin (IMD) plays an important role in maintaining vascular homeostasis. However, whether IMD inhibits AAA by inhibiting Notch1-mediated inflammation is unclear. In this study, we found Notch intracellular domain (NICD) and hes1 expression were higher in AAA patients’ aortas than in healthy controls. In angiotensin II (AngII)-induced AAA mouse model, IMD treatment significantly reduced AAA incidence and maximal aortic diameter. IMD inhibited AngII-enlarged aortas and -degraded elastic lamina, reduced NICD, hes1 and inflammatory factors expression, decreased infiltration of CD68 positive macrophages and the NOD-like receptor family pyrin domain containing 3 protein level. IMD inhibited lipopolysaccharide-induced macrophage migration in vitro and regulated macrophage polarization. Moreover, IMD overexpression significantly reduced CaCl2-induced AAA incidence and down-regulated NICD and hes1 expression. However, IMD deficiency showed opposite results. Mechanically, IMD treatment significantly decreased cleavage enzyme-a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) level. Pre-incubation with IMD17-47 (IMD receptors blocking peptide) and the phosphatidylinositol 3-kinase/protein kinase b (PI3K/Akt) inhibitor LY294002 reversed ADAM10 level. In conclusion, exogenous and endogenous IMD could inhibit the development of AAA by inhibiting Notch1 signaling-mediated inflammation via reducing ADAM10 through IMD receptor and PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xian-Qiang Ni
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Ya-Rong Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Li-Xin Jia
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, Beijing 100029, China
| | - Wei-Wei Lu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Qing Zhu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Jin-Ling Ren
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Yao Chen
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Lin-Shuang Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Xin Liu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Yan-Rong Yu
- Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Mo-Zhi Jia
- Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Zhong-Ping Ning
- Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Jie Du
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, Beijing 100029, China
| | - Chao-Shu Tang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing 100083, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing 100083, China.,Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing 100083, China
| |
Collapse
|
16
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
DAPT, a potent Notch inhibitor regresses actively growing abdominal aortic aneurysm via divergent pathways. Clin Sci (Lond) 2020; 134:1555-1572. [PMID: 32490531 DOI: 10.1042/cs20200456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a localized pathological dilation of the aorta exceeding the normal diameter (∼20 mm) by more than 50% of its original size (≥30 mm), accounting for approximately 150000-200000 deaths worldwide per year. We previously reported that Notch inhibition does not decrease the size of pre-established AAA at late stage of the disease. Here, we examined whether a potent pharmacologic inhibitor of Notch signaling (DAPT (N-[N-(3,5-Difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester)), regresses an actively growing AAA. In a mouse model of an aneurysm (Apoe-/- mice; n=44); DAPT (n=17) or vehicle (n=17) was randomly administered at day 14 of angiotensin II (AngII; 1 µg/min/kg), three times a week and mice were killed on day 42. Progressive increase in aortic stiffness and maximal intraluminal diameter (MILD) was observed in the AngII + vehicle group, which was significantly prevented by DAPT (P<0.01). The regression of aneurysm with DAPT was associated with reduced F4/80+Cd68+ (cluster of differentiation 68) inflammatory macrophages. DAPT improved structural integrity of aorta by reducing collagen fibrils abnormality and restoring their diameter. Mechanistically, C-C chemokine receptor type 7 (Ccr7)+F4/80- dendritic cells (DCs), implicated in the regression of aneurysm, were increased in the aorta of DAPT-treated mice. In the macrophages stimulated with AngII or lipopolysaccharide (LPS), DAPT reverted the expression of pro-inflammatory genes Il6 and Il12 back to baseline within 6 h compared with vehicle (P<0.05). DAPT also significantly increased the expression of anti-inflammatory genes, including c-Myc, Egr2, and Arg1 at 12-24 h in the LPS-stimulated macrophages (P<0.05). Overall, these regressive effects of Notch signaling inhibitor emphasize its therapeutic implications to prevent the progression of active AAAs.
Collapse
|
18
|
Abstract
Purpose Oestrogen receptor β is believed to exert a cardioprotective effect against ischaemic injury. Nonetheless, the mechanism underlying its protective action remains to be fully elucidated. Recently, increased attention has been focused on Notch1 signalling for ameliorating cardiac ischaemic injury. Here, we hypothesised that oestrogen receptor β activation attenuates myocardial infarction (MI)-induced cardiac damage by modulating the Notch1 signalling pathway. Methods Male C57BL/6 mice were used to establish an MI model through the ligation of the anterior descending branch of the left coronary artery. Two chemical drugs, 2,3-Bis(4-hydroxyphenyl)-propionitrile (DPN) and N-[N-(3,5-difluorophenacetyl)-l-alanyl]-s-phenylglycine t-butyl ester (DAPT), a specific inhibitor of Notch1 signalling) were administered via intraperitoneal injection to change oestrogen receptor β and Notch1 activities. Immunohistochemistry, western blot analysis, enzyme-linked immunosorbent assay (Elisa) assessment and echocardiography were used in this study to analyse cardiac oxidative stress, apoptosis, infraction volume, fibrosis and cardiac function. Results DPN-mediated oestrogen receptor β activation effectively protected cardiomyocytes from MI-induced oxidative damage and apoptosis. Furthermore, oestrogen receptor β activation reduced the infarct size and lowered the levels of myocardial enzymes in the serum, thereby leading to greater overall cardiac function improvement. Ischaemic injury–induced myocardial fibrosis was attenuated by oestrogen receptor β activation. Nevertheless, all of these cardioprotective effects of oestrogen receptor β activation were almost abrogated by DAPT administration, i.e. DAPT attenuated the anti-oxidative and anti-apoptotic effects and the decrease in infarct and fibrotic areas and reversed cardiac functional recovery. The levels of phospho-phosphatidylinositol-3-kinase (PI3K) and phospho-protein kinase B (Akt) were increased after DPN administration, and this change was reversed after DAPT was administered. Conclusions All of these new findings indicate that oestrogen receptor β activation is effective in ameliorating MI-induced cardiac dysfunction by enhancing Notch1 signalling and that PI3K/Akt signalling is the downstream mediator. Electronic supplementary material The online version of this article (10.1007/s10557-020-06949-3) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Hao Y, Wang X, Zhang F, Wang M, Wang Y, Wang H, Du Y, Wang T, Fu F, Gao Z, Zhang L. Inhibition of notch enhances the anti-atherosclerotic effects of LXR agonists while reducing fatty liver development in ApoE-deficient mice. Toxicol Appl Pharmacol 2020; 406:115211. [PMID: 32853627 DOI: 10.1016/j.taap.2020.115211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/08/2020] [Accepted: 08/21/2020] [Indexed: 01/03/2023]
Abstract
Liver X receptor (LXR) activation can achieve satisfactory anti-atherosclerotic activity, but can also lead to the development of fatty liver and hypertriglyceridemia. In contrast, Notch inhibition can suppress both atherosclerosis and the hepatic accumulation of lipids. In the present study, we sought to assess whether combining LXR ligand agonists (T317) with Notch receptor inhibitors (DAPT) would lead to enhanced anti-atherosclerotic activity while overcoming the adverse events associated with LXR ligand agonist therapy. The impact of the combined T317 + DAPT therapeutic regimen on atherosclerosis, fatty liver development, and hypertriglyceridemia was assessed using ApoE deficient (ApoE-/-) mice. The results of this analysis suggested that DAPT was able to improve the anti-atherosclerotic activity of T317 without reducing the stability of lesion plaques while simultaneously reducing blood lipids in treated ApoE-/- mice. This combination T317 + DAPT treatment was also linked with a significant upregulation of ABCA1 and the stimulation of reverse cholesterol transport (RCT), as well as with decreases in the levels of intercellular cell adhesion molecule-1 (ICAM-1) and p-p65, and with altered M1/M2 macrophage proportions within atherosclerotic plaques. Importantly, DAPT was also able to reduce T317-mediated lipid accumulation within the liver owing to its ability to reduce SREBP-1 expression while simultaneously increasing that of Pi-AMPKα and PPARα. Together, our results suggest that administering Notch receptor inhibitors to ApoE-/- mice may be an effective means of enhancing the anti-atherosclerotic activity of LXR ligand agonists while simultaneously limiting associated fatty liver and hypertriglyceridemia development in these animals.
Collapse
Affiliation(s)
- Yanfei Hao
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Xinlin Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Fenglan Zhang
- Yantai Yuhuangding Hospital, The Affiliated Hospital of Qingdao University, Yantai 264000, China
| | - Meiling Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Yanfang Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Hao Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Yuan Du
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Tian Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Fenghua Fu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
| | - Zhuye Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100089, China.
| | - Leiming Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China.
| |
Collapse
|
20
|
Jones B, Tonniges JR, Debski A, Albert B, Yeung DA, Gadde N, Mahajan A, Sharma N, Calomeni EP, Go MR, Hans CP, Agarwal G. Collagen fibril abnormalities in human and mice abdominal aortic aneurysm. Acta Biomater 2020; 110:129-140. [PMID: 32339711 DOI: 10.1016/j.actbio.2020.04.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/27/2020] [Accepted: 04/10/2020] [Indexed: 12/19/2022]
Abstract
Vascular diseases like abdominal aortic aneurysms (AAA) are characterized by a drastic remodeling of the vessel wall, accompanied with changes in the elastin and collagen content. At the macromolecular level, the elastin fibers in AAA have been reported to undergo significant structural alterations. While the undulations (waviness) of the collagen fibers is also reduced in AAA, very little is understood about changes in the collagen fibril at the sub-fiber level in AAA as well as in other vascular pathologies. In this study we investigated structural changes in collagen fibrils in human AAA tissue extracted at the time of vascular surgery and in aorta extracted from angiotensin II (AngII) infused ApoE-/- mouse model of AAA. Collagen fibril structure was examined using transmission electron microscopy and atomic force microscopy. Images were analyzed to ascertain length and depth of D-periodicity, fibril diameter and fibril curvature. Abnormal collagen fibrils with compromised D-periodic banding were observed in the excised human tissue and in remodeled regions of AAA in AngII infused mice. These abnormal fibrils were characterized by statistically significant reduction in depths of D-periods and an increased curvature of collagen fibrils. These features were more pronounced in human AAA as compared to murine samples. Thoracic aorta from Ang II-infused mice, abdominal aorta from saline-infused mice, and abdominal aorta from non-AAA human controls did not contain abnormal collagen fibrils. The structural alterations in abnormal collagen fibrils appear similar to those reported for collagen fibrils subjected to mechanical overload or chronic inflammation in other tissues. Detection of abnormal collagen could be utilized to better understand the functional properties of the underlying extracellular matrix in vascular as well as other pathologies. STATEMENT OF SIGNIFICANCE: Several vascular diseases including abdominal aortic aneurysm (AAA) are characterized by extensive remodeling in the vessel wall. Although structural alterations in elastin fibers are well characterized in vascular diseases, very little is known about the collagen fibril structure in these diseases. We report here a comprehensive ultrastructural evaluation of the collagen fibrils in AAA, using high-resolution microscopy techniques like transmission electron microscopy (TEM) and atomic force microscopy (AFM). We elucidate how abnormal collagen fibrils with compromised D-periodicity and increased fibril curvature are present in the vascular tissue in both clinical AAA as well as in murine models. We discuss how these abnormal collagen fibrils are likely a consequence of mechanical overload accompanying AAA and could impact the functional properties of the underlying tissue.
Collapse
Affiliation(s)
- Blain Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Jeffrey R Tonniges
- Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Anna Debski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Benjamin Albert
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - David A Yeung
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Nikhit Gadde
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Advitiya Mahajan
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, OH, USA; Department of Cardiovascular Medicine, Dalton Cardiovascular Research Center, and Medical Pharmacology and Physiology Columbia, University of Missouri, USA
| | - Neekun Sharma
- Department of Cardiovascular Medicine, Dalton Cardiovascular Research Center, and Medical Pharmacology and Physiology Columbia, University of Missouri, USA
| | - Edward P Calomeni
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Michael R Go
- Division of Vascular Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Chetan P Hans
- Center for Cardiovascular Research, Nationwide Children's Hospital, Columbus, OH, USA; Department of Cardiovascular Medicine, Dalton Cardiovascular Research Center, and Medical Pharmacology and Physiology Columbia, University of Missouri, USA.
| | - Gunjan Agarwal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA; Biophysics Graduate Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
21
|
Sharma N, Belenchia AM, Toedebusch R, Pulakat L, Hans CP. AT2R agonist NP-6A4 mitigates aortic stiffness and proteolytic activity in mouse model of aneurysm. J Cell Mol Med 2020; 24:7393-7404. [PMID: 32420690 PMCID: PMC7339180 DOI: 10.1111/jcmm.15342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical and experimental studies show that angiotensin II (AngII) promotes vascular pathology via activation of AngII type 1 receptors (AT1Rs). We recently reported that NP-6A4, a selective peptide agonist for AngII type 2 receptor (AT2R), exerts protective effects on human vascular cells subjected to serum starvation or doxorubicin exposure. In this study, we investigated whether NP-6A4-induced AT2R activation could mitigate AngII-induced abdominal aortic aneurism (AAA) using AngII-treated Apoe-/- mice. Male Apoe-/- mice were infused with AngII (1 µg/kg/min) by implanting osmotic pumps subcutaneously for 28 days. A subset of mice was pre-treated subcutaneously with NP-6A4 (2.5 mg/kg/day) or vehicle for 14 days prior to AngII, and treatments were continued for 28 days. NP-6A4 significantly reduced aortic stiffness of the abdominal aorta induced by AngII as determined by ultrasound functional analyses and histochemical analyses. NP-6A4 also increased nitric oxide bioavailability in aortic tissues and suppressed AngII-induced increases in monocyte chemotactic protein-1, osteopontin and proteolytic activity of the aorta. However, NP-6A4 did not affect maximal intraluminal aortic diameter or AAA incidences significantly. These data suggest that the effects of AT2R agonist on vascular pathologies are selective, affecting the aortic stiffness and proteolytic activity without affecting the size of AAA.
Collapse
Affiliation(s)
- Neekun Sharma
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Anthony M Belenchia
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Ryan Toedebusch
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Lakshmi Pulakat
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.,Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.,Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Chetan P Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.,Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
22
|
Rizzo P, Vieceli Dalla Sega F, Fortini F, Marracino L, Rapezzi C, Ferrari R. COVID-19 in the heart and the lungs: could we "Notch" the inflammatory storm? Basic Res Cardiol 2020; 115:31. [PMID: 32274570 PMCID: PMC7144545 DOI: 10.1007/s00395-020-0791-5] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 03/30/2020] [Indexed: 01/08/2023]
Abstract
From January 2020, coronavirus disease (COVID-19) originated in China has spread around the world. The disease is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The presence of myocarditis, cardiac arrest, and acute heart failure in COVID-19 patients suggests the existence of a relationship between SARS-CoV-2 infection and cardiac disease. The Notch signalling is a major regulator of cardiovascular function and it is also implicated in several biological processes mediating viral infections. In this report we discuss the possibility to target Notch signalling to prevent SARS-CoV-2 infection and interfere with the progression of COVID-19- associated heart and lungs disease.
Collapse
Affiliation(s)
- Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy.
| | | | | | - Luisa Marracino
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Claudio Rapezzi
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
- Cardiovascular Center, University Hospital of Cona, Ferrara, Italy
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
- Cardiovascular Center, University Hospital of Cona, Ferrara, Italy
| |
Collapse
|
23
|
Jabłońska A, Neumayer C, Bolliger M, Burghuber C, Klinger M, Demyanets S, Nanobachvili J, Huk I. Insight into the expression of toll-like receptors 2 and 4 in patients with abdominal aortic aneurysm. Mol Biol Rep 2020; 47:2685-2692. [PMID: 32146682 DOI: 10.1007/s11033-020-05366-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/29/2020] [Indexed: 12/15/2022]
Abstract
An abdominal aortic aneurysm (AAA) is a relatively common, life-threatening disease prevalent in persons over the age of 65. In recent years, an increasing number of studies have suggested that pattern-recognition receptors (PRRs), including Toll-like receptors (TLRs), may serve as important regulators in the development of AAAs. In this study, we evaluated the TLR2 and TLR4 expression in the aortic wall and blood of patients with AAA. The TLR2 and TLR4 mRNA expression were significantly higher in the blood of patients with AAA than in the blood of healthy volunteers (p = 0.009 and p = 0.010, respectively). The expression of TLR2 and TLR4 transcripts was also higher in the blood compared with the aortic wall tissue of AAA patients (p = 0.001 for both). Higher TLR2 protein expression was observed in the aortic wall of AAA patients compared with the blood (p = 0.026). A significantly higher concentration of TNF-α and IL-4 in patients with AAA than in healthy volunteers (p < 0.001 for both) was noticed. This study suggests that TLR2 may play a role in the inflammatory response in the aorta, both locally and systemically, in patients with AAA.
Collapse
Affiliation(s)
- Agnieszka Jabłońska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, 106 St., 93-232, Lodz, Poland. .,Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria.
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Michael Bolliger
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Christopher Burghuber
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Markus Klinger
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Josif Nanobachvili
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| | - Ihor Huk
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, Vienna General Hospital, Vienna, Austria
| |
Collapse
|
24
|
Sharma N, Sun Z, Hill MA, Hans CP. Measurement of Pulse Propagation Velocity, Distensibility and Strain in an Abdominal Aortic Aneurysm Mouse Model. J Vis Exp 2020:10.3791/60515. [PMID: 32150160 PMCID: PMC7890464 DOI: 10.3791/60515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
An abdominal aortic aneurysm (AAA) is defined as a localized dilation of the abdominal aorta that exceeds the maximal intraluminal diameter (MILD) by 1.5 times of its original size. Clinical and experimental studies have shown that small aneurysms may rupture, while a subpopulation of large aneurysms may remain stable. Thus, in addition to the measurement of intraluminal diameter of the aorta, knowledge of structural traits of the vessel wall may provide important information to assess the stability of the AAA. Aortic stiffening has recently emerged as a reliable tool to determine early changes in the vascular wall. Pulse propagation velocity (PPV) along with the distensibility and radial strain are highly useful ultrasound-based methods relevant for assessing aortic stiffness. The primary purpose of this protocol is to provide a comprehensive technique for the use of ultrasound imaging system to acquire images and analyze the structural and functional properties of the aorta as determined by MILD, PPV, distensibility and radial strain.
Collapse
Affiliation(s)
- Neekun Sharma
- Division of Cardiovascular Medicine, University of Missouri; Dalton Cardiovascular Research Center, University of Missouri
| | - Zhe Sun
- Medical Pharmacology and Physiology, University of Missouri; Dalton Cardiovascular Research Center, University of Missouri
| | - Michael A Hill
- Division of Cardiovascular Medicine, University of Missouri; Medical Pharmacology and Physiology, University of Missouri; Dalton Cardiovascular Research Center, University of Missouri
| | - Chetan P Hans
- Division of Cardiovascular Medicine, University of Missouri; Medical Pharmacology and Physiology, University of Missouri; Dalton Cardiovascular Research Center, University of Missouri;
| |
Collapse
|
25
|
Li Z, Kong W. Cellular signaling in Abdominal Aortic Aneurysm. Cell Signal 2020; 70:109575. [PMID: 32088371 DOI: 10.1016/j.cellsig.2020.109575] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022]
Abstract
Abdominal aortic aneurysms (AAAs) are highly lethal cardiovascular diseases without effective medications. However, the molecular and signaling mechanisms remain unclear. A series of pathological cellular processes have been shown to contribute to AAA formation, including vascular extracellular matrix remodeling, inflammatory and immune responses, oxidative stress, and dysfunction of vascular smooth muscle cells. Each cellular process involves complex cellular signaling, such as NF-κB, MAPK, TGFβ, Notch and inflammasome signaling. In this review, we discuss how cellular signaling networks function in various cellular processes during the pathogenesis and progression of AAA. Understanding the interaction of cellular signaling networks with AAA pathogenesis as well as the crosstalk of different signaling pathways is essential for the development of novel therapeutic approaches to and personalized treatments of AAA diseases.
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
26
|
Sharma N, Dev R, Belenchia AM, Aroor AR, Whaley-Connell A, Pulakat L, Hans CP. Deficiency of IL12p40 (Interleukin 12 p40) Promotes Ang II (Angiotensin II)-Induced Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2019; 39:212-223. [PMID: 30580570 DOI: 10.1161/atvbaha.118.311969] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Objective- Abdominal aortic aneurysm is caused by the accumulation of inflammatory cells in the aortic wall. Our recent studies demonstrated that inhibition of Notch signaling attenuates abdominal aortic aneurysm formation by shifting the macrophage balance towards anti-inflammatory (M2) phenotype. Using IL12p40-/- (interleukin 12 p40) mice, we investigated the effects of M2-predominant macrophages on the development of abdominal aortic aneurysm. Approach and Results- Male (8-10 week-old) wild-type and IL12p40-/- mice (n=15) on C57BL/6 background were infused with Ang II (angiotensin II, 1000 ng/kg per minute) by implanting osmotic pumps subcutaneously for 28 days. In the IL12p40-/- mice, Ang II significantly increased the maximal intraluminal diameter (9/15) as determined by transabdominal ultrasound imaging. In addition, IL12p40-deletion significantly increased aortic stiffness in response to Ang II as measured by pulse wave velocity and atomic force microscopy. Histologically, IL12p40-/- mice exhibited increased maximal external diameter of aorta and aortic lesions associated with collagen deposition and increased elastin fragmentation compared with wild-type mice infused with Ang II. Mechanistically, IL12p40 deficiency by siRNA (small interfering RNA) augmented the Tgfβ2-mediated Mmp2 expression in wild-type bone marrow-derived macrophages without affecting the expression of Mmp9. No such effects of IL12p40 deficiency on MMP2/MMP9 was observed in human aortic smooth muscle cells or fibroblasts. Depletion of macrophages in IL12p40-/- mice by clodronate liposomes significantly decreased the maximal external diameter of aorta and aortic stiffness in response to Ang II as determined by imaging and atomic force microscopy. Conclusions- IL12p40 depletion promotes the development of abdominal aortic aneurysm, in part, by facilitating recruitment of M2-like macrophages and potentiating aortic stiffness and fibrosis mediated by Tgfβ2.
Collapse
Affiliation(s)
- Neekun Sharma
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Rishabh Dev
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Anthony M Belenchia
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Annayya R Aroor
- Department of Medical Pharmacology and Physiology (A.R.A., C.P.H.), University of Missouri, Columbia
| | - Adam Whaley-Connell
- Harry S. Truman Memorial Veterans' Hospital (A.W.-C.), University of Missouri, Columbia
| | - Lakshmi Pulakat
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| | - Chetan P Hans
- From the Department of Cardiovascular Medicine (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia.,Department of Medical Pharmacology and Physiology (A.R.A., C.P.H.), University of Missouri, Columbia.,Dalton Cardiovascular Research Center (N.S., R.D., A.M.B., L.P., C.P.H.), University of Missouri, Columbia
| |
Collapse
|
27
|
Yoshimura K, Morikage N, Nishino-Fujimoto S, Furutani A, Shirasawa B, Hamano K. Current Status and Perspectives on Pharmacologic Therapy for Abdominal Aortic Aneurysm. Curr Drug Targets 2019; 19:1265-1275. [PMID: 29284386 PMCID: PMC6182934 DOI: 10.2174/1389450119666171227223331] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 01/16/2023]
Abstract
Background: Abdominal aortic aneurysm (AAA), a common disease involving the segmen-tal expansion and rupture of the aorta, has a high mortality rate. Therapeutic options for AAA are cur-rently limited to surgical repair to prevent catastrophic rupture. Non-surgical approaches, particularly pharmacotherapy, are lacking for the treatment of AAA. Objective: We review both basic and clinical studies and discuss the current challenges to developing medical therapy that reduces AAA progression. Results: Studies using animal models of AAA progression and human AAA explant cultures have identified several potential targets for preventing AAA growth. However, no clinical studies have con-vincingly confirmed the efficacy of any pharmacologic treatment against the growth of AAA. Thus, there is as yet no strong recommendation regarding pharmacotherapy to reduce the risk of AAA pro-gression and rupture. Conclusion: This review identifies concerns that need to be addressed for the field to progress and dis-cusses the challenges that must be overcome in order to develop effective pharmacotherapy to reduce AAA progression in the future.
Collapse
Affiliation(s)
- Koichi Yoshimura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan.,Graduate School of Health and Welfare, Yamaguchi Prefectural University, Yamaguchi, 753-8502, Japan
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Shizuka Nishino-Fujimoto
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Akira Furutani
- Department of Surgery, Yamaguchi Rosai Hospital, Sanyo-Onoda, 756-0095, Japan
| | - Bungo Shirasawa
- Department of Medical Education, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| |
Collapse
|
28
|
Sharma N, Dev R, Ruiz-Rosado JDD, Partida-Sanchez S, Guerau-de-Arellano M, Dhakal P, Kuivaniemi H, Hans CP. Pharmacological inhibition of Notch signaling regresses pre-established abdominal aortic aneurysm. Sci Rep 2019; 9:13458. [PMID: 31530833 PMCID: PMC6748927 DOI: 10.1038/s41598-019-49682-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by transmural infiltration of myeloid cells at the vascular injury site. Previously, we reported preventive effects of Notch deficiency on the development of AAA by reduction of infiltrating myeloid cells. In this study, we examined if Notch inhibition attenuates the progression of pre-established AAA and potential implications. Pharmacological Notch inhibitor (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-(S)-phenylglycine t-butyl ester; DAPT) was administered subcutaneously three times a week starting at day 28 of angiotensin II (AngII) infusion. Progressive increase in pulse wave velocity (PWV), maximal intra-luminal diameter (MILD) and maximal external aortic diameter (MEAD) were observed at day 56 of the AngII. DAPT prevented such increase in MILD, PWV and MEAD (P < 0.01). Histologically, the aortae of DAPT-treated Apoe-/- mice had significant reduction in inflammatory response and elastin fragmentation. Naked collagen microfibrils and weaker banded structure observed in the aortae of Apoe-/- mice in response to AngII, were substantially diminished by DAPT. A significant decrease in the proteolytic activity in the aneurysmal tissues and vascular smooth muscle cells (vSMCs) was observed with DAPT (P < 0.01). In human and mouse AAA tissues, increased immunoreactivity of activated Notch signaling correlated strongly with CD38 expression (R2 = 0.61). Collectively, we propose inhibition of Notch signaling as a potential therapeutic target for AAA progression.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/metabolism
- Angiotensin II/adverse effects
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/diagnostic imaging
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/metabolism
- Cells, Cultured
- Collagen/metabolism
- Cytokines/metabolism
- Dipeptides/pharmacology
- Disease Models, Animal
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Gene Expression Regulation/drug effects
- Humans
- Male
- Membrane Glycoproteins/metabolism
- Mice
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Receptors, Notch/antagonists & inhibitors
- Receptors, Notch/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Neekun Sharma
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA
| | - Rishabh Dev
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA
| | - Juan de Dios Ruiz-Rosado
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Medical Laboratory Science Division, The Ohio State University, Columbus, OH, USA
| | - Pramod Dhakal
- Animal Science Research Center, University of Missouri, Columbia, USA
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Chetan P Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA.
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA.
- Medical Pharmacology and Physiology, University of Missouri, Columbia, USA.
| |
Collapse
|
29
|
Yan H, Hu Y, Akk A, Ye K, Bacon J, Pham CTN. Interleukin-12 and -23 blockade mitigates elastase-induced abdominal aortic aneurysm. Sci Rep 2019; 9:10447. [PMID: 31320700 PMCID: PMC6639297 DOI: 10.1038/s41598-019-46909-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/05/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages play an important role in the inflammatory process that contributes to the development of abdominal aortic aneurysm (AAA). Studies of human and mouse AAA tissue reveal expanded populations of macrophages producing an abundance of pro-inflammatory cytokines, including TNF-α, IL-12p40 and high level of metalloprotease 9 (MMP-9) at the late stages of disease. Herein, we show that blockade of IL-12p40 in the early phase of aneurysm development suppresses macrophage expansion, inflammatory cytokine and MMP-9 production and mitigates AAA development. Since IL-12 and IL-23 are related cytokines that share the common p40 subunit, we also evaluate the effect of direct IL-23 blockade on the development of AAA. Specific IL-23p19 blockade prevents AAA progression with the same efficiency as IL-12p40 antagonism, suggesting that the efficacy of anti-IL-12p40 treatment may reflect IL-23 blockade. IL-12p40 and IL-23p19 are also abundantly expressed in human AAA tissue. Our findings have potential translational value since IL-12p40 and IL-23p19 antagonists already exist as FDA-approved therapeutics for various chronic inflammatory conditions.
Collapse
Affiliation(s)
- Huimin Yan
- John Cochran VA Medical Center, Saint Louis, Missouri, USA.,Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Ying Hu
- John Cochran VA Medical Center, Saint Louis, Missouri, USA.,Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Antonina Akk
- John Cochran VA Medical Center, Saint Louis, Missouri, USA.,Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Karen Ye
- John Cochran VA Medical Center, Saint Louis, Missouri, USA.,Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - John Bacon
- John Cochran VA Medical Center, Saint Louis, Missouri, USA.,Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Christine T N Pham
- John Cochran VA Medical Center, Saint Louis, Missouri, USA. .,Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, Missouri, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
30
|
Hans CP, Sharma N, Sen S, Zeng S, Dev R, Jiang Y, Mahajan A, Joshi T. Transcriptomics Analysis Reveals New Insights into the Roles of Notch1 Signaling on Macrophage Polarization. Sci Rep 2019; 9:7999. [PMID: 31142802 PMCID: PMC6541629 DOI: 10.1038/s41598-019-44266-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/29/2019] [Indexed: 12/24/2022] Open
Abstract
Naïve macrophages (Mφ) polarize in response to various environmental cues to a spectrum of cells that have distinct biological functions. The extreme ends of the spectrum are classified as M1 and M2 macrophages. Previously, we demonstrated that Notch1 deficiency promotes Tgf-β2 dependent M2-polarization in a mouse model of abdominal aortic aneurysm. The present studies aimed to characterize the unique set of genes regulated by Notch1 signaling in macrophage polarization. Bone marrow derived macrophages isolated from WT or Notch1+/- mice (n = 12) were differentiated to Mφ, M1 or M2-phenotypes by 24 h exposure to vehicle, LPS/IFN-γ or IL4/IL13 respectively and total RNA was subjected to RNA-Sequencing (n = 3). Bioinformatics analyses demonstrated that Notch1 haploinsufficiency downregulated the expression of 262 genes at baseline level, 307 genes with LPS/IFN-γ and 254 genes with IL4/IL13 treatment. Among these, the most unique genes downregulated by Notch1 haploinsufficiency included fibromodulin (Fmod), caspase-4, Has1, Col1a1, Alpl and Igf. Pathway analysis demonstrated that extracellular matrix, macrophage polarization and osteogenesis were the major pathways affected by Notch1 haploinsufficiency. Gain and loss-of-function studies established a strong correlation between Notch1 haploinsufficiency and Fmod in regulating Tgf-β signaling. Collectively, our studies suggest that Notch1 haploinsufficiency increases M2 polarization through these newly identified genes.
Collapse
Affiliation(s)
- Chetan P Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA.
- Medical Pharmacology and Physiology, University of Missouri, Columbia, USA.
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA.
| | - Neekun Sharma
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA
| | - Sidharth Sen
- MU Informatics Institute, University of Missouri, Columbia, USA
| | - Shuai Zeng
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, USA
| | - Rishabh Dev
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA
| | - Yuexu Jiang
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, USA
| | - Advitiya Mahajan
- Department of Cardiovascular Medicine, University of Missouri, Columbia, USA
| | - Trupti Joshi
- MU Informatics Institute, University of Missouri, Columbia, USA
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, USA
- Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, USA
- Christopher S. Bond Life Science Center, University of Missouri, Columbia, USA
| |
Collapse
|
31
|
Balistreri CR, Forte M, Greco E, Paneni F, Cavarretta E, Frati G, Sciarretta S. An overview of the molecular mechanisms underlying development and progression of bicuspid aortic valve disease. J Mol Cell Cardiol 2019; 132:146-153. [PMID: 31103478 DOI: 10.1016/j.yjmcc.2019.05.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/14/2019] [Indexed: 12/31/2022]
Abstract
Bicuspid aortic valve (BAV) is a common congenital heart malformation frequently associated with the development of aortic valve diseases and severe aortopathy, such as aortic dilatation, aneurysm and dissection. To date, different genetic loci have been identified in syndromic and non- syndromic forms of BAV. Among these, genes involved in the regulation of extracellular matrix remodelling, epithelial to mesenchymal transition and nitric oxide metabolism appear to be the main contributors to BAV pathogenesis. However, no- single gene model explains BAV inheritance, suggesting that more factors are simultaneously involved. In this regard, characteristic epigenetic and immunological profiles have been documented to contradistinguish BAV individuals. In this review, we provide a comprehensive overview addressing molecular mechanisms involved in BAV development and progression.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy.
| | | | - Ernesto Greco
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiological, and Geriatric Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Switzerland; University Heart Center, Cardiology, University Hospital Zurich, Switzerland
| | - Elena Cavarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Naples, Italy
| | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, IS, Italy; Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
32
|
Abstract
Current management of aortic aneurysms relies exclusively on prophylactic operative repair of larger aneurysms. Great potential exists for successful medical therapy that halts or reduces aneurysm progression and hence alleviates or postpones the need for surgical repair. Preclinical studies in the context of abdominal aortic aneurysm identified hundreds of candidate strategies for stabilization, and data from preoperative clinical intervention studies show that interventions in the pathways of the activated inflammatory and proteolytic cascades in enlarging abdominal aortic aneurysm are feasible. Similarly, the concept of pharmaceutical aorta stabilization in Marfan syndrome is supported by a wealth of promising studies in the murine models of Marfan syndrome-related aortapathy. Although some clinical studies report successful medical stabilization of growing aortic aneurysms and aortic root stabilization in Marfan syndrome, these claims are not consistently confirmed in larger and controlled studies. Consequently, no medical therapy can be recommended for the stabilization of aortic aneurysms. The discrepancy between preclinical successes and clinical trial failures implies shortcomings in the available models of aneurysm disease and perhaps incomplete understanding of the pathological processes involved in later stages of aortic aneurysm progression. Preclinical models more reflective of human pathophysiology, identification of biomarkers to predict severity of disease progression, and improved design of clinical trials may more rapidly advance the opportunities in this important field.
Collapse
Affiliation(s)
- Jan H. Lindeman
- Dept. Vascular Surgery, Leiden University Medical Center, The Netherlands
| | - Jon S. Matsumura
- Division of Vascular Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
33
|
Harrison OJ, Torrens C, Salhiyyah K, Modi A, Moorjani N, Townsend PA, Ohri SK, Cagampang F. Defective NOTCH signalling drives smooth muscle cell death and differentiation in bicuspid aortic valve aortopathy. Eur J Cardiothorac Surg 2019; 56:117-125. [DOI: 10.1093/ejcts/ezy464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/02/2018] [Accepted: 12/10/2018] [Indexed: 01/05/2023] Open
Affiliation(s)
- Oliver J Harrison
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Cardiac Surgery, University Hospital Southampton, Southampton, UK
| | - Christopher Torrens
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Kareem Salhiyyah
- Department of Cardiac Surgery, University Hospital Southampton, Southampton, UK
| | | | - Narain Moorjani
- Department of Cardiac Surgery, Royal Papworth Hospital, University of Cambridge, Cambridge, UK
| | - Paul A Townsend
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, Manchester Cancer Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sunil K Ohri
- Department of Cardiac Surgery, University Hospital Southampton, Southampton, UK
| | - Felino Cagampang
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
34
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 682] [Impact Index Per Article: 97.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
35
|
Deregulation of Notch1 pathway and circulating endothelial progenitor cell (EPC) number in patients with bicuspid aortic valve with and without ascending aorta aneurysm. Sci Rep 2018; 8:13834. [PMID: 30218064 PMCID: PMC6138685 DOI: 10.1038/s41598-018-32170-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/30/2018] [Indexed: 12/29/2022] Open
Abstract
Bicuspid aortic valve (BAV) is frequently associated with the development of ascending aortic aneurysm, even if the underlying mechanisms remain to be clarified. Here, we investigated if a deregulation of Notch1 signaling pathway and endothelial progenitor cells (EPCs) number is associated with BAV disease and an early ascending aortic aneurysm (AAA) onset. For this purpose, 70 subjects with BAV (M/F 50/20; mean age: 58.8 ± 14.8 years) and 70 subjects with tricuspid aortic valve (TAV) (M/F 35/35; mean age: 69.1 ± 12.8 years) and AAA complicated or not, were included. Interestingly, patients with AAA showed a significant increase in circulating Notch1 levels and EPC number than subjects without AAA. However, circulating Notch1 levels and EPC number were significantly lower in BAV subjects than TAV patients either in the presence or absence of AAA. Finally, Notch pathway was activated to a greater extent in aortic aneurysmatic portions with respect to healthy aortic fragments in both BAV and TAV patients. However, the expression of genes encoding components and ligands of Notch pathway in aortic tissues was significantly lower in BAV than TAV subjects. Our study demonstrates that BAV subjects are characterized by a significant decrease in both tissue and circulating levels of Notch pathway, and in blood EPC number than TAV patients, either in presence or absence of AAA disease.
Collapse
|
36
|
Shen M, Hu M, Fedak PWM, Oudit GY, Kassiri Z. Cell-Specific Functions of ADAM17 Regulate the Progression of Thoracic Aortic Aneurysm. Circ Res 2018; 123:372-388. [PMID: 29930147 DOI: 10.1161/circresaha.118.313181] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/03/2018] [Accepted: 06/20/2018] [Indexed: 12/31/2022]
Abstract
RATIONALE ADAM17 (a disintegrin and metalloproteinase-17) is a membrane-bound enzyme that regulates bioavailability of multiple transmembrane proteins by proteolytic processing. ADAM17 has been linked to several pathologies, but its role in thoracic aortic aneurysm (TAA) has not been determined. OBJECTIVE The objective of this study was to explore the cell-specific functions of vascular ADAM17 in the pathogenesis and progression of TAA. METHODS AND RESULTS In aneurysmal thoracic aorta from patients, ADAM17 was increased in tunica media and intima. To determine the function of ADAM17 in the major cells types within these regions, we generated mice lacking ADAM17 in smooth muscle cells (SMC; Adam17f/f/Sm22Cre/+ ) or endothelial cells (Adam17f/f/Tie2Cre/+ ). ADAM17 deficiency in either cell type was sufficient to suppress TAA dilation markedly and adverse remodeling in males and females (in vivo) although through different mechanisms. ADAM17 deficiency in SMCs prevented the contractile-to-synthetic phenotypic switching in these cells after TAA induction, preventing perivascular fibrosis, inflammation, and adverse aortic remodeling. Loss of ADAM17 in endothelial cells protected the integrity of the intimal barrier by preserving the adherens junction (vascular endothelial-cadherin) and tight junctions (junctional adhesion molecule-A and claudin). In vitro studies on primary mouse thoracic SMCs and human primary aortic SMCs and endothelial cells (±ADAM17 small interfering RNA) confirmed the cell-specific functions of ADAM17 and demonstrated the cross-species validity of these findings. To determine the impact of ADAM17 inhibition in treating TAA, we used an ADAM17-selective inhibitor (PF-548) before or 3 days after TAA induction. In both cases, ADAM17 inhibition prevented progression of aneurysmal growth. CONCLUSIONS We have identified distinct cell-specific functions of ADAM17 in TAA progression, promoting pathological remodeling of SMC and impairing integrity of the intimal endothelial cell barrier. The dual impact of ADAM17 deficiency (or inhibition) in protecting 2 major cell types in the aortic wall highlights the unique position of this proteinase as a critical treatment target for TAA.
Collapse
Affiliation(s)
- Mengcheng Shen
- From the Department of Physiology (M.S., M.H., Z.K.).,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| | - Mei Hu
- From the Department of Physiology (M.S., M.H., Z.K.).,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| | - Paul W M Fedak
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada; Section of Cardiac Surgery, Department of Cardiac Sciences, University of Calgary, Libin Cardiovascular Institute of Alberta, Canada (P.W.M.F.).,Division of Cardiac Surgery, Bluhm Cardiovascular Institute, Northwestern Memorial Hospital, Chicago, IL (P.W.M.F.)
| | - Gavin Y Oudit
- Department of Medicine (G.Y.O.).,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| | - Zamaneh Kassiri
- From the Department of Physiology (M.S., M.H., Z.K.) .,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| |
Collapse
|
37
|
Roman ÁC, Carvajal-Gonzalez JM, Merino JM, Mulero-Navarro S, Fernández-Salguero PM. The aryl hydrocarbon receptor in the crossroad of signalling networks with therapeutic value. Pharmacol Ther 2017; 185:50-63. [PMID: 29258844 DOI: 10.1016/j.pharmthera.2017.12.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is well-known for its major contributions to the cellular responses against environmental toxins and carcinogens. Notably, AhR has also emerged as a key transcription factor controlling many physiological processes including cell proliferation and apoptosis, differentiation, adhesion and migration, pluripotency and stemness. These novel functions have broadened our understanding of the signalling pathways and molecular intermediates interacting with AhR under both homeostatic and pathological conditions. Recent discoveries link AhR with the function of essential organs such as liver, skin and gonads, and with complex organismal structures including the immune and cardiovascular systems. The identification of potential endogenous ligands able to regulate AhR activity, opens the possibility of designing ad hoc molecules with pharmacological and/or therapeutic value to treat human diseases in which AhR may have a causal role. Integration of experimental data from in vitro and in vivo studies with "omic" analyses of human patients affected with cancer, immune diseases, inflammation or neurological disorders will likely contribute to validate the clinical relevance of AhR and the possible benefits of modulating its activity by pharmacologically-driven strategies. In this review, we will highlight signalling pathways involved in human diseases that could be targetable by AhR modulators and discuss the feasibility of using such molecules in therapy. The pros and cons of AhR-aimed approaches will be also mentioned.
Collapse
Affiliation(s)
- Ángel C Roman
- Champalimaud Neuroscience Programme, Champalimoud Center for the Unknown, Lisbon, Portugal
| | - José M Carvajal-Gonzalez
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Jaime M Merino
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Sonia Mulero-Navarro
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
| | - Pedro M Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
| |
Collapse
|
38
|
van de Pol V, Kurakula K, DeRuiter MC, Goumans MJ. Thoracic Aortic Aneurysm Development in Patients with Bicuspid Aortic Valve: What Is the Role of Endothelial Cells? Front Physiol 2017; 8:938. [PMID: 29249976 PMCID: PMC5714935 DOI: 10.3389/fphys.2017.00938] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/06/2017] [Indexed: 12/28/2022] Open
Abstract
Bicuspid aortic valve (BAV) is the most common type of congenital cardiac malformation. Patients with a BAV have a predisposition for the development of thoracic aortic aneurysm (TAA). This pathological aortic dilation may result in aortic rupture, which is fatal in most cases. The abnormal aortic morphology of TAAs results from a complex series of events that alter the cellular structure and extracellular matrix (ECM) composition of the aortic wall. Because the major degeneration is located in the media of the aorta, most studies aim to unravel impaired smooth muscle cell (SMC) function in BAV TAA. However, recent studies suggest that endothelial cells play a key role in both the initiation and progression of TAAs by influencing the medial layer. Aortic endothelial cells are activated in BAV mediated TAAs and have a substantial influence on ECM composition and SMC phenotype, by secreting several key growth factors and matrix modulating enzymes. In recent years there have been significant advances in the genetic and molecular understanding of endothelial cells in BAV associated TAAs. In this review, the involvement of the endothelial cells in BAV TAA pathogenesis is discussed. Endothelial cell functioning in vessel homeostasis, flow response and signaling will be highlighted to give an overview of the importance and the under investigated potential of endothelial cells in BAV-associated TAA.
Collapse
Affiliation(s)
- Vera van de Pol
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Kondababu Kurakula
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Marco C. DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
39
|
Bai H, Lee JS, Hu H, Wang T, Isaji T, Liu S, Guo J, Liu H, Wolf K, Ono S, Guo X, Yatsula B, Xing Y, Fahmy TM, Dardik A. Transforming Growth Factor-β1 Inhibits Pseudoaneurysm Formation After Aortic Patch Angioplasty. Arterioscler Thromb Vasc Biol 2017; 38:195-205. [PMID: 29146747 DOI: 10.1161/atvbaha.117.310372] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Pseudoaneurysms remain a significant complication after vascular procedures. We hypothesized that TGF-β (transforming growth factor-β) signaling plays a mechanistic role in the development of pseudoaneurysms. APPROACH AND RESULTS Rat aortic pericardial patch angioplasty was associated with a high incidence (88%) of pseudoaneurysms at 30 days, with increased smad2 phosphorylation in small pseudoaneurysms but not in large pseudoaneurysms; TGF-β1 receptors were increased in small pseudoaneurysms and preserved in large pseudoaneurysms. Delivery of TGF-β1 via nanoparticles covalently bonded to the patch stimulated smad2 phosphorylation both in vitro and in vivo and significantly decreased pseudoaneurysm formation (6.7%). Inhibition of TGF-β1 signaling with SB431542 decreased smad2 phosphorylation both in vitro and in vivo and significantly induced pseudoaneurysm formation by day 7 (66.7%). CONCLUSIONS Normal healing after aortic patch angioplasty is associated with increased TGF-β1 signaling, and recruitment of smad2 signaling may limit pseudoaneurysm formation; loss of TGF-β1 signaling is associated with the formation of large pseudoaneurysms. Enhancement of TGF-β1 signaling may be a potential mechanism to limit pseudoaneurysm formation after vascular intervention.
Collapse
Affiliation(s)
- Hualong Bai
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Jung Seok Lee
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Haidi Hu
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Tun Wang
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Toshihiko Isaji
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Shirley Liu
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Jianming Guo
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Haiyang Liu
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Katharine Wolf
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Shun Ono
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Xiangjiang Guo
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Bogdan Yatsula
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Ying Xing
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Tarek M Fahmy
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.)
| | - Alan Dardik
- From the Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China (H.B.); Basic Medical College of Zhengzhou University, Henan, China (H.B., Y.X.); Vascular Biology and Therapeutics Program (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), Department of Surgery (H.B., H.H., T.W., T.I., S.L., J.G., H.L., K.W., S.O., X.G., B.Y., A.D.), and Department of Immunobiology (T.M.F.), Yale University School of Medicine, New Haven, CT; Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.); and Department of Surgery, VA Connecticut Healthcare System, West Haven, CT (A.D.).
| |
Collapse
|
40
|
Sachdeva J, Mahajan A, Cheng J, Baeten JT, Lilly B, Kuivaniemi H, Hans CP. Smooth muscle cell-specific Notch1 haploinsufficiency restricts the progression of abdominal aortic aneurysm by modulating CTGF expression. PLoS One 2017; 12:e0178538. [PMID: 28562688 PMCID: PMC5451061 DOI: 10.1371/journal.pone.0178538] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 05/15/2017] [Indexed: 02/06/2023] Open
Abstract
Aims Infiltration of macrophages and apoptosis of vascular smooth muscle cells (VSMCs) promote the development of abdominal aortic aneurysm (AAA). Previously, we demonstrated that global Notch1 deficiency prevents the formation of AAA in a mouse model. Herein, we sought to explore the cell-specific roles of Notch1 in AAA development. Methods and results Cell-specific Notch1 haploinsufficient mice, generated on Apoe-/- background using Cre-lox technology, were infused with angiotensin II (1000 ng/min/kg) for 28 days. Notch1 haploinsufficiency in myeloid cells (n = 9) prevented the formation of AAA attributed to decreased inflammation. Haploinsufficiency of Notch1 in SMCs (n = 14) per se did not prevent AAA formation, but histoarchitectural traits of AAA including elastin degradation and aortic remodeling, were minimal in SMC-Notch1+/-;Apoe-/- mice compared to Apoe-/- mice (n = 33). Increased immunostaining of the contractile SMC-phenotype markers and concomitant decreased expression of synthetic SMC-phenotype markers were observed in the aortae of SMC-Notch1+/-;Apoe-/- mice. Expression of connective tissue growth factor (CTGF), a matrix-associated protein that modulates the synthetic VSMC phenotype, increased in the abdominal aorta of Apoe-/- mice and in the adventitial region of the abdominal aorta in human AAA. Notch1 haploinsufficiency decreased the expression of Ctgf in the aorta and in vitro cell culture system. In vitro studies on SMCs using the Notch1 intracellular domain (NICD) plasmid, dominant negative mastermind-like (dnMAML), or specific siRNA suggest that Notch1, not Notch3, directly modulates the expression of CTGF. Conclusions Our data suggest that lack of Notch1 in SMCs limits dilation of the abdominal aorta by maintaining contractile SMC-phenotype and preventing matrix-remodeling.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Cells, Cultured
- Coculture Techniques
- Connective Tissue Growth Factor/metabolism
- Haploinsufficiency
- Matrix Metalloproteinases/biosynthesis
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Receptor, Notch1/metabolism
Collapse
Affiliation(s)
| | - Advitiya Mahajan
- Cardiology, Medical Pharmacology & Physiology and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Jeeyun Cheng
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Jeremy T. Baeten
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Brenda Lilly
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Chetan P. Hans
- Ohio State University, Columbus, Ohio, United States of America
- Cardiology, Medical Pharmacology & Physiology and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
41
|
张 家, 陈 坤, 张 福, 李 少, 吴 源, 冯 靖, 王 武, 闫 玉. [Establishment of a rabbit model of small diameter vascular graft replacement]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:687-692. [PMID: 28539296 PMCID: PMC6780461 DOI: 10.3969/j.issn.1673-4254.2017.05.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To establish an rabbit model that mimics the hemodynamics of the bypass graft after coronary artery bypass surgery. METHODS Sixteen New Zealand rabbits were randomly divided into two groups for abdominal aortic artery replacement using a 3-cm-long ePTFE graft with an inner diameter 4 mm through an incision at 1/3 from the middle to the lower part of the abdomen (group A) or in the lower abdomen (group B). The general conditions of the rabbits, operative time, number of collateral vessels that needed to be ligated, rate of massive intraoperative bleeding, fluctuation of vascular anastomosis after surgery, patency rate of the graft on day 7 after the operation were compared between the two groups. RESULTS The two groups of rabbits had similar body weight, diameter of the abdominal aortic artery, intraoperative bleeding rate and occlusion rate of the vascular graft at 7 days after the procedure. The operative time was longer in group A, but the difference was not statistically significant. In group A, the number of the vascular branches that needed to be ligated was smaller and the rate normal femoral artery pulsation was higher than those in group B. CONCLUSION It is feasible to establish models of small diameter vascular graft replacement in rabbits, and the patency rate of the graft can be monitored by observation of the general condition and ultrasound examination of the rabbits.
Collapse
Affiliation(s)
- 家庆 张
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 坤棠 陈
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 福伟 张
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 少彬 李
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 源周 吴
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 靖 冯
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 武军 王
- 南方医科大学南方医院胸心外科,广东 广州 510515Department of Cardiothoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 玉生 闫
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
42
|
AT1 receptor signaling pathways in the cardiovascular system. Pharmacol Res 2017; 125:4-13. [PMID: 28527699 DOI: 10.1016/j.phrs.2017.05.008] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 01/14/2023]
Abstract
The importance of the renin angiotensin aldosterone system in cardiovascular physiology and pathophysiology has been well described whereas the detailed molecular mechanisms remain elusive. The angiotensin II type 1 receptor (AT1 receptor) is one of the key players in the renin angiotensin aldosterone system. The AT1 receptor promotes various intracellular signaling pathways resulting in hypertension, endothelial dysfunction, vascular remodeling and end organ damage. Accumulating evidence shows the complex picture of AT1 receptor-mediated signaling; AT1 receptor-mediated heterotrimeric G protein-dependent signaling, transactivation of growth factor receptors, NADPH oxidase and ROS signaling, G protein-independent signaling, including the β-arrestin signals and interaction with several AT1 receptor interacting proteins. In addition, there is functional cross-talk between the AT1 receptor signaling pathway and other signaling pathways. In this review, we will summarize an up to date overview of essential AT1 receptor signaling events and their functional significances in the cardiovascular system.
Collapse
|
43
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
44
|
Wilson NK, Gould RA, Gallo MacFarlane E, Consortium ML. Pathophysiology of aortic aneurysm: insights from human genetics and mouse models. Pharmacogenomics 2016; 17:2071-2080. [PMID: 27922338 DOI: 10.2217/pgs-2016-0127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aneurysms are local dilations of an artery that predispose the vessel to sudden rupture. They are often asymptomatic and undiagnosed, resulting in a high mortality rate. The predisposition to develop thoracic aortic aneurysms is often genetically inherited and associated with syndromes affecting connective tissue homeostasis. This review discusses how elucidation of the genetic causes of syndromic forms of thoracic aortic aneurysm has helped identify pathways that contribute to disease progression, including those activated by TGF-β, angiotensin II and Notch ligands. We also discuss how pharmacological manipulation of these signaling pathways has provided further insight into the mechanism of disease and identified compounds with therapeutic potential in these and related disorders.
Collapse
Affiliation(s)
- Nicole K Wilson
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Russell A Gould
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Miller Research Building 532, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | | |
Collapse
|
45
|
An TH, He QW, Xia YP, Chen SC, Baral S, Mao L, Jin HJ, Li YN, Wang MD, Chen JG, Zhu LQ, Hu B. MiR-181b Antagonizes Atherosclerotic Plaque Vulnerability Through Modulating Macrophage Polarization by Directly Targeting Notch1. Mol Neurobiol 2016; 54:6329-6341. [PMID: 27722924 DOI: 10.1007/s12035-016-0163-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022]
Abstract
Atherosclerotic plaque vulnerability is the major cause for acute stroke and could be regulated by macrophage polarization. MicroRNA-181b (miR-181b) was involved in macrophage differential. Here, we explore whether miR-181b could regulate atherosclerotic plaque vulnerability by modulating macrophage polarization and the underline mechanisms. In acute stroke patients with atherosclerotic plaque, we found that the serum level of miR-181b was decreased. Eight-week apolipoprotein E knockout (ApoE-/-) mice were randomly divided into three groups (N = 10): mice fed with normal saline (Ctrl), mice fed with high-fat diet, and tail vein injection with miRNA agomir negative control (AG-NC)/miR-181b agomir (181b-AG, a synthetic miR-181b agonist). We found that the serum level of miR-181b in AG-NC group was lower than that in Ctrl group. Moreover, 181b-AG could upregulate miR-181b expression, reduce artery burden and attenuate atherosclerotic plaque vulnerability by modulating macrophage polarization. In RAW264.7 cells treated with oxidized low-density lipoprotein (ox-LDL), we found miR-181b could reverse the function of ox-LDL on M1/M2 markers at both mRNA and protein levels. Furthermore, by employing luciferase reporter assay, we found that Notch1 was a direct target of miR-181b and could be regulated by miR-181b in vivo and in vitro. Finally, inhibition of Notch1 could abolish the function of downregulating miR-181b on increasing M2 phenotype macrophages. Our study demonstrates that administration of miR-181b could reduce atherosclerotic plaque vulnerability partially through modulating macrophage phenotype by directly targeting Notch1.
Collapse
Affiliation(s)
- Tian-Hui An
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Quan-Wei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sheng-Cai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Suraj Baral
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui-Juan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ya-Nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Meng-Die Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jian-Guo Chen
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling-Qiang Zhu
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Abdominal aortic aneurysm (AAA) is a pathological condition of permanent dilation that portends the potentially fatal consequence of aortic rupture. This review emphasizes recent advances in mechanistic insight into aneurysm pathogenesis and potential pharmacologic therapies that are on the horizon for AAAs. RECENT FINDINGS An increasing body of evidence demonstrates that genetic factors, including 3p12.3, DAB2IP, LDLr, LRP1, matrix metalloproteinase (MMP)-3, TGFBR2, and SORT1 loci, are associated with AAA development. Current human studies and animal models have shown that many leukocytes and inflammatory mediators, such as IL-1, IL-17, TGF-β, and angiotensin II, are involved in the pathogenesis of AAAs. Leukocytic infiltration into aortic media leads to smooth muscle cell depletion, generation of reactive oxygen species, and extracellular matrix fragmentation. Preclinical investigations into pharmacological therapies for AAAs have provided intriguing insight into the roles of microRNAs in regulating many pathological pathways in AAA development. Several large clinical trials are ongoing, seeking to translate preclinical findings into therapeutic options. SUMMARY Recent studies have identified many potential mechanisms involved in AAA pathogenesis that provide insight into the development of a medical treatment for this disease.
Collapse
|
47
|
Balistreri CR, Madonna R, Melino G, Caruso C. The emerging role of Notch pathway in ageing: Focus on the related mechanisms in age-related diseases. Ageing Res Rev 2016; 29:50-65. [PMID: 27328278 DOI: 10.1016/j.arr.2016.06.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/10/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022]
Abstract
Notch signaling is an evolutionarily conserved pathway, which is fundamental for the development of all tissues, organs and systems of human body. Recently, a considerable and still growing number of studies have highlighted the contribution of Notch signaling in various pathological processes of the adult life, such as age-related diseases. In particular, the Notch pathway has emerged as major player in the maintenance of tissue specific homeostasis, through the control of proliferation, migration, phenotypes and functions of tissue cells, as well as in the cross-talk between inflammatory cells and the innate immune system, and in onset of inflammatory age-related diseases. However, until now there is a confounding evidence about the related mechanisms. Here, we discuss mechanisms through which Notch signaling acts in a very complex network of pathways, where it seems to have the crucial role of hub. Thus, we stress the possibility to use Notch pathway, the related molecules and pathways constituting this network, both as innovative (predictive, diagnostic and prognostic) biomarkers and targets for personalised treatments for age-related diseases.
Collapse
|
48
|
Yin J, Hu H, Li X, Xue M, Cheng W, Wang Y, Xuan Y, Li X, Yang N, Shi Y, Yan S. Inhibition of Notch signaling pathway attenuates sympathetic hyperinnervation together with the augmentation of M2 macrophages in rats post-myocardial infarction. Am J Physiol Cell Physiol 2015; 310:C41-53. [PMID: 26491050 DOI: 10.1152/ajpcell.00163.2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/13/2015] [Indexed: 02/05/2023]
Abstract
Inflammation-dominated sympathetic sprouting adjacent to the necrotic region following myocardial infarction (MI) has been implicated in the etiology of arrhythmias resulting in sudden cardiac death; however, the mechanisms responsible remain to be elucidated. Although being a key immune mediator, the role of Notch has yet to be explored. We investigated whether Notch regulates macrophage responses to inflammation and affects cardiac sympathetic reinnervation in rats undergoing MI. MI was induced by coronary artery ligation. A high level of Notch intracellular domain was observed in the macrophages that infiltrated the infarct area at 3 days post-MI. The administration of the Notch inhibitor N-N-(3,5-difluorophenacetyl-L-alanyl)-S-phenylglycine-t-butyl ester (DAPT) (intravenously 30 min before MI and then daily until death) decreased the number of macrophages and significantly increased the M2 macrophage activation profile in the early stages and attenuated the expression of nerve growth factor (NGF). Eventually, NGF-induced sympathetic hyperinnervation was blunted, as assessed by the immunofluorescence of tyrosine hydroxylase. At 7 days post-MI, the arrhythmia score of programmed electric stimulation in the vehicle-treated infarcted rats was higher than that in rats treated with DAPT. Further deterioration in cardiac function and decreases in the plasma levels of TNF-α and IL-1β were also detected. In vitro studies revealed that LPS/IFN-γ upregulated the surface expression of NGF in M1 macrophages in a Notch-dependent manner. We concluded that Notch inhibition during the acute inflammatory response phase is associated with the downregulation of NGF, probably through a macrophage-dependent pathway, thus preventing the process of sympathetic hyperinnervation.
Collapse
Affiliation(s)
- Jie Yin
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Hesheng Hu
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xiaolu Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Mei Xue
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Wenjuan Cheng
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Ye Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yongli Xuan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xinran Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Na Yang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yugen Shi
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Suhua Yan
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
49
|
González MJ, Ruiz-García A, Monsalve EM, Sánchez-Prieto R, Laborda J, Díaz-Guerra MJM, Ruiz-Hidalgo MJ. DLK1 is a novel inflammatory inhibitor which interferes with NOTCH1 signaling in TLR-activated murine macrophages. Eur J Immunol 2015; 45:2615-27. [PMID: 26115479 DOI: 10.1002/eji.201545514] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/19/2015] [Accepted: 06/23/2015] [Indexed: 12/19/2022]
Abstract
Delta-like protein 1 (DLK1) is a noncanonical ligand that inhibits NOTCH1 receptor activity and regulates multiple differentiation processes. In macrophages, NOTCH signaling increases TLR-induced expression of key pro-inflammatory mediators. We have investigated the role of DLK1 in macrophage activation and inflammation using Dlk1-deficient mice and Raw 264.7 cells overexpressing Dlk1. In the absence of Dlk1, NOTCH1 expression is increased and the activation of macrophages with TLR3 or TLR4 agonists leads to higher production of IFN-β and other pro-inflammatory cytokines, including TNF-α, IL-12, and IL-23. The expression of key proteins involved in IFN-β signaling, such as IRF3, IRF7, IRF1, or STAT1, as well as cRel, or RelB, which are responsible for the generation of IL-12 and IL-23, is enhanced in Dlk1 KO macrophages. Consistently, Dlk1 KO mice are more sensitive to LPS-induced endotoxic shock. These effects seem to be mediated through the modulation of NOTCH1 signaling. TLR4 activation reduces DLK1 expression, whereas increases NOTCH1 levels. In addition, DLK1 expression diminishes during differentiation of human U937 cells to macrophages. Overall, these results reveal a novel role for DLK1 as a regulator of NOTCH-mediated, pro-inflammatory macrophage activation, which could help to ensure a baseline level preventing constant tissue inflammation.
Collapse
Affiliation(s)
- María J González
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Almudena Ruiz-García
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Eva M Monsalve
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Ricardo Sánchez-Prieto
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Jorge Laborda
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - María J M Díaz-Guerra
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - María J Ruiz-Hidalgo
- Facultad de Medicina, Centro Regional de Investigaciones Biomédicas (CRIB), Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| |
Collapse
|