101
|
Cardiovascular disease in autoimmune rheumatic diseases. Autoimmun Rev 2013; 12:1004-15. [PMID: 23541482 DOI: 10.1016/j.autrev.2013.03.013] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 03/07/2013] [Indexed: 12/18/2022]
Abstract
Various autoimmune rheumatic diseases (ARDs), including rheumatoid arthritis, spondyloarthritis, vasculitis and systemic lupus erythematosus, are associated with premature atherosclerosis. However, premature atherosclerosis has not been uniformly observed in systemic sclerosis. Furthermore, although experimental models of atherosclerosis support the role of antiphospholipid antibodies in atherosclerosis, there is no clear evidence of premature atherosclerosis in antiphospholipid syndrome (APA). Ischemic events in APA are more likely to be caused by pro-thrombotic state than by enhanced atherosclerosis. Cardiovascular disease (CVD) in ARDs is caused by traditional and non-traditional risk factors. Besides other factors, inflammation and immunologic abnormalities, the quantity and quality of lipoproteins, hypertension, insulin resistance/hyperglycemia, obesity and underweight, presence of platelets bearing complement protein C4d, reduced number and function of endothelial progenitor cells, apoptosis of endothelial cells, epigenetic mechanisms, renal disease, periodontal disease, depression, hyperuricemia, hypothyroidism, sleep apnea and vitamin D deficiency may contribute to the premature CVD. Although most research has focused on systemic inflammation, vascular inflammation may play a crucial role in the premature CVD in ARDs. It may be involved in the development and destabilization of both atherosclerotic lesions and of aortic aneurysms (a known complication of ARDs). Inflammation in subintimal vascular and perivascular layers appears to frequently occur in CVD, with a higher frequency in ARD than in non-ARD patients. It is possible that this inflammation is caused by infections and/or autoimmunity, which might have consequences for treatment. Importantly, drugs targeting immunologic factors participating in the subintimal inflammation (e.g., T- and B-cells) might have a protective effect on CVD. Interestingly, vasa vasorum and cardiovascular adipose tissue may play an important role in atherogenesis. Inflammation and complement depositions in the vessel wall are likely to contribute to vascular stiffness. Based on biopsy findings, also inflammation in the myocardium and small vessels may contribute to premature CVD in ARDs (cardiac ischemia and heart failure). There is an enormous need for an improved CVD prevention in ARDs. Studies examining the effect of DMARDs/biologics on vascular inflammation and CV risk are warranted.
Collapse
|
102
|
Tanaka H, Zaima N, Sasaki T, Hayasaka T, Goto-Inoue N, Onoue K, Ikegami K, Morita Y, Yamamoto N, Mano Y, Sano M, Saito T, Sato K, Konno H, Setou M, Unno N. Adventitial vasa vasorum arteriosclerosis in abdominal aortic aneurysm. PLoS One 2013; 8:e57398. [PMID: 23460850 PMCID: PMC3583902 DOI: 10.1371/journal.pone.0057398] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 01/21/2013] [Indexed: 12/03/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a common disease among elderly individuals. However, the precise pathophysiology of AAA remains unknown. In AAA, an intraluminal thrombus prevents luminal perfusion of oxygen, allowing only the adventitial vaso vasorum (VV) to deliver oxygen and nutrients to the aortic wall. In this study, we examined changes in the adventitial VV wall in AAA to clarify the histopathological mechanisms underlying AAA. We found marked intimal hyperplasia of the adventitial VV in the AAA sac; further, immunohistological studies revealed proliferation of smooth muscle cells, which caused luminal stenosis of the VV. We also found decreased HemeB signals in the aortic wall of the sac as compared with those in the aortic wall of the neck region in AAA. The stenosis of adventitial VV in the AAA sac and the malperfusion of the aortic wall observed in the present study are new aspects of AAA pathology that are expected to enhance our understanding of this disease.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Nobuhiro Zaima
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Applied Biological Chemistry, Kinki University, Higashiosaka City, Japan
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takahiro Hayasaka
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoko Goto-Inoue
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kenji Onoue
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Koji Ikegami
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoshifumi Morita
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoto Yamamoto
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuuki Mano
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masaki Sano
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takaaki Saito
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kohji Sato
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroyuki Konno
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsutoshi Setou
- Department of Cell Biology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- * E-mail: (NU); (MS)
| | - Naoki Unno
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
- * E-mail: (NU); (MS)
| |
Collapse
|
103
|
Toba H, Wang J, Ohigashi M, Kobara M, Nakata T. Telmisartan Protects against Vascular Dysfunction with Peroxisome Proliferator-Activated Receptor-γ Activation in Hypertensive 5/6 Nephrectomized Rats. Pharmacology 2013; 92:265-75. [DOI: 10.1159/000355482] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/06/2013] [Indexed: 11/19/2022]
|
104
|
Wang F, Zhao XQ, Liu JN, Wang ZH, Wang XL, Hou XY, Liu R, Gao F, Zhang MX, Zhang Y, Bu PL. Antagonist of microRNA-21 improves balloon injury-induced rat iliac artery remodeling by regulating proliferation and apoptosis of adventitial fibroblasts and myofibroblasts. J Cell Biochem 2012; 113:2989-3001. [PMID: 22565856 DOI: 10.1002/jcb.24176] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Molecular pathways involved in adventitial fibroblasts (AFs) and myofibroblasts (MFs) proliferation and apoptosis contribute to vascular remodeling. MicroRNA-21 (miR-21) plays an important role in regulating cellular proliferation and apoptosis of many cell types; however, the effect of miR-21 on AFs and MFs is still unknown. In this study, we found that miR-21 was expressed in AFs and overexpressed in MFs. Inhibition of miR-21 decreased proliferation and increased apoptosis of AFs and MFs, and overexpression of miR-21 with pre-miR-21 had the reverse effect. Programmed cell death 4 (PDCD4), related to cell proliferation and apoptosis, was validated as a direct target of miR-21 by dual-luciferase reporter assay and gain and loss of function of miR-21 in AFs and MFs. PDCD4 knockdown with siRNA partly rescued the reduced proliferation with miR-21 inhibition and alleviated the increased apoptosis induced by miR-21 inhibition in AFs and MFs. Moreover, increasing PDCD4 expression by miR-21 inhibition significantly decreased JNK/c-Jun activity. In contrast, decreasing PDCD4 expression by pre-miR-21 treatment increased JNK/c-Jun activity, while the effect of miR-21 inhibition on JNK/c-Jun activity could be rescued by PDCD4 siRNA. Moreover, miR-21 inhibition could regulate proliferation and apoptosis of vascular AFs and MFs in vivo. Furthermore, miR-21 inhibition reversed vascular remodeling induced by balloon injury. In summary, our findings demonstrate that miR-21 may have a critical role in regulating proliferation and apoptosis of AFs and MFs, and PDCD4 is a functional target gene involved in the miR-21-mediated cellular effects in vascular remodeling by a miR-21/PDCD4/JNK/c-Jun pathway.
Collapse
Affiliation(s)
- Fei Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Shandong University Qilu Hospital, Jinan, Shandong Province, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Laaksamo E, Tulamo R, Liiman A, Baumann M, Friedlander RM, Hernesniemi J, Kangasniemi M, Niemelä M, Laakso A, Frösen J. Oxidative Stress Is Associated With Cell Death, Wall Degradation, and Increased Risk of Rupture of the Intracranial Aneurysm Wall. Neurosurgery 2012; 72:109-17. [DOI: 10.1227/neu.0b013e3182770e8c] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract
BACKGROUND:
The cause of rupture of intracranial aneurysms (IA) is not well understood. We previously demonstrated that loss of cells from the IA wall is associated with wall degeneration and rupture.
OBJECTIVE:
To investigate the mechanisms mediating cell death in the IA wall.
METHODS:
Snap-frozen tissue samples from aneurysm fundi were studied with terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining and immunostaining (14 unruptured and 20 ruptured), as well as with Western blot (12 unruptured and 12 ruptured).
RESULTS:
Ruptured IA walls had more TUNEL-positive cells than unruptured walls (P < .001). Few cells positive for cleaved caspase-3 were detected. Cleaved caspase-9 (intrinsic activation of apoptosis) was significantly increased in ruptured IA walls, whereas cleaved caspase-8 (extrinsic activation of apoptosis) was not detected. Increased expression of hemeoxygenase-1, a marker for oxidative stress, was associated with IA wall degeneration and rupture.
CONCLUSION:
Our results show that programmed cell death is activated in the IA wall via the intrinsic pathway. High oxidative stress in the IA wall is probably a significant cause of the intrinsic activation of cell death.
Collapse
Affiliation(s)
| | | | - Arto Liiman
- Neurosurgery Research Group, Biomedicum Helsinki,
| | - Marc Baumann
- Protein Chemistry/Proteomics Laboratory, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Robert M. Friedlander
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Neuroapoptosis Laboratory, Harvard Medical School, Boston, Massachusetts
| | - Juha Hernesniemi
- Neurosurgery Research Group, Biomedicum Helsinki,
- Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Marko Kangasniemi
- Neurosurgery Research Group, Biomedicum Helsinki,
- Department of Radiology, University of Helsinki and HUS Radiology (Medical Imaging Center), Helsinki, Finland
| | - Mika Niemelä
- Neurosurgery Research Group, Biomedicum Helsinki,
- Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Aki Laakso
- Neurosurgery Research Group, Biomedicum Helsinki,
- Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Juhana Frösen
- Neurosurgery Research Group, Biomedicum Helsinki,
- Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
- Neuroapoptosis Laboratory, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
106
|
Michel JB, Delbosc S, Ho-Tin-Noé B, Leseche G, Nicoletti A, Meilhac O, Martin-Ventura JL. From intraplaque haemorrhages to plaque vulnerability. J Cardiovasc Med (Hagerstown) 2012; 13:628-34. [DOI: 10.2459/jcm.0b013e328357face] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
107
|
Li Y, Tao J, Zhang J, Tian X, Liu S, Sun M, Zhang X, Yan C, Han Y. Cellular repressor E1A-stimulated genes controls phenotypic switching of adventitial fibroblasts by blocking p38MAPK activation. Atherosclerosis 2012; 225:304-14. [PMID: 23040447 DOI: 10.1016/j.atherosclerosis.2012.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 08/02/2012] [Accepted: 08/13/2012] [Indexed: 11/16/2022]
Abstract
AIMS Phenotypic modulation of adventitial fibroblasts (AFs) plays an important role in the pathogenesis of proliferative vascular diseases. The current study aimed to identify the role of cellular repressor E1A-stimulated genes (CREG), a critical mediator in the maintenance of vascular homeostasis, in AF phenotypic modulation and adventitial remodeling. METHOD AND RESULTS Using in situ double-immunofluorescence staining, we ascertained that CREG expression was significantly down-regulated in the adventitia after vascular injury, and its expression pattern was conversely correlated with the expression of smooth muscle α-actin (α-SMA), a marker for differentiation of AFs into myofibroblasts. In vitro data confirmed the association of CREG in angiotensin II (Ang II)-induced AF differentiation. Additionally, overexpression of CREG attenuated Ang II-induced α-SMA expression in AFs. CREGoverexpressing AFs showed decreased levels of proliferation on days 2-5 following stimulation by Ang II compared with controls, with changes in the cell cycle profile as shown by BrdU incorporation assay and fluorescence activated cell sorting analysis. Moreover, wound healing assay and transwell migration model demonstrated that upregulation of CREG expression inhibited Ang II-induced AF migration. We found that CREG-mediated its counterbalancing effects in Ang II-induced phenotypic modulation, proliferation and migration by inhibition of the p38MAPK signaling pathway, validated by pharmacological blockade of p38MAPK with SB 203580 and by overexpression of p38MAPK with transfectants expressing constitutively active p38αMAPK. CONCLUSION Our findings suggest that CREG is a novel AF phenotypic modulator in a p38MAPK-dependent manner. Modulating CREG on the local vascular wall may become a new therapeutic target against proliferative vascular diseases.
Collapse
Affiliation(s)
- Yang Li
- Graduate School of Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Li XD, Chen J, Ruan CC, Zhu DL, Gao PJ. Vascular endothelial growth factor-induced osteopontin expression mediates vascular inflammation and neointima formation via Flt-1 in adventitial fibroblasts. Arterioscler Thromb Vasc Biol 2012; 32:2250-8. [PMID: 22814749 DOI: 10.1161/atvbaha.112.255216] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Adventitia acts as an active participant in vascular inflammation but the precise mechanism underlying adventitia-mediated vascular inflammation is not fully understood. In this study, we sought to determine whether vascular endothelial growth factor (VEGF) regulates osteopontin (OPN) expression through Flt-1 in adventitial fibroblasts (AFs) to mediate vascular inflammation and neointima formation. METHODS AND RESULTS In primary cultured AFs, VEGF increased intracellular and secreted OPN expression in a time- and dose-dependent manner, which was effectively suppressed by a specific anti-Flt-1 hexapeptide. Interestingly, VEGF treatment of AFs enhanced the capability of AF-conditioned medium to stimulate macrophages chemotaxis, and this effect was attenuated after blockade of OPN from AF-conditioned medium. Furthermore, perivascular delivery of anti-Flt-1 peptide preferentially concentrated in the adventitia resulted in a decrease of neointima formation after balloon injury in carotid arteries. The inhibition of neointima formation was preceded by significant reduction of VEGF and OPN expression with concurrent macrophage infiltration into adventitia after injury. Activation of extracellular signal-regulated kinase 1/2 pathway was involved in OPN upregulation and macrophage chemotaxis. CONCLUSIONS These results demonstrate that VEGF/Flt-1 signaling plays a significant role in vascular inflammation and neointima formation by regulating OPN expression in AFs and provide insight into Flt-1 as a potential therapeutic target for vascular diseases.
Collapse
Affiliation(s)
- Xiao-Dong Li
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
109
|
Weih F, Gräbner R, Hu D, Beer M, Habenicht AJR. Control of dichotomic innate and adaptive immune responses by artery tertiary lymphoid organs in atherosclerosis. Front Physiol 2012; 3:226. [PMID: 22783198 PMCID: PMC3390894 DOI: 10.3389/fphys.2012.00226] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 06/04/2012] [Indexed: 12/29/2022] Open
Abstract
Tertiary lymphoid organs (TLOs) emerge in tissues in response to non-resolving inflammation such as chronic infection, graft rejection, and autoimmune disease. We identified artery TLOs (ATLOs) in the adventitia adjacent to atherosclerotic plaques of aged hyperlipidemic ApoE−/− mice. ATLOs are structured into T cell areas harboring conventional dendritic cells and monocyte-derived DCs; B cell follicles containing follicular dendritic cells within activated germinal centers; and peripheral niches of plasma cells. ATLOs also show extensive neoangiogenesis, aberrant lymphangiogenesis, and high endothelial venule (HEV) neogenesis. Newly formed conduit networks connect the external lamina of the artery with HEVs in T cell areas. ATLOs recruit and generate lymphocyte subsets with opposing activities including activated CD4+ and CD8+ effector T cells, natural and induced CD4+ T regulatory (nTregs; iTregs) cells as well as B-1 and B-2 cells at different stages of differentiation. These data indicate that ATLOs organize dichotomic innate and adaptive immune responses in atherosclerosis. In this review we discuss the novel concept that dichotomic immune responses toward atherosclerosis-specific antigens are carried out by ATLOs in the adventitia of the arterial wall and that malfunction of the tolerogenic arm of ATLO immunity triggers transition from silent autoimmune reactivity to clinically overt disease.
Collapse
Affiliation(s)
- Falk Weih
- Leibniz-Institute for Age Research, Fritz-Lipmann-Institute Jena, Germany
| | | | | | | | | |
Collapse
|
110
|
Sarda-Mantel L, Alsac JM, Boisgard R, Hervatin F, Montravers F, Tavitian B, Michel JB, Le Guludec D. Comparison of 18F-fluoro-deoxy-glucose, 18F-fluoro-methyl-choline, and 18F-DPA714 for positron-emission tomography imaging of leukocyte accumulation in the aortic wall of experimental abdominal aneurysms. J Vasc Surg 2012; 56:765-73. [PMID: 22726755 DOI: 10.1016/j.jvs.2012.01.069] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/20/2012] [Accepted: 01/29/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a frequent form of atherothrombotic disease, whose natural history is to enlarge and rupture. Indicators other than AAA diameter would be useful for preventive surgery decision-making, including positron-emission tomography (PET) methods permitting visualization of aortic wall leukocyte activation relevant to prognostic AAA evaluation. In this study, we compare three PET tracers of activated leukocytes, 18F-fluoro-deoxy-glucose (FDG), 18F-fluoro-methyl-choline (FCH), and 18F-DPA714 (a peripheral benzodiazepine receptor antagonist) for in vivo PET quantification of aortic wall inflammation in rat experimental AAAs, in correlation with histopathological studies of lesions. METHODS AAAs were induced by orthotopic implantation of decellularized guinea pig abdominal aorta in 46 Lewis rats. FDG-PET (n = 20), FCH-PET (n = 8), or both (n = 12) were performed 2 weeks to 4 months after the graft, 1 hour after tracer injection (30 MBq). Six rats (one of which had FDG-PET) underwent 18F-DPA714-PET. Rats were sacrificed after imaging; AAAs and normal thoracic aortas were cut into axial sections for quantitative autoradiography and histologic studies, including ED1 (macrophages) and CD8 T lymphocyte immunostaining. Ex vivo staining of AAAs and thoracic aortas with 18F-DPA714 and unlabeled competitors was performed. RESULTS AAAs developed in 35 out of 46 cases. FCH uptake in AAAs was lower than that of FDG in all cases on imaging, with lower AAA-to-background maximal standardized uptake value (SUV(max)) ratios (1.78 ± 0.40 vs 2.71 ± 0.54; P < .01 for SUV(max) ratios), and lower AAA-to-normal aorta activity ratios on autoradiography (3.52 ± 1.26 vs 8.55 ± 4.23; P < .005). FDG AAA-to-background SUV(max) ratios correlated with the intensity of CD8 + ED1 staining (r = .76; P < .03). FCH AAA-to-background SUV(max) ratios correlated with the intensity of ED1 staining (r = .80; P < .03). 18F-DPA714 uptake was similar in AAAs and in normal aortas, both in vivo and ex vivo. CONCLUSIONS In rat experimental AAA, characterized by an important aortic wall leukocytes activity, FDG-PET showed higher sensitivity than FCH-PET and 18F-DPA714-PET to detect activated leukocytes. This enhances potential interest of this tracer for prognostic evaluation of AAA in patients.
Collapse
Affiliation(s)
- Laure Sarda-Mantel
- Institut National de la Santé et de la Recherche Médicale Unit 698, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
111
|
Frösen J, Tulamo R, Paetau A, Laaksamo E, Korja M, Laakso A, Niemelä M, Hernesniemi J. Saccular intracranial aneurysm: pathology and mechanisms. Acta Neuropathol 2012; 123:773-86. [PMID: 22249619 DOI: 10.1007/s00401-011-0939-3] [Citation(s) in RCA: 326] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 12/22/2011] [Accepted: 12/31/2011] [Indexed: 01/06/2023]
Abstract
Saccular intracranial aneurysms (sIA) are pouch-like pathological dilatations of intracranial arteries that develop when the cerebral artery wall becomes too weak to resist hemodynamic pressure and distends. Some sIAs remain stable over time, but in others mural cells die, the matrix degenerates, and eventually the wall ruptures, causing life-threatening hemorrhage. The wall of unruptured sIAs is characterized by myointimal hyperplasia and organizing thrombus, whereas that of ruptured sIAs is characterized by a decellularized, degenerated matrix and a poorly organized luminal thrombus. Cell-mediated and humoral inflammatory reaction is seen in both, but inflammation is clearly associated with degenerated and ruptured walls. Inflammation, however, seems to be a reaction to the ongoing degenerative processes, rather than the cause. Current data suggest that the loss of mural cells and wall degeneration are related to impaired endothelial function and high oxidative stress, caused in part by luminal thrombosis. The aberrant flow conditions caused by sIA geometry are the likely cause of the endothelial dysfunction, which results in accumulation of cytotoxic and pro-inflammatory substances into the sIA wall, as well as thrombus formation. This may start the processes that eventually can lead to the decellularized and degenerated sIA wall that is prone to rupture.
Collapse
Affiliation(s)
- Juhana Frösen
- Department of Neurosurgery, Helsinki University Central Hospital, Topeliuksenkatu 5, 00260 Helsinki, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
112
|
|
113
|
Blood-borne proteases are the main determinant of wall enlargement and rupture in abdominal aortic aneurysms and vulnerable plaque rupture through wall fibrino-cruoric formation. Vascul Pharmacol 2012. [DOI: 10.1016/j.vph.2011.08.099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
114
|
Skilton MR, Sullivan TR, Ayer JG, Harmer JA, Toelle BG, Webb K, Marks GB, Celermajer DS. Carotid extra-medial thickness in childhood: early life effects on the arterial adventitia. Atherosclerosis 2012; 222:478-82. [PMID: 22534523 DOI: 10.1016/j.atherosclerosis.2012.03.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 03/23/2012] [Accepted: 03/24/2012] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Structural modification of the arterial adventitia may be an early event in atherosclerosis. Carotid extra-medial thickness is a new measure of arterial adventitial thickness. We examined the association of cardiovascular risk factors with extra-medial thickness, in childhood. METHODS Carotid extra-medial thickness was assessed by high-resolution ultrasound in 389 non-diabetic children aged 8-years. A non-fasting blood sample was collected from 314 participants. Associations of gender, age, lipoproteins, blood pressure, BMI z-score, waist:height ratio and parental history of early vascular disease, with extra-medial thickness were examined. RESULTS Carotid extra-medial thickness was lower in girls (r=-.163, P=.001) and directly associated with systolic (r=.128, P=.009), diastolic blood pressure (r=.130, P=.009), and height (r=.170, P=.0006). These associations remained after adjustment for carotid intima-media thickness. In multivariable analysis including carotid intima-media thickness, only gender and height were significantly associated with carotid extra-medial thickness. In gender-stratified analysis, the strongest associations with extra-medial thickness were BMI z-score (r=.181, P=.01), height (r=.210, P=.003) and diastolic blood pressure (r=.167, P=.02) for boys; and systolic blood pressure (r=.153, P=.03) and parental history of premature cardiovascular disease (r=.139, P=.05) for girls. The association of BMI z-score with extra-medial thickness differed by gender (P-interaction=.04). CONCLUSIONS Carotid extra-medial thickness is independently associated with gender and height in childhood. Extra-medial thickness may provide important information concerning early arterial health, particularly related to the arterial adventitia.
Collapse
Affiliation(s)
- Michael R Skilton
- Boden Institute of Obesity, Nutrition, Exercise and Eating Disorders, University of Sydney, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Lacolley P, Regnault V, Nicoletti A, Li Z, Michel JB. The vascular smooth muscle cell in arterial pathology: a cell that can take on multiple roles. Cardiovasc Res 2012; 95:194-204. [DOI: 10.1093/cvr/cvs135] [Citation(s) in RCA: 477] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
116
|
Miyama N, Dua MM, Schultz GM, Kosuge H, Terashima M, Pisani LJ, Dalman RL, McConnell MV. Bioluminescence and Magnetic Resonance Imaging of Macrophage Homing to Experimental Abdominal Aortic Aneurysms. Mol Imaging 2012. [DOI: 10.2310/7290.2011.00033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Macrophage infiltration is a prominent feature of abdominal aortic aneurysm (AAA) progression. We used a combined imaging approach with bioluminescence (BLI) and magnetic resonance imaging (MRI) to study macrophage homing and accumulation in experimental AAA disease. Murine AAAs were created via intra-aortic infusion of porcine pancreatic elastase. Mice were imaged over 14 days after injection of prepared peritoneal macrophages. For BLI, macrophages were from transgenic mice expressing luciferase. For MRI, macrophages were labeled with iron oxide particles. Macrophage accumulation during aneurysm progression was observed by in situ BLI and by in vivo 7T MRI. Mice were sacrificed after imaging for histologic analysis. In situ BLI ( n = 32) demonstrated high signal in the AAA by days 7 and 14, which correlated significantly with macrophage number and aortic diameter. In vivo 7T MRI ( n = 13) at day 14 demonstrated T2* signal loss in the AAA and not in sham mice. Immunohistochemistry and Prussian blue staining confirmed the presence of injected macrophages in the AAA. BLI and MRI provide complementary approaches to track macrophage homing and accumulation in experimental AAAs. Similar dual imaging strategies may aid the study of AAA biology and the evaluation of novel therapies.
Collapse
Affiliation(s)
- Noriyuki Miyama
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Monica M. Dua
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Geoffrey M. Schultz
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Hisanori Kosuge
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Masahiro Terashima
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Laura J. Pisani
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Ronald L. Dalman
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Michael V. McConnell
- From the Divisions of Vascular Surgery and Cardiovascular Medicine and the Department of Radiology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
117
|
Ramos-Mozo P, Madrigal-Matute J, Vega de Ceniga M, Blanco-Colio LM, Meilhac O, Feldman L, Michel JB, Clancy P, Golledge J, Norman PE, Egido J, Martin-Ventura JL. Increased plasma levels of NGAL, a marker of neutrophil activation, in patients with abdominal aortic aneurysm. Atherosclerosis 2012; 220:552-6. [DOI: 10.1016/j.atherosclerosis.2011.11.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 10/26/2011] [Accepted: 11/16/2011] [Indexed: 10/15/2022]
|
118
|
Vascular effects of glycoprotein130 ligands--part I: pathophysiological role. Vascul Pharmacol 2011; 56:34-46. [PMID: 22197898 DOI: 10.1016/j.vph.2011.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 12/02/2011] [Accepted: 12/09/2011] [Indexed: 12/25/2022]
Abstract
The vessel wall is no longer considered as only an anatomical barrier for blood cells but is recognized as an active endocrine organ. Dysfunction of the vessel wall occurs in various disease processes including atherosclerosis, hypertension, peripheral artery disease, aneurysms, and transplant and diabetic vasculopathies. Different cytokines were shown to modulate the behavior of the cells, which constitute the vessel wall such as immune cells, endothelial cells and smooth muscle cells. Glycoprotein 130 (gp130) is a common cytokine receptor that controls the activity of a group of cytokines, namely, interleukin (IL)-6, oncostatin M (OSM), IL-11, ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), cardiotrophin-1 (CT-1), cardiotrophin-like cytokine (CLC), IL-27, and neuropoietin (NP). Gp130 and associated cytokines have abundantly diverse functions. Part I of this review focuses on the pathophysiological functions of gp130 ligands. We specifically describe vascular effects of these molecules and discuss the respective underlying molecular and cellular mechanisms.
Collapse
|
119
|
Ramos-Mozo P, Madrigal-Matute J, Martinez-Pinna R, Blanco-Colio LM, Lopez JA, Camafeita E, Meilhac O, Michel JB, Aparicio C, de Ceniga MV, Egido J, Martín-Ventura JL. Proteomic Analysis of Polymorphonuclear Neutrophils Identifies Catalase as a Novel Biomarker of Abdominal Aortic Aneurysm: Potential Implication of Oxidative Stress in Abdominal Aortic Aneurysm Progression. Arterioscler Thromb Vasc Biol 2011; 31:3011-9. [DOI: 10.1161/atvbaha.111.237537] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Polymorphonuclear neutrophils (PMNs) play a main role in abdominal aortic aneurysm (AAA) progression. We have analyzed circulating PMNs isolated from AAA patients and controls by a proteomic approach to identify proteins potentially involved in AAA pathogenesis.
Methods and Results—
PMNs from 8 AAA patients (4 large AAA >5 cm and 4 small AAA 3–5 cm) and 4 controls were analyzed by 2D differential in-gel electrophoresis. Among differentially expressed spots, several proteins involved in redox balance were identified by mass spectrometry (eg, cyclophilin, thioredoxin reductase, catalase). Diminished catalase expression and activity were observed in PMNs from AAA patients compared with controls. In contrast, PMNs from AAA patients displayed higher H
2
O
2
and myeloperoxidase levels than PMNs from controls. Moreover, a significant decrease in catalase mRNA levels was observed in PMNs after phorbol 12-myristate 13-acetate incubation. Catalase plasma levels were also decreased in large (n=47) and small (n=56) AAA patients compared with controls (n=34). We observed catalase expression in AAA thrombus and thrombus-conditioned medium, associated with PMN infiltration. Furthermore, increased H
2
O
2
levels were observed in AAA thrombus-conditioned medium compared with the media layer.
Conclusion—
Diminished catalase levels in circulating PMNs and plasma are observed in AAA patients, supporting an important role of oxidative stress in AAA evolution.
Collapse
Affiliation(s)
- Priscila Ramos-Mozo
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Julio Madrigal-Matute
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Roxana Martinez-Pinna
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Luis Miguel Blanco-Colio
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Juan Antonio Lopez
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Emilio Camafeita
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Olivier Meilhac
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Jean-Baptiste Michel
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Cesar Aparicio
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Melina Vega de Ceniga
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - Jesus Egido
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| | - José Luis Martín-Ventura
- From the Vascular Research Laboratory (P.R.-M., J.M.-M., R.M.-P., L.M.B.-C., J.E., J.L.M.-V.) and Vascular Surgery (C.A.), Instituto de Investigaciones Sanitarias, Fundación Jiménez Diaz, Autonoma University, Madrid, Spain; Unidad de Proteomica, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (J.A.L., E.C.); Institut National de la Santé et de la Recherche Médicale, U698, Univ Paris 7, College Hospital Universitary, X-Bichat, Paris, France (O.M., J.-B.M.); Galdakao Hospital,
| |
Collapse
|
120
|
Ho-Tin-Noé B, Le Dall J, Gomez D, Louedec L, Vranckx R, El-Bouchtaoui M, Legrès L, Meilhac O, Michel JB. Early atheroma-derived agonists of peroxisome proliferator-activated receptor-γ trigger intramedial angiogenesis in a smooth muscle cell-dependent manner. Circ Res 2011; 109:1003-14. [PMID: 21885829 DOI: 10.1161/circresaha.110.235390] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Neovascularization favors intraplaque hemorrhage and plaque rupture. Development of therapeutic strategies against atheromatous angiogenesis requires elucidation of its initiating factors. OBJECTIVE We investigated the contribution of smooth muscle cells (SMCs) and atheroma-derived lipids to the initiation of atheroma-associated neoangiogenesis. METHODS AND RESULTS Forty human aortic segments, each harvested from a different donor, were classified as healthy or as bearing early atheromatous lesions, including fatty streaks and fibrolipidic atheroma, according to their histological features. Immunostaining for blood vessels and vascular endothelial growth factor-A (VEGF-A), as well as measurement of VEGF-A protein and mRNA levels by ELISA and real-time PCR, revealed that angiogenesis and VEGF-A production were enhanced in the medial layer of atheromatous aortas. The intramedial vessel density and invasiveness and the production of VEGF-A by medial SMCs were indeed increased in atheromatous aortas compared with healthy aortas. Furthermore, intimal layers of atheromatous aortas were enriched in soluble lipid mediators capable of inducing a sustained increase in VEGF-A production by medial SMCs, turning these cells into potent inducers of angiogenesis when incorporated into mouse Matrigel implants. Both effects were inhibited by the peroxisome proliferator-activated receptor-γ inhibitor GW9662 and mimicked by its agonist, rosiglitazone. CONCLUSIONS We show that VEGF-A production is upregulated in medial SMCs of human atheromatous aortas and that peroxisome proliferator-activated receptor-γ agonists derived from early intimal lesions are likely to contribute to this phenotypic change. Our findings suggest that medial SMCs are central organizers of an angiogenic response initiated by the subendothelial accumulation of atherogenic lipids.
Collapse
|
121
|
Hu Y, Xu Q. Adventitial biology: differentiation and function. Arterioscler Thromb Vasc Biol 2011; 31:1523-9. [PMID: 21677295 DOI: 10.1161/atvbaha.110.221176] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent evidence indicates that stem/progenitor cells are present in the adventitia and participate in vascular repair and the formation of neointimal lesions in severely damaged vessels. Data have also demonstrated that these resident stem/progenitor cells could differentiate into endothelial or smooth muscle cells in response to different stimuli. Under pathological conditions, adventitial inflammation results in releasing a panel of cytokines, such as stromal cell-derived factor-1 and tumor necrosis factor-α, that may lead to local stem/progenitor mobilization and differentiation. Overall, these data support the impact of the adventitial progenitors in pathophysiological processes of lesion development in the arterial wall. In the present review, we aim to summarize the data concerning the presence of the resident stem cells and discuss the pathological impact of the adventitia in vascular diseases. We will also discuss the possible signal pathways orchestrating stem cell differentiation toward vascular lineage and highlight controversial issues related to the role of adventitial progenitors.
Collapse
Affiliation(s)
- Yanhua Hu
- Cardiovascular Division, King's College London British Heart Foundation, Centre, London, United Kingdom
| | | |
Collapse
|
122
|
Alvira CM, Guignabert C, Kim YM, Chen C, Wang L, Duong TT, Yeung RSM, Li DY, Rabinovitch M. Inhibition of transforming growth factor β worsens elastin degradation in a murine model of Kawasaki disease. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1210-20. [PMID: 21356372 DOI: 10.1016/j.ajpath.2010.11.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2010] [Revised: 11/03/2010] [Accepted: 11/09/2010] [Indexed: 12/13/2022]
Abstract
Kawasaki disease (KD) is an acute inflammatory illness marked by coronary arteritis. However, the factors increasing susceptibility to coronary artery lesions are unknown. Because transforming growth factor (TGF) β increases elastin synthesis and suppresses proteolysis, we hypothesized that, in contrast to the benefit observed in aneurysms forming in those with Marfan syndrome, inhibition of TGF-β would worsen inflammatory-induced coronary artery lesions. By using a murine model of KD in which injection of Lactobacillus casei wall extract (LCWE) induces coronary arteritis, we show that LCWE increased TGF-β signaling in the coronary smooth muscle cells beginning at 2 days and continuing through 14 days, the point of peak coronary inflammation. By 42 days, LCWE caused fragmentation of the internal and external elastic lamina. Blocking TGF-β by administration of a neutralizing antibody accentuated the LCWE-mediated fragmentation of elastin and induced an overall loss of medial elastin without increasing the inflammatory response. We attributed these increased pathological characteristics to a reduction in the proteolytic inhibitor, plasminogen activator inhibitor-1, and an associated threefold increase in matrix metalloproteinase 9 activity compared with LCWE alone. Therefore, our data demonstrate that in the coronary arteritis associated with KD, TGF-β suppresses elastin degradation by inhibiting plasmin-mediated matrix metalloproteinase 9 activation. Thus, strategies to block TGF-β, used in those with Marfan syndrome, are unlikely to be beneficial and could be detrimental.
Collapse
Affiliation(s)
- Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305-5162, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Delbosc S, Alsac JM, Journe C, Louedec L, Castier Y, Bonnaure-Mallet M, Ruimy R, Rossignol P, Bouchard P, Michel JB, Meilhac O. Porphyromonas gingivalis participates in pathogenesis of human abdominal aortic aneurysm by neutrophil activation. Proof of concept in rats. PLoS One 2011; 6:e18679. [PMID: 21533243 PMCID: PMC3076426 DOI: 10.1371/journal.pone.0018679] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 03/08/2011] [Indexed: 11/29/2022] Open
Abstract
Background Abdominal Aortic Aneurysms (AAAs) represent a particular form of atherothrombosis where neutrophil proteolytic activity plays a major role. We postulated that neutrophil recruitment and activation participating in AAA growth may originate in part from repeated episodes of periodontal bacteremia. Methods and Findings Our results show that neutrophil activation in human AAA was associated with Neutrophil Extracellular Trap (NET) formation in the IntraLuminal Thrombus, leading to the release of cell-free DNA. Human AAA samples were shown to contain bacterial DNA with high frequency (11/16), and in particular that of Porphyromonas gingivalis (Pg), the most prevalent pathogen involved in chronic periodontitis, a common form of periodontal disease. Both DNA reflecting the presence of NETs and antibodies to Pg were found to be increased in plasma of patients with AAA. Using a rat model of AAA, we demonstrated that repeated injection of Pg fostered aneurysm development, associated with pathological characteristics similar to those observed in humans, such as the persistence of a neutrophil-rich luminal thrombus, not observed in saline-injected rats in which a healing process was observed. Conclusions Thus, the control of periodontal disease may represent a therapeutic target to limit human AAA progression.
Collapse
Affiliation(s)
- Sandrine Delbosc
- INSERM (Institut National de la Santé et de la Recherche Médicale) U698, Paris, France
- Université Denis Diderot, Paris, France
| | - Jean-Marc Alsac
- INSERM (Institut National de la Santé et de la Recherche Médicale) U698, Paris, France
- Service de chirurgie cardiovasculaire, Hôpital Européen Georges Pompidou, APHP (Assistance Publique Hôpitaux de Paris), Paris, France
| | - Clement Journe
- INSERM (Institut National de la Santé et de la Recherche Médicale) U698, Paris, France
- Université Denis Diderot, Paris, France
| | - Liliane Louedec
- INSERM (Institut National de la Santé et de la Recherche Médicale) U698, Paris, France
- Université Denis Diderot, Paris, France
| | - Yves Castier
- Service de chirurgie thoracique et vasculaire, Hôpital Xavier Bichat-Claude Bernard, APHP (Assistance Publique Hôpitaux de Paris), Paris, France
| | - Martine Bonnaure-Mallet
- Equipe de Microbiologie, UPRES-EA (Unité Propre de Recherche de l'Enseignement Superieur-Equipe d'Accueil) 1254, Université Européenne de Bretagne, Université de Rennes I, Rennes, France
| | - Raymond Ruimy
- Service de bactériologie et virologie, Hôpital Xavier Bichat-Claude Bernard, APHP (Assistance Publique Hôpitaux de Paris), Paris, France
| | - Patrick Rossignol
- CHU (Centre Hospitalier Universitaire) de Nancy, CIC (Centre d'Investigation Clinique); CIC9501; Université Nancy, Faculté de Médecine; Inserm, U961, Vandoeuvre lès Nancy, France; Service de médecine vasculaire et hypertension, Hôpital Européen Georges Pompidou, Paris, France
| | - Philippe Bouchard
- Université Denis Diderot, Paris, France
- Département de Parodontologie, Service d'odontologie, Hôpital Garancière Rothschild, APHP (Assistance Publique Hôpitaux de Paris), Paris, France
| | - Jean-Baptiste Michel
- INSERM (Institut National de la Santé et de la Recherche Médicale) U698, Paris, France
- Université Denis Diderot, Paris, France
| | - Olivier Meilhac
- INSERM (Institut National de la Santé et de la Recherche Médicale) U698, Paris, France
- Université Denis Diderot, Paris, France
- * E-mail:
| |
Collapse
|
124
|
Fluid flow mechanotransduction in vascular smooth muscle cells and fibroblasts. Ann Biomed Eng 2011; 39:1608-19. [PMID: 21479754 DOI: 10.1007/s10439-011-0309-2] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/04/2011] [Indexed: 12/29/2022]
Abstract
Understanding how vascular wall endothelial cells (ECs), smooth muscle cells (SMCs), and fibroblasts (FBs) sense and transduce the stimuli of hemodynamic forces (shear stress, cyclic strain, and hydrostatic pressure) into intracellular biochemical signals is critical to prevent vascular disease development and progression. ECs lining the vessel lumen directly sense alterations in blood flow shear stress and then communicate with medial SMCs and adventitial FBs to regulate vessel function and disease. Shear stress mechanotransduction in ECs has been extensively studied and reviewed. In the case of endothelial damage, blood flow shear stress may directly act on the superficial layer of SMCs and transmural interstitial flow may be elevated on medial SMCs and adventitial FBs. Therefore, it is also important to investigate direct shear effects on vascular SMCs as well as FBs. The work published in the last two decades has shown that shear stress and interstitial flow have significant influences on vascular SMCs and FBs. This review summarizes work that considered direct shear effects on SMCs and FBs and provides the first comprehensive overview of the underlying mechanisms that modulate SMC secretion, alignment, contraction, proliferation, apoptosis, differentiation, and migration in response to 2-dimensional (2D) laminar, pulsatile, and oscillating flow shear stresses and 3D interstitial flow. A mechanistic model of flow sensing by SMCs is also provided to elucidate possible mechanotransduction pathways through surface glycocalyx, integrins, membrane receptors, ion channels, and primary cilia. Understanding flow-mediated mechanotransduction in SMCs and FBs and the interplay with ECs should be helpful in exploring strategies to prevent flow-initiated atherosclerosis and neointima formation and has implications in vascular tissue engineering.
Collapse
|
125
|
Michel JB, Virmani R, Arbustini E, Pasterkamp G. Intraplaque haemorrhages as the trigger of plaque vulnerability. Eur Heart J 2011; 32:1977-85, 1985a, 1985b, 1985c. [PMID: 21398643 PMCID: PMC3155759 DOI: 10.1093/eurheartj/ehr054] [Citation(s) in RCA: 261] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Atherothrombosis remains one of the main causes of morbidity and mortality in the western countries. Human atherothrombotic disease begins early in life in relation to circulating lipid retention in the inner vascular wall. Risk factors enhance the progression towards clinical expression: dyslipidaemia, diabetes, smoking, hypertension, ageing, etc. The evolution from the initial lipid retention in the arterial wall to clinical events is a continuum of increasingly complex biological processes. Current strategies to fight the consequences of atherothrombosis are orientated either towards the promotion of a healthy life style and preventive treatment of risk factors, or towards late interventional strategies. Despite this therapeutic arsenal, the incidence of clinical events remains dramatically high, dependent, at least in part, on the increasing frequency of type 2 diabetes and ageing. But some medical treatments, focusing only on prevention of the metabolic risk, have failed to reduce cardiovascular mortality, thus illustrating that our understanding of the pathophysiology of human atherothrombosis leading to clinical events remain incomplete. New paradigms are now emerging which may give rise to novel experimental strategies to improve therapeutic efficacy and prediction of disease progression. Recent studies strengthen the concept that the intraplaque neovascularization and bleeding (Figure 1, upper panel) are events that could play a major role in plaque progression and leucocyte infiltration, and may also serve as a measure of risk for the development of future events. The recent advances in our understanding of IntraPlaque Hemorrhage as a critical event in triggering acute clinical events have important implications for clinical research and possibly future clinical practice.
Collapse
Affiliation(s)
- Jean-Baptiste Michel
- UMR 698 Inserm, Paris 7-Denis Diderot University, Xavier Bichat Hospital, 46 rue Henri Huchard, 75018 Paris, France.
| | | | | | | |
Collapse
|
126
|
Skilton MR, Boussel L, Bonnet F, Bernard S, Douek PC, Moulin P, Serusclat A. Carotid intima-media and adventitial thickening: comparison of new and established ultrasound and magnetic resonance imaging techniques. Atherosclerosis 2011; 215:405-10. [PMID: 21300355 DOI: 10.1016/j.atherosclerosis.2010.12.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 12/13/2010] [Accepted: 12/30/2010] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Carotid intima-media thickness is a well established non-invasive surrogate marker of cardiovascular disease, however there is evidence that structural modification of the arterial adventitia also accompanies cardiovascular risk factors and might be involved causally in atherosclerosis. We sought to determine the relative contributions of the intima-media and adventitia to variation in ultrasound and magnetic resonance derived measures of carotid wall thickness. METHODS Carotid ultrasound and magnetic resonance imaging were undertaken in 20 participants. Carotid intima-media thickness, carotid extra-media thickness (which incorporates the arterial adventitia) and total wall thickness (a combined near-wall intima-media thickness and carotid extra-media thickness) using high-resolution ultrasound, and wall thickness using magnetic resonance imaging, were obtained. RESULTS All ultrasound-derived measures of the arterial wall thickness were highly correlated with wall thickness by magnetic resonance imaging (all P<0.001); as expected the total wall thickness by ultrasound measure was most tightly correlated (correlation coefficient=0.814, P<0.0001). In multivariable models, there was evidence that both carotid intima-media thickness and carotid extra-media thickness contributed independently to the variance in wall thickness by magnetic resonance imaging, especially for the most severe focal thickening. Measures of carotid wall thickness that incorporated all three layers of the arterial wall were more closely correlated with the number of cardiovascular risk factors than carotid intima-media thickness alone. CONCLUSIONS These results indicate that the arterial adventitia is an important contributor to the wall thickness measure derived by magnetic resonance imaging, and that carotid extra-media thickness likely provides additional information concerning arterial structure than that obtained from carotid intima-media thickness alone.
Collapse
Affiliation(s)
- Michael R Skilton
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
127
|
Tieu BC, Ju X, Lee C, Sun H, Lejeune W, Recinos A, Brasier AR, Tilton RG. Aortic adventitial fibroblasts participate in angiotensin-induced vascular wall inflammation and remodeling. J Vasc Res 2010; 48:261-72. [PMID: 21099231 DOI: 10.1159/000320358] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 07/26/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The role of adventitial fibroblasts in the vascular inflammation observed in the adventitia of large vessels in numerous cardiovascular diseases remains unclear. Our objective was to explore the contribution of these cells to angiotensin II (Ang II)-induced aortic inflammation and adventitial expansion. METHODS Cytokine production by primary human aortic adventitial fibroblasts (AoAF) in tissue culture was detected using multiplex ELISA, and increases in cytokine mRNA following Ang II stimulation were quantitated by real-time PCR. The ability of AoAF-derived MCP-1 to attract monocytes was studied in vitro using Boyden assays, and the resulting effect of the monocyte-AoAF interaction on fibroblast proliferation was measured in vitro using proliferation and (3)H-thymidine incorporation assays. Ang II-induced fibroblast proliferation was measured in vivo using aortic digestion of single cells followed by flow cytometric quantification of fibroblast numbers as well as fibroblast and PCNA immunostaining. The ability of monocytes to induce AoAF proliferation was demonstrated in vivo using CCR2(+/+) wild-type monocyte adoptive transfer into Ang II-stimulated CCR2-null mice which can produce MCP-1 but have cells lacking the MCP-1 receptor - CCR2. RESULTS AoAF constitutively secreted numerous proinflammatory cytokines, particularly IL-6 and MCP-1, whose gene expressions were further upregulated in response to Ang II stimulation. AoAF-derived MCP-1 was potent in recruiting THP-1 monocytes in vitro, and these monocytes stimulated AoAF proliferation based on a flow cytometric assessment of cell number and (3)H-thymidine incorporation in tissue culture. In vivo, Ang II induced fibroblast proliferation, increased fibroblast and PCNA adventitial staining, and blunted inflammatory responses in the CCR2(-/-) background. Injection of CCR2(+/+) monocytes into Ang II-treated CCR2(-/-) mice restored adventitial thickening which resulted in increased fibrosis secondary to adventitial fibroblast proliferation. CONCLUSIONS Our results suggest that Ang II-stimulates AoAF to recruit monocytes via fibroblast-derived MCP-1, and the recruited monocytes further activate fibroblast proliferation, adventitial thickening, and additional cytokine production. This fibroblast-monocyte amplification loop may critically mediate hallmarks of adventitial inflammation common to many cardiovascular diseases.
Collapse
Affiliation(s)
- Brian C Tieu
- Department of Biochemistry, University of Texas Medical Branch, Galveston, TX 77555-1060, USA
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Soluble TWEAK plasma levels predict expansion of human abdominal aortic aneurysms. Atherosclerosis 2010; 214:486-9. [PMID: 21130992 DOI: 10.1016/j.atherosclerosis.2010.11.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 10/21/2010] [Accepted: 11/05/2010] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Diminished soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) concentrations are associated with cardiovascular diseases. We have analyzed sTWEAK levels and its relation with expansion rate in subjects with abdominal aortic aneurysm (AAA). METHODS sTWEAK levels were measured by ELISA. RESULTS sTWEAK concentrations were diminished in small AAA (≤ 5 cm; 353 ± 12 pg/mL; n = 25, p = 0.03) and large AAA (>5 cm; 315 ± 21 pg/mL; n = 18, p = 0.004) compared with healthy subjects (411 ± 22 pg/mL; n=27). Moreover, sTWEAK concentrations were negatively associated with AAA size (r = -0.4; p = 0.008). sTWEAK was also negatively associated with AAA expansion rate with 5 years of follow-up (n = 79, r = -0.263; p = 0.031). Multivariate regression analysis revealed that sTWEAK levels were independently associated with AAA growth rate (β = -0.208; p = 0.046). CONCLUSIONS sTWEAK plasma levels were decreased in subjects with AAA and were independently related with AAA expansion rate indicating that this protein could be a novel diagnostic and prognostic biomarker of AAA.
Collapse
|
129
|
Olive M, Harten I, Mitchell R, Beers J, Djabali K, Cao K, Erdos MR, Blair C, Funke B, Smoot L, Gerhard-Herman M, Machan JT, Kutys R, Virmani R, Collins FS, Wight TN, Nabel EG, Gordon LB. Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging. Arterioscler Thromb Vasc Biol 2010; 30:2301-9. [PMID: 20798379 PMCID: PMC2965471 DOI: 10.1161/atvbaha.110.209460] [Citation(s) in RCA: 311] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Children with Hutchinson-Gilford progeria syndrome (HGPS) exhibit dramatically accelerated cardiovascular disease (CVD), causing death from myocardial infarction or stroke between the ages of 7 and 20 years. We undertook the first histological comparative evaluation between genetically confirmed HGPS and the CVD of aging. METHODS AND RESULTS We present structural and immunohistological analysis of cardiovascular tissues from 2 children with HGPS who died of myocardial infarction. Both had features classically associated with the atherosclerosis of aging, as well as arteriolosclerosis of small vessels. In addition, vessels exhibited prominent adventitial fibrosis, a previously undescribed feature of HGPS. Importantly, although progerin was detected at higher rates in the HGPS coronary arteries, it was also present in non-HGPS individuals. Between the ages of 1 month and 97 years, progerin staining increased an average of 3.34% per year (P<0.0001) in coronary arteries. CONCLUSIONS We find concordance among many aspects of cardiovascular pathology in both HGPS and geriatric patients. HGPS generates a more prominent adventitial fibrosis than typical CVD. Vascular progerin generation in young non-HGPS individuals, which significantly increases throughout life, strongly suggests that progerin has a role in cardiovascular aging of the general population.
Collapse
Affiliation(s)
- Michelle Olive
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ingrid Harten
- Hope Heart Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101-2795
- Department of Pathology, University of Washington, Seattle, WA 98195
| | - Richard Mitchell
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School Boston, MA 02115
| | - Jeanette Beers
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Karima Djabali
- Department of Dermatology, University Technique of Munich (TUM), 85748 Munich-Garching, Germany
| | - Kan Cao
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michael R. Erdos
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Cecilia Blair
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Birgit Funke
- Laboratory for Molecular Medicine, Cambridge, MA. Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Leslie Smoot
- Department of Cardiology, Children’s Hospital Boston, and Harvard Medical School Boston, MA 02115
| | - Marie Gerhard-Herman
- Department of Cardiology, Brigham and Women’s Hospital and Harvard Medical School Boston, MA 02115
| | - Jason T. Machan
- Biostatistics, Rhode Island Hospital Departments of Orthopaedics and Surgery, Warren Alpert Medical School of Brown University, Providence, RI 02912
| | | | | | - Francis S. Collins
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Thomas N. Wight
- Hope Heart Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101-2795
- Department of Pathology, University of Washington, Seattle, WA 98195
| | - Elizabeth G. Nabel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Leslie B. Gordon
- Department of Pediatrics, Hasbro Children’s Hospital and Warren Alpert Medical School of Brown University, Providence, RI 02912
- Department of Anesthesia, Children’s Hospital Boston, and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
130
|
Abstract
Arterial remodeling over time is a cornerstone of normal systemic aging. The age-associated arterial structural and functional changes in the intima, the media, and the adventitia are closely linked to angiotensin II (Ang II) signaling. A growing line of evidence indicates that essential elements of Ang II signaling, which encompasses milk fat globule epidermal growth factor-8, calpain-1, transforming growth factor-β1, matrix metalloproteinase-2/9, monocyte chemoattractant protein-1, nicotinamide adenine dinucleotide phosphate-oxidase, and reactive oxygen species, are upregulated within the central arterial wall in rats, nonhuman primates, and humans during aging. In vitro studies show that the elevation of Ang II signaling induces the accumulation of collagen and advanced glycated end-products, the degradation of elastin, and the increased cell cycle disorder, invasion, and hypertrophy of endothelial and vascular smooth muscle cells. Further, in vivo studies demonstrate that increased Ang II signaling accelerates arterial aging. Conversely, attenuating Ang II signaling via an inhibition of angiotensin conversing enzyme or a blockade of AT1 activation retards age-associated arterial remodeling. This review attempts to integrate complex facts of Ang II signaling within the aged central arterial wall and may shed light on new therapeutic targets for arterial aging.
Collapse
Affiliation(s)
- Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | | | | |
Collapse
|
131
|
Tulamo R, Frösen J, Paetau A, Seitsonen S, Hernesniemi J, Niemelä M, Järvelä I, Meri S. Lack of complement inhibitors in the outer intracranial artery aneurysm wall associates with complement terminal pathway activation. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:3224-32. [PMID: 20971742 DOI: 10.2353/ajpath.2010.091172] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Inflammation and activation of the complement system predispose to intracranial artery aneurysm (IA) rupture. Because disturbances in complement regulation may lead to increased susceptibility to complement activation and inflammation, we looked for evidence for dysregulation of the complement system in 26 unruptured and 26 ruptured IAs resected intraoperatively. Immunohistochemical and immunofluorescence results of parallel IA sections showed that deposition of the complement activation end-product C5b-9 was lacking from the luminal part of the IA wall that contained complement inhibitors factor H, C4b binding protein, and protectin as well as glycosaminoglycans. In contrast, the outer, less cellular part of the IA wall lacked protectin and had enabled full complement activation and C5b-9 formation. Decay accelerating factor and membrane cofactor protein had less evident roles in complement regulation. The Factor H Y402H variant, studied in 97 IA patients, was seen as often in aneurysm patients with or without aneurysm rupture as in the control population. The regulatory capacity of the complement system thus appears disturbed in the outer part of the IA wall, allowing full proinflammatory complement activation to occur before aneurysm rupture. Insufficient complement control might be due to matrix remodeling and cell loss by mechanical hemodynamics and/or inflammatory stress. Apparently, disturbed complement regulation leads to an increased susceptibility to complement activation, inflammation, and tissue damage in the IA wall.
Collapse
Affiliation(s)
- Riikka Tulamo
- Neurosurgery Research Group, Room B408a2, Biomedicum Helsinki 1, P.O. Box 700, FI-00029 Huch, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Efficiency of drug delivery to the coronary arteries in swine is dependent on the route of administration: assessment of luminal, intimal, and adventitial coronary artery and venous delivery methods. J Vasc Interv Radiol 2010; 21:1555-64. [PMID: 20813544 DOI: 10.1016/j.jvir.2010.05.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 04/13/2010] [Accepted: 05/26/2010] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To compare the efficiency of five different drug delivery methods to the coronary artery in swine. MATERIALS AND METHODS A nanoparticle-albumin-bound, nonradioactive isotopic marker was administered within the left anterior descending coronary artery (LAD) through a microinfusion catheter (MIC: adventitial, n = 8, and luminal, n = 4), a porous drug infusion balloon (DIB: intimal, n = 4), and a straight catheter (SC: luminal, n = 2) and within the superior vena cava (SC: intravenous, luminal, n = 2). The distribution of the marker in heart, lung, liver, kidney, muscle, blood, urine, and bile was determined 68-84 minutes after delivery. The heart was sectioned into six axial slices and each slice divided into four quadrants. The marker content was assayed by neutron bombardment and the total counts of disintegrations per minute (DPM) expressed as a percentage of the control for each device delivery control. RESULTS After luminal delivery with the nonactuated MIC (MIC-NA) or intimal delivery with the DIB, 0.17% ± 0.07 and 0.39% ± 0.09, respectively, less than 0.39% of the total marker was detected in the heart. After adventitial delivery with the actuated MIC (MIC-A), 63.1% ± 9.9 of the total marker was detected in the heart. Marker was only detected in quadrants containing the coronary LAD, with the highest level in the middle slice and lower marker levels in consecutive proximal and distal heart slices. The nonactuated MIC-NA and DIB drug infusion balloon patterns of marker distribution were similar to those of actuated MIC-A, although with reduced levels. These delivery methods were also associated with considerably more marker detected in the lungs and liver: at least 22% compared with 1.34% ± 1.34 for the actuated MIC-A There was one delivery failure with the actuated MIC. CONCLUSIONS Catheter-based adventitial delivery with the MIC-A represents a more efficient delivery method for retention of vascular therapeutics.
Collapse
|
133
|
Arapoglou V, Kondi-Pafiti A, Rizos D, Carvounis E, Frangou-Plemenou M, Kotsis T, Katsenis K. The Influence of Diabetes on Degree of Abdominal Aortic Aneurysm Tissue Inflammation. Vasc Endovascular Surg 2010; 44:454-9. [DOI: 10.1177/1538574410363748] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abdominal aortic aneurysm (AAA) progression and disease resistance are related to transmural degenerative processes and an inflammatory infiltration (INF). Diabetes is associated with low prevalence and growth rate of AAA. We sought to characterize INF in established AAA (INFAAA), in diabetic patients. From 89 male patients aged 52 to 83 years, aneurysm specimens obtained at open asymptomatic nonruptured AAA repair were graded for INF and immunostained using antibodies against T-lymphocytes (CD3) and macrophages (CD68). Diabetic patients had an odds ratio (OR) 3.8, 95% confidence interval ([CI] 1.14-12.96), P = .03, of experiencing above-median INFAAA. These associations were affected by serum glucose (SG) levels (OR 3.6, 95% CI [0.72-18.77]; P = .1). Macrophage subpopulations higher in diabetic patients (1.44 ± 0.78 versus 0.98 ± 0.76; P = .02) were correlated with SG (r = .21, P = .044). Abdominal aortic aneurysms in diabetic patients are associated with higher INF. Macrophage densities are correlated with SG.
Collapse
Affiliation(s)
- Vassilis Arapoglou
- Vascular Surgical Unit, 2nd Surgical Department, Aretaeion Hospital, Medical School, University of Athens, Greece,
| | - Agathi Kondi-Pafiti
- Pathology Laboratory, Areteion Hospital, Medical School, University of Athens, Greece
| | - Demetrios Rizos
- Hormone Laboratory, Areteion Hospital, Medical School, University of Athens, Greece
| | - Eleni Carvounis
- Pathology Laboratory, Areteion Hospital, Medical School, University of Athens, Greece
| | | | - Thomas Kotsis
- Vascular Surgical Unit, 2nd Surgical Department, Aretaeion Hospital, Medical School, University of Athens, Greece
| | - Konstantinos Katsenis
- Vascular Surgical Unit, 2nd Surgical Department, Aretaeion Hospital, Medical School, University of Athens, Greece
| |
Collapse
|
134
|
T-cadherin expression in cardiac allograft vasculopathy: Bench to bedside translational investigation. J Heart Lung Transplant 2010; 29:792-9. [DOI: 10.1016/j.healun.2010.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/02/2010] [Accepted: 03/03/2010] [Indexed: 11/19/2022] Open
|
135
|
Dai E, Liu LY, Wang H, McIvor D, Sun YM, Macaulay C, King E, Munuswamy-Ramanujam G, Bartee MY, Williams J, Davids J, Charo I, McFadden G, Esko JD, Lucas AR. Inhibition of chemokine-glycosaminoglycan interactions in donor tissue reduces mouse allograft vasculopathy and transplant rejection. PLoS One 2010; 5:e10510. [PMID: 20463901 PMCID: PMC2865544 DOI: 10.1371/journal.pone.0010510] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 04/05/2010] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Binding of chemokines to glycosaminoglycans (GAGs) is classically described as initiating inflammatory cell migration and creating tissue chemokine gradients that direct local leukocyte chemotaxis into damaged or transplanted tissues. While chemokine-receptor binding has been extensively studied during allograft transplantation, effects of glycosaminoglycan (GAG) interactions with chemokines on transplant longevity are less well known. Here we examine the impact of interrupting chemokine-GAG interactions and chemokine-receptor interactions, both locally and systemically, on vascular disease in allografts. METHODOLOGY/PRINCIPAL FINDINGS Analysis of GAG or CC chemokine receptor 2 (CCR2) deficiency were coupled with the infusion of viral chemokine modulating proteins (CMPs) in mouse aortic allograft transplants (n = 239 mice). Inflammatory cell invasion and neointimal hyperplasia were significantly reduced in N-deacetylase-N-sulfotransferase-1 (Ndst1(f/f)TekCre(+)) heparan sulfate (GAG)-deficient (Ndst1(-/-), p<0.044) and CCR2-deficient (Ccr2(-/-), p<0.04) donor transplants. Donor tissue GAG or CCR2 deficiency markedly reduced inflammation and vasculopathy, whereas recipient deficiencies did not. Treatment with three CMPs was also investigated; Poxviral M-T1 blocks CC chemokine receptor binding, M-T7 blocks C, CC, and CXC GAG binding, and herpesviral M3 binds receptor and GAG binding for all classes. M-T7 reduced intimal hyperplasia in wild type (WT) (Ccr2(+/+), p< or =0.003 and Ccr2(-/-), p=0.027) aortic allografts, but not in Ndst1(-/-) aortic allografts (p = 0.933). M-T1 and M3 inhibited WT (Ccr2(+/+) and Ndst1(+/+), p< or =0.006) allograft vasculopathy, but did not block vasculopathy in Ccr2(-/-) (p = 0.61). M-T7 treatment alone, even without immunosuppressive drugs, also significantly prolonged survival of renal allograft transplants (p< or =0.001). CONCLUSIONS/SIGNIFICANCE Interruption of chemokine-GAG interactions, even in the absence of chemokine-receptor blockade, is a highly effective approach to reduction of allograft rejection, reducing vascular inflammation and prolonging allograft survival. Although chemokines direct both local and systemic cell migration, interruption of inherent chemokine responses in the donor tissue unexpectedly had a greater therapeutic impact on allograft vasculopathy.
Collapse
Affiliation(s)
- Erbin Dai
- Vascular Biology Research Group, Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Li-Ying Liu
- Vascular Biology Research Group, Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Hao Wang
- Departments of Medicine and Surgery, and Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - Dana McIvor
- Departments of Medicine and Surgery, and Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - Yun ming Sun
- Vascular Biology Research Group, Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- Viron Therapeutics, Inc., London, Ontario, Canada
| | | | - Elaine King
- Viron Therapeutics, Inc., London, Ontario, Canada
| | - Ganesh Munuswamy-Ramanujam
- Vascular Biology Research Group, Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Mee Yong Bartee
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Jennifer Williams
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Jennifer Davids
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Israel Charo
- Gladstone Institute, San Francisco, California, United States of America
| | - Grant McFadden
- Departments of Medicine and Surgery, and Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, United States of America
| | - Alexandra R. Lucas
- Vascular Biology Research Group, Robarts Research Institute, The University of Western Ontario, London, Ontario, Canada
- Departments of Medicine and Surgery, and Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
136
|
Tsuruda T, Imamura T, Hatakeyama K, Asada Y, Kitamura K. Stromal cell biology--a way to understand the evolution of cardiovascular diseases. Circ J 2010; 74:1042-50. [PMID: 20378995 DOI: 10.1253/circj.cj-10-0024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Stromal cells, composed of fibroblasts, microvascular endothelial cells, immune cells and inflammatory cells, are critical determinants of the mechanical properties and function of the heart and vasculature, and the mechanisms whereby these types of cells are activated are important to understand the progression of cardiovascular diseases. Emerging studies have suggested that the activation of autocrine and paracrine signaling pathways by stromal cell-derived growth factors, cytokines and bioactive molecules contributes to disease progression. Disruption of the stromal network will result in alterations in the geometry and function in these organs. Interventions targeting the stromal cells (eg, myofibroblasts, microvascular endothelial cells, inflammatory cells) by pharmacological agents or direct gene delivery/small interfering RNA would be potential novel therapeutic strategies to prevent/attenuate the progression of cardiovascular disorders.
Collapse
Affiliation(s)
- Toshihiro Tsuruda
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, University of Miyazaki, Miyazaki, Japan.
| | | | | | | | | |
Collapse
|
137
|
Brasier AR. The nuclear factor-kappaB-interleukin-6 signalling pathway mediating vascular inflammation. Cardiovasc Res 2010; 86:211-8. [PMID: 20202975 DOI: 10.1093/cvr/cvq076] [Citation(s) in RCA: 432] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vascular inflammation is a common pathophysiological response to diverse cardiovascular disease processes, including atherosclerosis, myocardial infarction, congestive heart failure, and aortic aneurysms/dissection. Inflammation is an ordered process initiated by vascular injury that produces enhanced leucocyte adherence, chemotaxis, and finally activation in situ. This process is coordinated by local secretion of adhesion molecules, chemotactic factors, and cytokines whose expression is the result of vascular injury-induced signal transduction networks. A wide variety of mediators of the vascular injury response have been identified; these factors include vasoactive peptides (angiotensin II, Ang II), CD40 ligands, oxidized cholesterol, and advanced glycation end-products. Downstream, the nuclear factor-kappaB (NF-kappaB) transcription factor performs an important signal integration step, responding to mediators of vascular injury in a stimulus-dependent and cell type-specific manner. The ultimate consequence of NF-kappaB signalling is the activation of inflammatory genes including adhesion molecules and chemotaxins. However, clinically, the hallmark of vascular NF-kappaB activation is the production of interleukin-6 (IL-6), whose local role in vascular inflammation is relatively unknown. The recent elucidation for the role of the IL-6 signalling pathway in Ang II-induced vascular inflammation as one that controls monocyte activation as well as its diverse signalling mechanism will be reviewed. These new discoveries further our understanding for the important role of the NF-kappaB-IL-6 signalling pathway in the process of vascular inflammation.
Collapse
Affiliation(s)
- Allan R Brasier
- Division of Endocrinology, Department of Internal Medicine, Sealy Center for Molecular Medicine and Institute for Translational Sciences, University of Texas Medical Branch, MRB 8.122, 301 University Blvd, Galveston, TX 77555-1060, USA.
| |
Collapse
|
138
|
Nchimi A, Defawe O, Brisbois D, Broussaud TKY, Defraigne JO, Magotteaux P, Massart B, Serfaty JM, Houard X, Michel JB, Sakalihasan N. MR Imaging of Iron Phagocytosis in Intraluminal Thrombi of Abdominal Aortic Aneurysms in Humans. Radiology 2010; 254:973-81. [DOI: 10.1148/radiol.09090657] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
139
|
Tulamo R, Frösen J, Junnikkala S, Paetau A, Kangasniemi M, Peláez J, Hernesniemi J, Niemelä M, Meri S. Complement system becomes activated by the classical pathway in intracranial aneurysm walls. J Transl Med 2010; 90:168-79. [PMID: 19997064 DOI: 10.1038/labinvest.2009.133] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Inflammation and activation of the complement system in the intracranial aneurysm (IA) wall predispose to IA rupture. We have previously shown that increased C5b-9 accumulation correlates with IA rupture and wall degeneration. To elucidate the underlying mechanisms, we investigated initiators and the pathway of complement activation in unruptured and ruptured IAs. Unruptured and ruptured IA wall samples were studied in parallel sections by immunohistochemical and immunofluorescence stainings for the location and relations of classical and alternative pathway complement components (C1q, C3b/iC3b, C3d, C4b/iC4b; n=35 and properdin, n=10), putative complement activators IgG (n=90), IgM, CRP and OxLDL (n=10), and complement activation endproduct C5b-9. Classical pathway components were seen in all IAs, and they were located mostly in the extracellular matrix. The early pathway complement components colocalized with each other, but were present in larger areas than C5b-9. The areas positive for complement component accumulation were significantly broader in ruptured than in unruptured IAs. The potential complement activators IgG, IgM, CRP and OxLDL were found mostly in the extracellular matrix and in partial overlap with C5b-9. Lipids were seen in Oil-Red-O staining in colocalization with C5b-9. Complement becomes activated by the classical pathway in the IA wall. The activation appears to be induced by multiple factors, which, in addition to the traditional activators (immunoglobulins, CRP, OxLDL), could involve vascular pressure-induced tissue damage. Despite wide early pathway activation, the terminal pathway is focused on a distinct lipid-rich layer. The profile of the complement components and the association of C5b-9 with lipids in the extracellular matrix indicate a long-term chronic inflammatory process rather than an acute targeted inflammatory reaction. The observed pattern of complement activation may be the consequence of local stress-induced insufficiency of complement regulation in IA walls.
Collapse
Affiliation(s)
- Riikka Tulamo
- Neurosurgery Research Group, Biomedicum Helsinki, Helsinki, Huch FI-00029, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Inhibition of renin–angiotensin system attenuates periadventitial inflammation and reduces atherosclerotic lesion formation. Biomed Pharmacother 2009; 63:754-61. [DOI: 10.1016/j.biopha.2009.02.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2008] [Accepted: 02/24/2009] [Indexed: 11/18/2022] Open
|
141
|
Tieu BC, Lee C, Sun H, Lejeune W, Recinos A, Ju X, Spratt H, Guo DC, Milewicz D, Tilton RG, Brasier AR. An adventitial IL-6/MCP1 amplification loop accelerates macrophage-mediated vascular inflammation leading to aortic dissection in mice. J Clin Invest 2009; 119:3637-51. [PMID: 19920349 DOI: 10.1172/jci38308] [Citation(s) in RCA: 370] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 09/23/2009] [Indexed: 12/14/2022] Open
Abstract
Vascular inflammation contributes to cardiovascular diseases such as aortic aneurysm and dissection. However, the precise inflammatory pathways involved have not been clearly defined. We have shown here that subcutaneous infusion of Ang II, a vasopressor known to promote vascular inflammation, into older C57BL/6J mice induced aortic production of the proinflammatory cytokine IL-6 and the monocyte chemoattractant MCP-1. Production of these factors occurred predominantly in the tunica adventitia, along with macrophage recruitment, adventitial expansion, and development of thoracic and suprarenal aortic dissections. In contrast, a reduced incidence of dissections was observed after Ang II infusion into mice lacking either IL-6 or the MCP-1 receptor CCR2. Further analysis revealed that Ang II induced CCR2+CD14hiCD11bhiF4/80- macrophage accumulation selectively in aortic dissections and not in aortas from Il6-/- mice. Adoptive transfer of Ccr2+/+ monocytes into Ccr2-/- mice resulted in selective monocyte uptake into the ascending and suprarenal aorta in regions of enhanced ROS stress, with restoration of IL-6 secretion and increased incidence of dissection. In vitro, coculture of monocytes and aortic adventitial fibroblasts produced MCP-1- and IL-6-enriched conditioned medium that promoted differentiation of monocytes into macrophages, induced CD14 and CD11b upregulation, and induced MCP-1 and MMP-9 expression. These results suggest that leukocyte-fibroblast interactions in the aortic adventitia potentiate IL-6 production, inducing local monocyte recruitment and activation, thereby promoting MCP-1 secretion, vascular inflammation, ECM remodeling, and aortic destabilization.
Collapse
Affiliation(s)
- Brian C Tieu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Dejouvencel T, Féron D, Rossignol P, Sapoval M, Kauffmann C, Piot JM, Michel JB, Fruitier-Arnaudin I, Meilhac O. Hemorphin 7 reflects hemoglobin proteolysis in abdominal aortic aneurysm. Arterioscler Thromb Vasc Biol 2009; 30:269-75. [PMID: 19910633 DOI: 10.1161/atvbaha.109.198309] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE In human abdominal aortic aneurysm, the accumulation of blood-derived cells and proteases within the mural thrombus plays a pivotal role in the evolution toward vessel wall rupture. We sought to identify peptides released from abdominal aortic aneurysm specimens, characterized by an intraluminal thrombus. METHODS AND RESULTS Intraluminal thrombus samples were analyzed by differential proteomics, using surface-enhanced laser desorption/ionization time-of-flight mass spectrometry. A 1309-Da peptide was detected in larger amounts in the newly formed luminal thrombus layer relative to older layers. It was identified as being LVVYPWTQRF (known as LVV-Hemorphin 7), a peptide generated from hemoglobin by cathepsin D. By immunohistochemical analysis, we showed that Hemorphin 7 (H7) colocalizes with cathepsin D and cathepsin G in the luminal layer of the intraluminal thrombus. In vitro, cathepsin G was able to generate H7 peptides at pH 7.4, whereas cathepsin D was only active in acidic conditions. Finally, H7 peptides were shown to be increased 3- to 4-fold in sera of abdominal aortic aneurysm patients relative to controls, and their levels were positively correlated with the volume of the thrombus. CONCLUSIONS Our results suggest that circulating H7 peptides may reflect proteolysis of hemoglobin in the aneurysmal intraluminal thrombus and may be used as a biological marker of pathological vascular remodeling.
Collapse
|
143
|
Witter K, Tonar Z, Matějka VM, Martinča T, Jonák M, Rokošný S, Pirk J. Tissue reaction to three different types of tissue glues in an experimental aorta dissection model: a quantitative approach. Histochem Cell Biol 2009; 133:241-59. [DOI: 10.1007/s00418-009-0656-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2009] [Indexed: 12/14/2022]
|
144
|
Arapoglou V, Kondi-Pafiti A, Rizos D, Kotsis T, Kalkandis C, Katsenis K. The Influence of Total Plasma Homocysteine and Traditional Atherosclerotic Risk Factors on Degree of Abdominal Aortic Aneurysm Tissue Inflammation. Vasc Endovascular Surg 2009; 43:473-9. [DOI: 10.1177/1538574409334345] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Objective: Modulating effects of genetic and environmental risk factors on severity of human abdominal aortic aneurysm (AAA) tissue inflammation remain unclear. We investigated the influence of total plasma homocysteine (tHcy) and traditional atherosclerotic risk factors (ARF) on degree of AAA tissue inflammation. Methods: Aneurysm specimens were obtained from 89 male patients aged 52 to 83 years, underwent asymptomatic not ruptured AAA (mean diameter 5.5 cm) open repair and graded for degree of histologic inflammation. Multivariate analysis was used to determine the association of tHcy and ARF, with degree of inflammation. Results: Current cigarette smoking, odds ratio (OR) 4.4, 95% confidence interval 1.3 to 15.2, P = .01 and no other ARF, neither tHcy levels OR 0.9 (0.9-1.02), P = .2 were associated with high-grade tissue inflammation. Conclusion: These results provide evidence against a major effect of tHcy levels on AAA tissue inflammation, while current cigarette smoking is a significant modulating factor.
Collapse
Affiliation(s)
- Vassilis Arapoglou
- Vascular Surgical Unit, 2nd Surgical Department, Aretaieion Hospital, Medical School, University of Athens, Greece,
| | - Agathi Kondi-Pafiti
- Pathology Laboratory, Aretaieion Hospital, Medical School, University of Athens, Greece
| | - Demetrios Rizos
- Hormone Laboratory, Aretaieion Hospital, Medical School, University of Athens, Greece
| | - Thomas Kotsis
- Vascular Surgical Unit, 2nd Surgical Department, Aretaieion Hospital, Medical School, University of Athens, Greece
| | - Christos Kalkandis
- Vascular Surgical Unit, 2nd Surgical Department, Aretaieion Hospital, Medical School, University of Athens, Greece
| | - Konstantinos Katsenis
- Vascular Surgical Unit, 2nd Surgical Department, Aretaieion Hospital, Medical School, University of Athens, Greece
| |
Collapse
|
145
|
Shi ZD, Ji XY, Qazi H, Tarbell JM. Interstitial flow promotes vascular fibroblast, myofibroblast, and smooth muscle cell motility in 3-D collagen I via upregulation of MMP-1. Am J Physiol Heart Circ Physiol 2009; 297:H1225-34. [PMID: 19465549 DOI: 10.1152/ajpheart.00369.2009] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neointima formation often occurs in regions where the endothelium has been damaged and the transmural interstitial flow is elevated. Vascular smooth muscle cells (SMCs) and fibroblasts/myofibroblasts (FBs/MFBs) contribute to intimal thickening by migrating from the media and adventitia into the site of injury. In this study, for the first time, the direct effects of interstitial flow on SMC and FB/MFB migration were investigated in an in vitro three-dimensional system. Collagen I gels were used to mimic three-dimensional extracellular matrix (ECM) for rat aortic SMCs and FBs/MFBs. Exposure to interstitial flow induced by 1 cmH(2)O pressure differential (shear stress, approximately 0.05 dyn/cm(2); flow velocity, approximately 0.5 microm/s; and Darcy permeability, approximately 10(-11) cm(2)) substantially enhanced cell motility. Matrix metalloproteinase (MMP) inhibitor (GM-6001) abolished flow-induced migration augmentation, which suggested that the enhanced motility was MMP dependent. The upregulation of MMP-1 played a critical role for the flow-enhanced motility, which was further confirmed by silencing MMP-1 gene expression. Longer exposures to higher flows suppressed the number of migrated cells, although MMP-1 gene expression remained high. This suppression was a result of both flow-induced tissue inhibitor of metalloproteinase-1 upregulation and increased apoptotic and necrotic cell death. Interstitial flow did not affect MMP-2 gene expression or activity in the collagen I gel for any cell type. Our findings shed light on the mechanism by which vascular SMCs and FBs/MFBs contribute to intimal thickening in regions of vascular injury where interstitial flow is elevated.
Collapse
Affiliation(s)
- Zhong-Dong Shi
- Department of Biomedical Engineering, City College of New York, City University of New York, New York 10031, USA
| | | | | | | |
Collapse
|
146
|
Adventitia: the vital wall of conduit arteries. ACTA ACUST UNITED AC 2009; 3:166-83. [DOI: 10.1016/j.jash.2009.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 03/05/2009] [Accepted: 03/06/2009] [Indexed: 01/12/2023]
|
147
|
Falk E, Thim T, Kristensen IB. Atherosclerotic Plaque, Adventitia, Perivascular Fat, and Carotid Imaging⁎⁎Editorials published in JACC: Cardiovascular Imaging reflect the views of the authors and do not necessarily represent the views of JACC: Cardiovaswcular Imaging or the American College of Cardiology. JACC Cardiovasc Imaging 2009; 2:183-6. [DOI: 10.1016/j.jcmg.2008.11.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 11/06/2008] [Accepted: 11/13/2008] [Indexed: 11/16/2022]
|
148
|
Skilton MR, Sérusclat A, Sethu AHA, Brun S, Bernard S, Balkau B, Moulin P, Bonnet F. Noninvasive Measurement of Carotid Extra-Media Thickness. JACC Cardiovasc Imaging 2009; 2:176-82. [DOI: 10.1016/j.jcmg.2008.09.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 08/11/2008] [Accepted: 09/09/2008] [Indexed: 10/21/2022]
|
149
|
Houard X, Ollivier V, Louedec L, Michel J, Back M. Differential inflammatory activity across human abdominal aortic aneurysms reveals neutrophilderived leukotriene B4 as a major chemotactic factor released from the intraluminal thrombus. FASEB J 2009; 23:1376-83. [DOI: 10.1096/fj.08-116202] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Xavier Houard
- INSERM U698 Paris 7 Denis Diderot UniversityCardiovascular HematologyBio‐Engineering and RemodelingBichat‐Claude Bernard HospitalParisFrance
| | - Veronique Ollivier
- INSERM U698 Paris 7 Denis Diderot UniversityCardiovascular HematologyBio‐Engineering and RemodelingBichat‐Claude Bernard HospitalParisFrance
| | - Liliane Louedec
- INSERM U698 Paris 7 Denis Diderot UniversityCardiovascular HematologyBio‐Engineering and RemodelingBichat‐Claude Bernard HospitalParisFrance
| | - Jean‐Baptiste Michel
- INSERM U698 Paris 7 Denis Diderot UniversityCardiovascular HematologyBio‐Engineering and RemodelingBichat‐Claude Bernard HospitalParisFrance
| | - Magnus Back
- INSERM U698 Paris 7 Denis Diderot UniversityCardiovascular HematologyBio‐Engineering and RemodelingBichat‐Claude Bernard HospitalParisFrance
- Department of Cardiology and Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| |
Collapse
|
150
|
Dai J, Louedec L, Philippe M, Michel JB, Houard X. Effect of blocking platelet activation with AZD6140 on development of abdominal aortic aneurysm in a rat aneurysmal model. J Vasc Surg 2008; 49:719-27. [PMID: 19028049 DOI: 10.1016/j.jvs.2008.09.057] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Revised: 08/01/2008] [Accepted: 09/27/2008] [Indexed: 12/21/2022]
Abstract
BACKGROUND Platelet activation and thrombus renewal are keys to intraluminal thrombus formation and progression of abdominal aortic aneurysms (AAA). This study explored the ability of AZD6140, a P2Y(12) receptor antagonist, to inhibit platelet activation and prevent aneurysm development in a rat experimental model of AAA. METHOD Aortic aneurysms were induced by implanting a segment of sodium dodecyl sulfate-decellularized guinea pig aorta in rat aortas. One day later, rats were randomized to AZD6140 (10 mg/kg twice daily by mouth) or diluent (n = 23 per group) for either 10 (n = 18) or 42 days (n = 28). Adenosine diphosphate (ADP)-mediated platelet aggregation, aneurysm expansion, intraluminal thrombus formation, inflammatory infiltration, matrix metalloproteinase-9 (MMP-9) expression, and smooth muscle cell colonization were measured. RESULTS AZD6140 inhibited ADP-induced platelet aggregation in vivo for 12 hours, justifying twice-daily administration in rats. The spontaneous increase in aortic diameter shown in the aneurysmal model (2.22 +/- 0.56 mm at day 10 vs 5.21 +/- 1.22 mm at day 42) was reduced with AZD6140 (3.61 +/- 1.46 mm at day 42, P < .01). This beneficial effect was associated with a significant reduction of thrombus development, platelet CD41 expression (P < .05), and leukocyte infiltration of the mural thrombus at days 10 and 42 (P < .01). MMP-9 expression correlated with mural thrombus area and was significantly reduced by AZD6140 (P < .05). AZD6140 limited elastic fiber degradation (P < .05) and enhanced progressive colonization of the thrombus by smooth muscle cells at day 42 (P < .01). CONCLUSIONS These data suggest that inhibition of platelet activation limits intraluminal thrombus biologic activities, thereby impairing aneurysm development.
Collapse
Affiliation(s)
- Jianping Dai
- INSERM U698, Cardiovascular Haematology, Bio-Engineering and Remodeling, Bichat-Claude Bernard Hospital, Paris, France
| | | | | | | | | |
Collapse
|