1
|
Hemmati F, Akinpelu A, Nweze DC, Mistriotis P. 3D confinement alters smooth muscle cell responses to chemical and mechanical cues. APL Bioeng 2024; 8:046103. [PMID: 39464377 PMCID: PMC11512639 DOI: 10.1063/5.0225569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Smooth muscle cell (SMC) phenotypic switching is a hallmark of many vascular diseases. Although prior work has established that chemical and mechanical cues contribute to SMC phenotypic switching, the impact of three-dimensional (3D) confinement on this process remains elusive. Yet, in vivo, arterial SMCs reside within confined environments. In this study, we designed a microfluidic assay to investigate the interplay between 3D confinement and different environmental stimuli in SMC function. Our results show that tightly, but not moderately, confined SMCs acquire a contractile phenotype when exposed to collagen I. Elevated compressive forces induced by hydrostatic pressure abolish this upregulation of the contractile phenotype and compromise SMC survival, particularly in tightly confined spaces. Transforming growth factor beta 1, which promotes the contractile state in moderate confinement, fails to enhance the contractility of tightly confined cells. Fibronectin and engagement of cadherin 2 suppress the contractile phenotype of SMCs regardless of the degree of confinement. In contrast, homophilic engagement of cadherin 11 upregulates SMC-specific genes and enhances contractility in both moderately and tightly confined cells. Overall, our work introduces 3D confinement as a regulator of SMC phenotypic responses to chemical and mechanical signals.
Collapse
Affiliation(s)
- Farnaz Hemmati
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Ayuba Akinpelu
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Daniel Chinedu Nweze
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | | |
Collapse
|
2
|
Pløen GG, Sørensen CB, Bentzon JF. Smooth muscle cells clonally expand in a murine carotid allograft model complicated by immune reactions to reporter transgenes. Transpl Immunol 2024; 87:102129. [PMID: 39260676 DOI: 10.1016/j.trim.2024.102129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/27/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND AIMS Most experimental studies of allograft vasculopathy (AV) have relied on transplantation between major histocompatibility complex-mismatched inbred mouse strains, but this leads to the complete eradication of donor smooth muscle cells (SMCs) and lesions formed by recipient cells. This is unlike human AV which is thought to form mainly by donor SMCs. Here, we studied sources of neointimal cells in a minor histocompatibility antigen-mismatched AV model by combining male-to-female orthotopic carotid transplantations and lineage tracing by SMC-specific expression of fluorescent proteins. METHODS To track SMC-derived cells in allograft vasculopathy, we used male donor mice with SMC-restricted Cre recombination of the mT/mG reporter transgene, which switches expression of membrane-bound red fluorescent protein (RFP) to green fluorescent protein (GFP), or the stochastically recombining Confetti reporter transgene, which yields a mosaic expression of four fluorescent proteins. Donor carotid segments were harvested and orthotopically allografted to female recipients that were wildtype or had non-recombined reporter transgenes. Inhibition of T cell responses by CTLA4Ig was used in some experiments. Sections of lesions harvested after 4 weeks were analyzed by fluorescence microscopy. RESULTS Donor-derived SMCs survived and gave rise to part of the neointimal cells in experiments where carotid segments from recombined mT/mG male mice were transplanted into wild-type or non-recombined mT/mG female mice. Sex-mismatched transplants developed significant lesions, increasing the intimal and medial area 4.6-fold (p = 0.038) and 2.0-fold (p = 0.024) compared to sex- and fluorescence-matched controls, respectively. Interestingly, sex-matched fluorescence-positive transplants developed intimal lesions in 50% of fluorescence-naïve recipient controls. To study the clonal structure of the neointimal donor-derived SMC lineage cells, we then transplanted male carotids with heterozygous or homozygous recombined Confetti transgenes into female recipients. These transplants developed lesions with few surviving donor SMCs, indicating that expression of the Confetti reporter increased rejection and donor-specific SMC death. Some of the few remaining donor SMCs underwent clonal expansion. CTLA4Ig administration at the time of surgery did not improve SMC survival in mT/mG or Confetti transplants. CONCLUSION Male-to-female transplant models feature donor-derived SMCs, some of which undergo clonal expansion, but immune rejection to fluorescence reporters appears to bias results in lineage tracing models. Overcoming these challenges with alternative reporter transgenes or tolerant recipients is necessary to study the mechanisms by which donor SMCs contribute to allograft vasculopathy.
Collapse
Affiliation(s)
| | | | - Jacob Fog Bentzon
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| |
Collapse
|
3
|
Conner AA, David D, Yim EKF. The Effects of Biomimetic Surface Topography on Vascular Cells: Implications for Vascular Conduits. Adv Healthc Mater 2024; 13:e2400335. [PMID: 38935920 DOI: 10.1002/adhm.202400335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/04/2024] [Indexed: 06/29/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality worldwide and represent a pressing clinical need. Vascular occlusions are the predominant cause of CVD and necessitate surgical interventions such as bypass graft surgery to replace the damaged or obstructed blood vessel with a synthetic conduit. Synthetic small-diameter vascular grafts (sSDVGs) are desired to bypass blood vessels with an inner diameter <6 mm yet have limited use due to unacceptable patency rates. The incorporation of biophysical cues such as topography onto the sSDVG biointerface can be used to mimic the cellular microenvironment and improve outcomes. In this review, the utility of surface topography in sSDVG design is discussed. First, the primary challenges that sSDVGs face and the rationale for utilizing biomimetic topography are introduced. The current literature surrounding the effects of topographical cues on vascular cell behavior in vitro is reviewed, providing insight into which features are optimal for application in sSDVGs. The results of studies that have utilized topographically-enhanced sSDVGs in vivo are evaluated. Current challenges and barriers to clinical translation are discussed. Based on the wealth of evidence detailed here, substrate topography offers enormous potential to improve the outcome of sSDVGs and provide therapeutic solutions for CVDs.
Collapse
Affiliation(s)
- Abigail A Conner
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Dency David
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Center for Biotechnology and Bioengineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
4
|
Karabayas M, Ibrahim HE, Roelofs AJ, Reynolds G, Kidder D, De Bari C. Vascular disease persistence in giant cell arteritis: are stromal cells neglected? Ann Rheum Dis 2024; 83:1100-1109. [PMID: 38684323 PMCID: PMC11420755 DOI: 10.1136/ard-2023-225270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Giant cell arteritis (GCA), the most common systemic vasculitis, is characterised by aberrant interactions between infiltrating and resident cells of the vessel wall. Ageing and breach of tolerance are prerequisites for GCA development, resulting in dendritic and T-cell dysfunction. Inflammatory cytokines polarise T-cells, activate resident macrophages and synergistically enhance vascular inflammation, providing a loop of autoreactivity. These events originate in the adventitia, commonly regarded as the biological epicentre of the vessel wall, with additional recruitment of cells that infiltrate and migrate towards the intima. Thus, GCA-vessels exhibit infiltrates across the vascular layers, with various cytokines and growth factors amplifying the pathogenic process. These events activate ineffective repair mechanisms, where dysfunctional vascular smooth muscle cells and fibroblasts phenotypically shift along their lineage and colonise the intima. While high-dose glucocorticoids broadly suppress these inflammatory events, they cause well known deleterious effects. Despite the emerging targeted therapeutics, disease relapse remains common, affecting >50% of patients. This may reflect a discrepancy between systemic and local mediators of inflammation. Indeed, temporal arteries and aortas of GCA-patients can show immune-mediated abnormalities, despite the treatment induced clinical remission. The mechanisms of persistence of vascular disease in GCA remain elusive. Studies in other chronic inflammatory diseases point to the fibroblasts (and their lineage cells including myofibroblasts) as possible orchestrators or even effectors of disease chronicity through interactions with immune cells. Here, we critically review the contribution of immune and stromal cells to GCA pathogenesis and analyse the molecular mechanisms by which these would underpin the persistence of vascular disease.
Collapse
Affiliation(s)
- Maira Karabayas
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Hafeez E Ibrahim
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Anke J Roelofs
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Gary Reynolds
- Centre for Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dana Kidder
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Cosimo De Bari
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
5
|
Yang J, Gao P, Li Q, Wang T, Guo S, Zhang J, Zhang T, Wu G, Guo Y, Wang Z, Tian Y. Arterial Adventitial Vasa Vasorum Hyperplasia involved in Atherosclerotic Plaque Formation in a Rabbit Model. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1273-1279. [PMID: 38796339 DOI: 10.1016/j.ultrasmedbio.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/28/2024]
Abstract
OBJECTIVE It was previously believed that atherosclerotic (AS) plaque starts to develop from the intima and that intraplaque vasa vasorum (VV) hyperplasia promotes adventitial VV (AVV) hyperplasia. However, recent studies have shown that arterial AVV hyperplasia precedes early intimal thickening, suggesting its possible role as an initiating factor of AS. To provide further insight into this process, in this study, we examine the evolution of AAV and VV development in a preclinical model of early AS with longitudinal ultrasound imaging. METHODS Models of early AS were established. Duplex ultrasound scanning and contrast-enhanced ultrasound were performed for diagnosis. Pearson correlation tests were used to analyze the relationships between AVV hyperplasia and VV hyperplasia, or between AVV hyperplasia and intima-media thickness (IMT). RESULTS During 0-12 wk of high-fat feeding, AVV gradually increased and intima-media thickened gradually in the observation area; in the 2nd wk of high-fat feeding, the observation area showed obvious AVV proliferation; at the 4th wk, the intima-media membrane became thicker; at the 12th wk, early plaque formation and intraplaque VV proliferation were observed. There was a strong positive correlation between AVV proliferation and IMT thickening and a strong negative correlation between AVV proliferation and the change rate of vessel diameter. CONCLUSION This study demonstrated that AVV proliferation in the arteries occurred earlier than IMT thickening and was positively correlated with IMT. At present, the indicators of ultrasonic diagnosis of AS, such as IMT, Intraplaque VV, Echo property, all appear in the advanced stage of AS. The AVV may be an innovative diagnostic target for the early stage of AS plaque.
Collapse
Affiliation(s)
- Jiemei Yang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China; Cardiac Ultrasound Division, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Penghao Gao
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Qiannan Li
- Department of General Practice, The Second Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Tengyu Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Shuyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Jingyu Zhang
- Department of Geriatrics, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Tianyi Zhang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Guodong Wu
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Yuanyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China; Department of Geriatrics, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Zeng Wang
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital, Cardiovascular Institute, Harbin Medical University, Harbin, Heilongjiang, PR China.
| |
Collapse
|
6
|
Pushevski V, Dejanov P, Rambabova-Bushljetikj I, Petrusevska G, Popov Z, Ivanovski N. Pathohistomorphometric and Immuno-Histologic Changes in Early Arteriovenous Fistula Failure in Patients with Chronic Kidney Disease. Pril (Makedon Akad Nauk Umet Odd Med Nauki) 2024; 45:13-20. [PMID: 39008640 DOI: 10.2478/prilozi-2024-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
BACKGROUND Hemodialysis is a prevalent treatment for the end-stage chronic kidney disease (CKD) worldwide. The primary arteriovenous fistula (AVF), widely considered the optimal hemodialysis access method, fails to mature in up to two-thirds of the cases. The etiology of the early AVF failure, defined as thrombosis or inability to use within three months post-creation remains less understood, and is influenced by various factors including patient demographics, surgical techniques, and genetic predispositions. Neointimal hyperplasia is a primary histological finding in stenotic lesions leading to the AVF failure. However, there are insufficient data on the cellular phenotypes and the impact of the preexisting CKD-related factors. This study aims to investigate the histological, morphometric, and immunohistochemical alterations in the fistula vein, pre-, peri-, and post-early failure. MATERIALS AND METHODS Eighty-nine stage 4-5 CKD patients underwent standard preoperative assessment, including the Doppler ultrasound, before a typical radio-cephalic AVF creation. Post-failure, a new AVF was created proximally. The vein specimens were collected during the surgery, processed, and analyzed for morphometric analyses and various cellular markers, including Vimentin, TGF, and Ki 67. RESULTS The study enrolled 89 CKD patients, analyzing various aspects of their condition and AVF failures. The histomorphometric analysis revealed substantial venous luminal stenosis and varied endothelial changes. The immunohistologic analysis showed differential marker expressions pre- and post-AVF creation. CONCLUSION This study highlights the complexity of the early AVF failures in CKD patients. The medial hypertrophy emerged as a significant preexisting lesion, while the postoperative analyses indicated a shift towards neointimal hyperplasia. The research underscores the nuanced interplay of vascular remodeling, endothelial damage, and cellular proliferation in the AVF outcomes.
Collapse
Affiliation(s)
- Vladimir Pushevski
- 1University Clinic of Nephrology, Skopje, RN Macedonia
- 2Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, RN Macedonia
| | - Petar Dejanov
- 1University Clinic of Nephrology, Skopje, RN Macedonia
- 2Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, RN Macedonia
| | - Irena Rambabova-Bushljetikj
- 1University Clinic of Nephrology, Skopje, RN Macedonia
- 2Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, RN Macedonia
| | - Gordana Petrusevska
- 2Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, RN Macedonia
- 3Insitutute of Pathology, Skopje, RN Macedonia
| | - Zivko Popov
- 2Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, RN Macedonia
- 4Macedonian Academy of Sciences and Arts, Skopje, RN Macedonia
| | - Ninoslav Ivanovski
- 2Faculty of Medicine, Ss. Cyril and Methodius University, Skopje, RN Macedonia
- 5Zan Mitrev Clinic, Skopje, RN Macedonia
| |
Collapse
|
7
|
Xu S, Jiemy WF, Brouwer E, Burgess JK, Heeringa P, van der Geest KSM, Alba-Rovira R, Corbera-Bellalta M, Boots AH, Cid MC, Sandovici M. Current evidence on the role of fibroblasts in large-vessel vasculitides: From pathogenesis to therapeutics. Autoimmun Rev 2024; 23:103574. [PMID: 38782083 DOI: 10.1016/j.autrev.2024.103574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Large-vessel vasculitides (LVV) comprise a group of chronic inflammatory diseases of the aorta and its major branches. The most common forms of LVV are giant cell arteritis (GCA) and Takayasu arteritis (TAK). Both GCA and TAK are characterized by granulomatous inflammation of the vessel wall accompanied by a maladaptive immune and vascular response that promotes vascular damage and remodeling. The inflammatory process in LVV starts in the adventitia where fibroblasts constitute the dominant cell population. Fibroblasts are traditionally recognized for synthesizing and renewing the extracellular matrix thereby being major players in maintenance of normal tissue architecture and in tissue repair. More recently, fibroblasts have emerged as a highly plastic cell population exerting various functions, including the regulation of local immune processes and organization of immune cells at the site of inflammation through production of cytokines, chemokines and growth factors as well as cell-cell interaction. In this review, we summarize and discuss the current knowledge on fibroblasts in LVV. Furthermore, we identify key questions that need to be addressed to fully understand the role of fibroblasts in the pathogenesis of LVV.
Collapse
Affiliation(s)
- Shuang Xu
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - William F Jiemy
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Elisabeth Brouwer
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Peter Heeringa
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, the Netherlands
| | - Kornelis S M van der Geest
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Roser Alba-Rovira
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Corbera-Bellalta
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Annemieke H Boots
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands
| | - Maria C Cid
- Vasculitis Research Group, Department of Autoimmune Diseases, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Sandovici
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, the Netherlands.
| |
Collapse
|
8
|
Chen YC, Shih CL, Wu CL, Fang YH, So EC, Wu SN. Exploring the Impact of BK Ca Channel Function in Cellular Membranes on Cardiac Electrical Activity. Int J Mol Sci 2024; 25:1537. [PMID: 38338830 PMCID: PMC10855144 DOI: 10.3390/ijms25031537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/14/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
This review paper delves into the current body of evidence, offering a thorough analysis of the impact of large-conductance Ca2+-activated K+ (BKCa or BK) channels on the electrical dynamics of the heart. Alterations in the activity of BKCa channels, responsible for the generation of the overall magnitude of Ca2+-activated K+ current at the whole-cell level, occur through allosteric mechanisms. The collaborative interplay between membrane depolarization and heightened intracellular Ca2+ ion concentrations collectively contribute to the activation of BKCa channels. Although fully developed mammalian cardiac cells do not exhibit functional expression of these ion channels, evidence suggests their presence in cardiac fibroblasts that surround and potentially establish close connections with neighboring cardiac cells. When cardiac cells form close associations with fibroblasts, the high single-ion conductance of these channels, approximately ranging from 150 to 250 pS, can result in the random depolarization of the adjacent cardiac cell membranes. While cardiac fibroblasts are typically electrically non-excitable, their prevalence within heart tissue increases, particularly in the context of aging myocardial infarction or atrial fibrillation. This augmented presence of BKCa channels' conductance holds the potential to amplify the excitability of cardiac cell membranes through effective electrical coupling between fibroblasts and cardiomyocytes. In this scenario, this heightened excitability may contribute to the onset of cardiac arrhythmias. Moreover, it is worth noting that the substances influencing the activity of these BKCa channels might influence cardiac electrical activity as well. Taken together, the BKCa channel activity residing in cardiac fibroblasts may contribute to cardiac electrical function occurring in vivo.
Collapse
Affiliation(s)
- Yin-Chia Chen
- Division of Cardiovascular Surgery, Department of Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Chia-Lung Shih
- Clinical Research Center, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60056, Taiwan
| | - Chao-Liang Wu
- Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Yi-Hsien Fang
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Edmund Cheung So
- Department of Anesthesia, An Nan Hospital, China Medical University, Tainan 70965, Taiwan
| | - Sheng-Nan Wu
- Department of Research and Education, An Nan Hospital, China Medical University, Tainan 70965, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung 80421, Taiwan
| |
Collapse
|
9
|
Lopes E, Machado-Oliveira G, Simões CG, Ferreira IS, Ramos C, Ramalho J, Soares MIL, Melo TMVDPE, Puertollano R, Marques ARA, Vieira OV. Cholesteryl Hemiazelate Present in Cardiovascular Disease Patients Causes Lysosome Dysfunction in Murine Fibroblasts. Cells 2023; 12:2826. [PMID: 38132146 PMCID: PMC10741512 DOI: 10.3390/cells12242826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
There is growing evidence supporting the role of fibroblasts in all stages of atherosclerosis, from the initial phase to fibrous cap and plaque formation. In the arterial wall, as with macrophages and vascular smooth muscle cells, fibroblasts are exposed to a myriad of LDL lipids, including the lipid species formed during the oxidation of their polyunsaturated fatty acids of cholesteryl esters (PUFA-CEs). Recently, our group identified the final oxidation products of the PUFA-CEs, cholesteryl hemiesters (ChE), in tissues from cardiovascular disease patients. Cholesteryl hemiazelate (ChA), the most prevalent lipid of this family, is sufficient to impact lysosome function in macrophages and vascular smooth muscle cells, with consequences for their homeostasis. Here, we show that the lysosomal compartment of ChA-treated fibroblasts also becomes dysfunctional. Indeed, fibroblasts exposed to ChA exhibited a perinuclear accumulation of enlarged lysosomes full of neutral lipids. However, this outcome did not trigger de novo lysosome biogenesis, and only the lysosomal transcription factor E3 (TFE3) was slightly transcriptionally upregulated. As a consequence, autophagy was inhibited, probably via mTORC1 activation, culminating in fibroblasts' apoptosis. Our findings suggest that the impairment of lysosome function and autophagy and the induction of apoptosis in fibroblasts may represent an additional mechanism by which ChA can contribute to the progression of atherosclerosis.
Collapse
Affiliation(s)
- Elizeth Lopes
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| | - Gisela Machado-Oliveira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| | - Catarina Guerreiro Simões
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| | - Inês S. Ferreira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| | - Cristiano Ramos
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| | - José Ramalho
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| | - Maria I. L. Soares
- Coimbra Chemistry Centre (CQC)–Institute of Molecular Sciences and Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal; (M.I.L.S.); (T.M.V.D.P.e.M.)
| | - Teresa M. V. D. Pinho e Melo
- Coimbra Chemistry Centre (CQC)–Institute of Molecular Sciences and Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal; (M.I.L.S.); (T.M.V.D.P.e.M.)
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA;
| | - André R. A. Marques
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| | - Otília V. Vieira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-069 Lisbon, Portugal; (E.L.); (G.M.-O.); (C.G.S.); (I.S.F.); (C.R.); (J.R.)
| |
Collapse
|
10
|
Yan A, Gotlieb AI. The microenvironment of the atheroma expresses phenotypes of plaque instability. Cardiovasc Pathol 2023; 67:107572. [PMID: 37595697 DOI: 10.1016/j.carpath.2023.107572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data from histopathology studies of human atherosclerotic tissue specimens and from vascular imaging studies support the concept that the local arterial microenvironment of a stable atheroma promotes destabilizing conditions that result in the transition to an unstable atheroma. Destabilization is characterized by several different plaque phenotypes that cause major clinical events such as acute coronary syndrome and cerebrovascular strokes. There are several rupture-associated phenotypes causing thrombotic vascular occlusion including simple fibrous cap rupture of an atheroma, fibrous cap rupture at site of previous rupture-and-repair of an atheroma, and nodular calcification with rupture. Endothelial erosion without rupture has more recently been shown to be a common phenotype to promote thrombosis as well. Microenvironment features that are linked to these phenotypes of plaque instability are neovascularization arising from the vasa vasorum network leading to necrotic core expansion, intraplaque hemorrhage, and cap rupture; activation of adventitial and perivascular adipose tissue cells leading to secretion of cytokines, growth factors, adipokines in the outer artery wall that destabilize plaque structure; and vascular smooth muscle cell phenotypic switching through transdifferentiation and stem/progenitor cell activation resulting in the promotion of inflammation, calcification, and secretion of extracellular matrix, altering fibrous cap structure, and necrotic core growth. As the technology evolves, studies using noninvasive vascular imaging will be able to investigate the transition of stable to unstable atheromas in real time. A limitation in the field, however, is that reliable and predictable experimental models of spontaneous plaque rupture and/or erosion are not currently available to study the cell and molecular mechanisms that regulate the conversion of the stable atheroma to an unstable plaque.
Collapse
Affiliation(s)
- Angela Yan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
KhokharVoytas A, Shahbaz M, Maqsood MF, Zulfiqar U, Naz N, Iqbal UZ, Sara M, Aqeel M, Khalid N, Noman A, Zulfiqar F, Al Syaad KM, AlShaqhaa MA. Genetic modification strategies for enhancing plant resilience to abiotic stresses in the context of climate change. Funct Integr Genomics 2023; 23:283. [PMID: 37642792 DOI: 10.1007/s10142-023-01202-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Enhancing the resilience of plants to abiotic stresses, such as drought, salinity, heat, and cold, is crucial for ensuring global food security challenge in the context of climate change. The adverse effects of climate change, characterized by rising temperatures, shifting rainfall patterns, and increased frequency of extreme weather events, pose significant threats to agricultural systems worldwide. Genetic modification strategies offer promising approaches to develop crops with improved abiotic stress tolerance. This review article provides a comprehensive overview of various genetic modification techniques employed to enhance plant resilience. These strategies include the introduction of stress-responsive genes, transcription factors, and regulatory elements to enhance stress signaling pathways. Additionally, the manipulation of hormone signaling pathways, osmoprotectant accumulation, and antioxidant defense mechanisms is discussed. The use of genome editing tools, such as CRISPR-Cas9, for precise modification of target genes related to stress tolerance is also explored. Furthermore, the challenges and future prospects of genetic modification for abiotic stress tolerance are highlighted. Understanding and harnessing the potential of genetic modification strategies can contribute to the development of resilient crop varieties capable of withstanding adverse environmental conditions caused by climate change, thereby ensuring sustainable agricultural productivity and food security.
Collapse
Affiliation(s)
| | - Muhammad Shahbaz
- Department of Botany, University of Agriculture, Faisalabad, Pakistan.
| | | | - Usman Zulfiqar
- Department of Agronomy, Faculty of Agriculture and Environment, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan.
| | - Nargis Naz
- Department of Botany, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Usama Zafar Iqbal
- Department of Botany, University of Agriculture, Faisalabad, Pakistan
| | - Maheen Sara
- Department of Nutritional Sciences, Government College Women University, Faisalabad, Pakistan
| | - Muhammad Aqeel
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems (SKLHIGA), College of Ecology, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Noreen Khalid
- Department of Botany, Government College Women University Sialkot, Sialkot, Pakistan
| | - Ali Noman
- Department of Botany, Government College University, Faisalabad, Pakistan
| | - Faisal Zulfiqar
- Department of Horticultural Sciences, Faculty of Agriculture and Environment, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Khalid M Al Syaad
- Department of Biology, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | | |
Collapse
|
12
|
Kalinin R, Suchkov I, Mzhavanadze N, Surov I. The role of endothelial-mesenchymal transition in vascular restenosis after endovascular treatment in patients with peripheral arterial disease of the lower extremities. RUSSIAN JOURNAL OF CARDIOLOGY AND CARDIOVASCULAR SURGERY 2023; 16:469. [DOI: 10.17116/kardio202316051469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Symptomatic atherosclerosis of the lower extremity arteries is becoming more prevalent each year. Atherosclerotic stenosis or occlusions of femoral and popliteal arteries may often co-exist with aorto-iliac or distal lesions. Treatment options depend on severity of disease, localization of atherosclerotic plaques and comorbidities. The main objective of treating patients with atherosclerotic peripheral artery disease affecting lower extremities is limb salvage and relief of symptoms. Endovascular interventions are able to improve limb salve rates and prognosis of disease, as well as to avoid open and often traumatic reconstructive surgical procedures on the lower extremities. However, postoperative restenosis is still one of the key problems following percutaneous angioplasty and/or stenting. Endothelial mesenchymal transition is regarded as a possible key pathogenetic mechanism underlying restenosis. This article is dedicated to EndMT and its possible role in restenosis.
Collapse
|
13
|
Shao X, Hou X, Zhang X, Zhang R, Zhu R, Qi H, Zheng J, Guo X, Feng R. Integrated single-cell RNA-seq analysis reveals the vital cell types and dynamic development signature of atherosclerosis. Front Physiol 2023; 14:1118239. [PMID: 37089432 PMCID: PMC10117136 DOI: 10.3389/fphys.2023.1118239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/06/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction: In the development of atherosclerosis, the remodeling of blood vessels is a key process involving plaque formation and rupture. So far, most reports mainly believe that macrophages, smooth muscle cells, and endothelial cells located at the intima and media of artery play the key role in this process. Few studies had focused on whether fibroblasts located at adventitia are involved in regulating disease process.Methods and results: In this study, we conducted in-depth analysis of single-cell RNA-seq data of the total of 18 samples from healthy and atherosclerotic arteries. This study combines several analysis methods including transcription regulator network, cell-cell communication network, pseudotime trajectory, gene set enrichment analysis, and differential expression analysis. We found that SERPINF1 is highly expressed in fibroblasts and is involved in the regulation of various signaling pathways.Conclusion: Our research reveals a potential mechanism of atherosclerosis, SERPINF1 regulates the formation and rupture of plaques through the Jak-STAT signaling pathway, which may provide new insights into the pathological study of disease. Moreover, we suggest that SRGN and IGKC as potential biomarkers for unstable arterial plaques.
Collapse
Affiliation(s)
- Xiuli Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiuyang Hou
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiaolin Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ruijia Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Rongli Zhu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - He Qi
- Department of Medical Biotechnology, Liaoning Vocational College of Medicine, Shenyang, China
| | - Jianling Zheng
- Department of Medical Biotechnology, Liaoning Vocational College of Medicine, Shenyang, China
| | - Xiaoling Guo
- Center of Scientific Research, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rui Feng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
- *Correspondence: Rui Feng,
| |
Collapse
|
14
|
Kawabata Y, Wakatsuki T, Yamaguchi K, Fukuda D, Ito H, Matsuura T, Kusunose K, Ise T, Yagi S, Yamada H, Soeki T, Tsuruo Y, Sata M. Association of Microluminal Structures Assessed by Optical Coherence Tomography With Local Inflammation in Adjacent Epicardial Adipose Tissue and Coronary Plaque Characteristics in Fresh Cadavers. Circ J 2023; 87:329-335. [PMID: 36244741 DOI: 10.1253/circj.cj-22-0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Coronary intraplaque microluminal structures (MS) are associated with plaque vulnerability, and the inward progression of vascular inflammation from the adventitia towards the media and intima has also been demonstrated. Therefore, in the present study we investigated the relationships among MS, local inflammation in adjacent epicardial adipose tissue (EAT), and coronary plaque characteristics. METHODS AND RESULTS Optical coherence tomography (OCT) revealed MS in the left anterior descending coronary artery in 10 fresh cadaveric hearts. We sampled 30 lesions and subdivided them based on the presence of MS: MS (+) group (n=19) and MS (-) group (n=11). We measured inflammatory molecule levels in the adjacent EAT and percentage lipid volume assessed by integrated backscatter intravascular ultrasound in each lesion. The expression levels of vascular endothelial growth factor B and C-C motif chemokine ligand 2 were significantly higher in the MS (+) group than in the MS (-) group (0.9±0.7 vs. 0.2±0.2 arbitrary units (AU), P=0.04 and 1.5±0.5 vs. 0.6±0.7 AU, P=0.02, respectively). Percentage lipid volume was significantly higher in the MS (+) group than in the MS (-) group (38.7±16.5 vs. 23.7±10.9%, P=0.03). CONCLUSIONS Intraplaque MS observed on OCT were associated with lipid-rich plaques and local inflammation in the adjacent EAT. Collectively, these results suggest that local inflammation in the EAT is associated with coronary plaque vulnerability via MS.
Collapse
Affiliation(s)
- Yutaka Kawabata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Tetsuzo Wakatsuki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Osaka Metropolitan University Hospital
| | - Hiroyuki Ito
- Department of Community Medicine for Cardiology, Tokushima University Graduate School of Biomedical Sciences
| | - Tomomi Matsuura
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takayuki Ise
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Shusuke Yagi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Hirotsugu Yamada
- Department of Community Medicine for Cardiology, Tokushima University Graduate School of Biomedical Sciences
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences
| | - Yoshihiro Tsuruo
- Department of Anatomy, Tokushima University Graduate School of Biomedical Sciences
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| |
Collapse
|
15
|
Fang Y, Wei H, Wu Z, Song W, Liu C, Li H, Gu C. Short and long-term outcomes after off-pump coronary endarterectomy stratified by different target vessels. J Cardiothorac Surg 2022; 17:339. [PMID: 36567322 PMCID: PMC9791732 DOI: 10.1186/s13019-022-02089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 12/11/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The efficacy of off-pump coronary endarterectomy (CE) has been proven in patients with diffuse coronary artery disease (DCAD). However, the clinical benefits of of-pump CE stratified by different target vessels remain controversial. This retrospective study assessed the effect of the territory and number of CE on short- and long-term outcomes of DCAD. METHODS From January 2012 to December 2014, 246 patients undergoing off-pump coronary artery bypass grafting (OPCABG) + CE were included. The patients were grouped by the territory and number of CE. The primary endpoints were postoperative acute myocardial infarction (PMI) and long-term major adverse cardiovascular and cerebrovascular events (MACCE). RESULTS Sixty-five patients (26.42%) were in the left anterior descending branch (LAD) group (CE on LAD), 134(54.47%) in the right coronary artery (RCA) group (CE on RCA), and 47(19.10%) in the multi-vessels group. PMI in the LAD group, RCA group, and multi-vessels group were 3.08%, 6.72%, and 14.89%, respectively (P = 0.08). Multi-vessels CE (OR = 9.042, 95%CI 2.198-37.193, P = 0.002), CE-plaque length ≥ 3 cm (OR = 6.247, 95%CI 2.162-18.052, P < 0.001), and type 2 diabetes mellitus (2DM) (OR = 4.072, 95%CI 1.598-10.374, P = 0.003) were independent risk factors of PMI. The long-term (mean 76 months) MACCE in the LAD group, RCA group, and multi-vessels group were 13.85%, 17.91%, and 10.64%, respectively (P = 0.552). Cox analysis indicated that PMI (HR = 7.113, 95%CI 3.129-16.171, P < 0.001) and Age ≥ 65 years (HR = 2.488, 95%CI 1.214-5.099, P = 0.013) increased the risk of long-term MACCE. CONCLUSIONS Multi-vessel CE and CE-plaque length ≥ 3 cm significantly increased risk of PMI after OPCABG + CE, but the territory and number of CE did not affect long-term MACCE.
Collapse
Affiliation(s)
- Ying Fang
- grid.411606.40000 0004 1761 5917Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Street No.2 Chaoyang District, Beijing, 100029 China
| | - Hua Wei
- grid.411606.40000 0004 1761 5917Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Street No.2 Chaoyang District, Beijing, 100029 China
| | - Zhen Wu
- grid.411606.40000 0004 1761 5917Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Street No.2 Chaoyang District, Beijing, 100029 China
| | - Wei Song
- grid.411606.40000 0004 1761 5917Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Street No.2 Chaoyang District, Beijing, 100029 China
| | - Changcheng Liu
- grid.411606.40000 0004 1761 5917Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Street No.2 Chaoyang District, Beijing, 100029 China
| | - Haiyang Li
- grid.411606.40000 0004 1761 5917Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Street No.2 Chaoyang District, Beijing, 100029 China
| | - Chengxiong Gu
- grid.411606.40000 0004 1761 5917Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Street No.2 Chaoyang District, Beijing, 100029 China
| |
Collapse
|
16
|
Wang Z, Li G, Li M, Hu L, Hao Z, Li Q, Sun C. Periostin contributes to the adventitial remodeling of atherosclerosis by activating adventitial fibroblasts. ATHEROSCLEROSIS PLUS 2022; 50:57-64. [PMID: 36643802 PMCID: PMC9833252 DOI: 10.1016/j.athplu.2022.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Background and aims Adventitial remodeling is an important pathological process of atherosclerosis, but cues implicated in adventitial remodeling are far from fully understood. Periostin (POSTN), a matricellular protein, has been demonstrated to have multiple roles in cardiovascular diseases. The aim of the study was to explore the function of POSTN in adventitial remodeling during atherosclerosis. Methods An atherosclerosis model was constructed based on ApoE-/- mice fed a high-fat and high-cholesterol diet. The expression of POSTN in the adventitia of mouse atherosclerotic vascular specimens was detected by immunohistochemical staining. The roles of POSTN in regulating adventitial fibroblast activation were assessed by cell contractility and activation marker α-smooth muscle actin (α-SMA) expression evaluation in adventitial fibroblasts overexpressing POSTN. In addition, we performed quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting to examine the expression of the proinflammatory chemokines transforming growth factor-β1 (TGF-β1) and monocyte chemotactic protein 1 (MCP1), as well as some extracellular matrix (ECM)-related proteins, in POSTN-overexpressing adventitial fibroblasts. Finally, the integrin-related signaling pathway was detected upon POSTN overexpression in adventitial fibroblasts. Results POSTN was highly expressed in the adventitia of atherosclerotic aortae in the mouse atherosclerosis model and promoted the activation and contraction of adventitial fibroblasts. Meanwhile, POSTN also induced adventitial fibroblasts to express TGF-β1, monocyte chemotactic protein-1 (MCP1), and ECM-related proteins and activated the phosphorylation of focal adhesion kinase (FAK) and Src. Conclusions Our results revealed that POSTN is elevated in adventitia during atherosclerosis and contributes to the adventitial remodeling of atherosclerosis by activating adventitial fibroblasts.
Collapse
Key Words
- Adventitial fibroblasts
- Adventitial remodeling
- Atherosclerosis
- COL1A1, collagen Ⅰ
- COL3A1, collagen Ⅲ
- DMEM, Dulbecco's modified Eagle's medium
- ECM, extracellular matrix
- FAK, focal adhesion kinase
- FBS, fetal bovine serum
- MCP1, monocyte chemotactic protein-1
- MMPs, matrix metalloproteinases
- POSTN
- POSTN, periostin
- TGF-β1
- TGF-β1, transforming growth factor-β1
- qRT-PCR, quantitative real-time polymerase chain reaction
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Zhonghua Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China,Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Guoliang Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mingpeng Li
- Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Lu Hu
- Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Zichen Hao
- Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Qian Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chaofeng Sun
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China,Corresponding author. Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
17
|
Rizzo C, La Barbera L, Miceli G, Tuttolomondo A, Guggino G. The innate face of Giant Cell Arteritis: Insight into cellular and molecular innate immunity pathways to unravel new possible biomarkers of disease. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:933161. [PMID: 39086970 PMCID: PMC11285707 DOI: 10.3389/fmmed.2022.933161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/11/2022] [Indexed: 08/02/2024]
Abstract
Giant cell arteritis (GCA) is an inflammatory chronic disease mainly occurring in elderly individuals. The pathogenesis of GCA is still far from being completely elucidated. However, in susceptible arteries, an aberrant immune system activation drives the occurrence of vascular remodeling which is mainly characterized by intimal hyperplasia and luminal obstruction. Vascular damage leads to ischemic manifestations involving extra-cranial branches of carotid arteries, mostly temporal arteries, and aorta. Classically, GCA was considered a pathological process resulting from the interaction between an unknown environmental trigger, such as an infectious agent, with local dendritic cells (DCs), activated CD4 T cells and effector macrophages. In the last years, the complexity of GCA has been underlined by robust evidence suggesting that several cell subsets belonging to the innate immunity can contribute to disease development and progression. Specifically, a role in driving tissue damage and adaptive immunity activation was described for dendritic cells (DCs), monocytes and macrophages, mast cells, neutrophils and wall components, such as endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). In this regard, molecular pathways related to cytokines, chemokines, growth factors, vasoactive molecules and reactive oxygen species may contribute to the inflammatory process underlying GCA. Altogether, innate cellular and molecular pathways may clarify many pathogenetic aspects of the disease, paving the way for the identification of new biomarkers and for the development of new treatment targets for GCA. This review aims to deeply dissect past and new evidence on the innate immunological disruption behind GCA providing a comprehensive description of disease development from the innate perspective.
Collapse
Affiliation(s)
- Chiara Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Palermo, Italy
| | - Lidia La Barbera
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Palermo, Italy
| | - Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Unit of Internal Medicine and Stroke Care, University of Palermo, Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Unit of Internal Medicine and Stroke Care, University of Palermo, Palermo, Italy
| | - Giuliana Guggino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Palermo, Italy
| |
Collapse
|
18
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Xiang P, Blanchard V, Francis GA. Smooth Muscle Cell—Macrophage Interactions Leading to Foam Cell Formation in Atherosclerosis: Location, Location, Location. Front Physiol 2022; 13:921597. [PMID: 35795646 PMCID: PMC9251363 DOI: 10.3389/fphys.2022.921597] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Cholesterol-overloaded cells or “foam cells” in the artery wall are the biochemical hallmark of atherosclerosis, and are responsible for much of the growth, inflammation and susceptibility to rupture of atherosclerotic lesions. While it has previously been thought that macrophages are the main contributor to the foam cell population, recent evidence indicates arterial smooth muscle cells (SMCs) are the source of the majority of foam cells in both human and murine atherosclerosis. This review outlines the timeline, site of appearance and proximity of SMCs and macrophages with lipids in human and mouse atherosclerosis, and likely interactions between SMCs and macrophages that promote foam cell formation and removal by both cell types. An understanding of these SMC-macrophage interactions in foam cell formation and regression is expected to provide new therapeutic targets to reduce the burden of atherosclerosis for the prevention of coronary heart disease, stroke and peripheral vascular disease.
Collapse
|
20
|
Clare J, Ganly J, Bursill CA, Sumer H, Kingshott P, de Haan JB. The Mechanisms of Restenosis and Relevance to Next Generation Stent Design. Biomolecules 2022; 12:biom12030430. [PMID: 35327622 PMCID: PMC8945897 DOI: 10.3390/biom12030430] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/04/2023] Open
Abstract
Stents are lifesaving mechanical devices that re-establish essential blood flow to the coronary circulation after significant vessel occlusion due to coronary vessel disease or thrombolytic blockade. Improvements in stent surface engineering over the last 20 years have seen significant reductions in complications arising due to restenosis and thrombosis. However, under certain conditions such as diabetes mellitus (DM), the incidence of stent-mediated complications remains 2–4-fold higher than seen in non-diabetic patients. The stents with the largest market share are designed to target the mechanisms behind neointimal hyperplasia (NIH) through anti-proliferative drugs that prevent the formation of a neointima by halting the cell cycle of vascular smooth muscle cells (VSMCs). Thrombosis is treated through dual anti-platelet therapy (DAPT), which is the continual use of aspirin and a P2Y12 inhibitor for 6–12 months. While the most common stents currently in use are reasonably effective at treating these complications, there is still significant room for improvement. Recently, inflammation and redox stress have been identified as major contributing factors that increase the risk of stent-related complications following percutaneous coronary intervention (PCI). The aim of this review is to examine the mechanisms behind inflammation and redox stress through the lens of PCI and its complications and to establish whether tailored targeting of these key mechanistic pathways offers improved outcomes for patients, particularly those where stent placement remains vulnerable to complications. In summary, our review highlights the most recent and promising research being undertaken in understanding the mechanisms of redox biology and inflammation in the context of stent design. We emphasize the benefits of a targeted mechanistic approach to decrease all-cause mortality, even in patients with diabetes.
Collapse
Affiliation(s)
- Jessie Clare
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Melbourne, VIC 3122, Australia; (J.C.); (J.G.); (P.K.)
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Justin Ganly
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Melbourne, VIC 3122, Australia; (J.C.); (J.G.); (P.K.)
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Christina A. Bursill
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia;
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- ARC Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5000, Australia
| | - Huseyin Sumer
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Melbourne, VIC 3122, Australia; (J.C.); (J.G.); (P.K.)
- Correspondence: (H.S.); (J.B.d.H.)
| | - Peter Kingshott
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Melbourne, VIC 3122, Australia; (J.C.); (J.G.); (P.K.)
- ARC Training Centre in Surface Engineering for Advanced Materials (SEAM), Swinburne University of Technology, Melbourne, VIC 3122, Australia
| | - Judy B. de Haan
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Melbourne, VIC 3122, Australia; (J.C.); (J.G.); (P.K.)
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Correspondence: (H.S.); (J.B.d.H.)
| |
Collapse
|
21
|
Dynamic Crosstalk between Vascular Smooth Muscle Cells and the Aged Extracellular Matrix. Int J Mol Sci 2021; 22:ijms221810175. [PMID: 34576337 PMCID: PMC8468233 DOI: 10.3390/ijms221810175] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/15/2023] Open
Abstract
Vascular aging is accompanied by the fragmentation of elastic fibers and collagen deposition, leading to reduced distensibility and increased vascular stiffness. A rigid artery facilitates elastin to degradation by MMPs, exposing vascular cells to greater mechanical stress and triggering signaling mechanisms that only exacerbate aging, creating a self-sustaining inflammatory environment that also promotes vascular calcification. In this review, we highlight the role of crosstalk between smooth muscle cells and the vascular extracellular matrix (ECM) and how aging promotes smooth muscle cell phenotypes that ultimately lead to mechanical impairment of aging arteries. Understanding the underlying mechanisms and the role of associated changes in ECM during aging may contribute to new approaches to prevent or delay arterial aging and the onset of cardiovascular diseases.
Collapse
|
22
|
Abstract
Abdominal aortic aneurysm (AAA) is a common disease associated with significant cardiovascular morbidity and mortality. Up to now, there is still controversy on the choice of treatment method of AAA. Even so, the mechanisms of AAA progression are poorly defined, making targeting new therapies problematic. Current evidence favors an interaction of the hemodynamic microenvironment with local and systemic immune responses. In this review, we aim to provide an update of mechanisms in AAA progression, involving hemodynamics, perivascular adipose tissue, adventitial fibroblasts, vasa vasorum remodeling, intraluminal thrombus, and distribution of macrophage subtypes.
Collapse
Affiliation(s)
- Jiang-Ping Gao
- Department of Vascular Surgery, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Wei Guo
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
Jolly AJ, Lu S, Strand KA, Dubner AM, Mutryn MF, Nemenoff RA, Majesky MW, Moulton KS, Weiser-Evans MCM. Heterogeneous subpopulations of adventitial progenitor cells regulate vascular homeostasis and pathological vascular remodeling. Cardiovasc Res 2021; 118:1452-1465. [PMID: 33989378 DOI: 10.1093/cvr/cvab174] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases are characterized by chronic vascular dysfunction and provoke pathological remodeling events such as neointima formation, atherosclerotic lesion development, and adventitial fibrosis. While lineage-tracing studies have shown that phenotypically modulated smooth muscle cells (SMCs) are the major cellular component of neointimal lesions, the cellular origins and microenvironmental signaling mechanisms that underlie remodeling along the adventitial vascular layer are not fully understood. However, a growing body of evidence supports a unique population of adventitial lineage-restricted progenitor cells expressing the stem cell marker, stem cell antigen-1 (Sca1; AdvSca1 cells) as important effectors of adventitial remodeling and suggests that they are at least partially responsible for subsequent pathological changes that occur in the media and intima. AdvSca1 cells are being studied in murine models of atherosclerosis, perivascular fibrosis, and neointima formation in response to acute vascular injury. Depending on the experimental conditions, AdvSca1 cells exhibit the capacity to differentiate into SMCs, endothelial cells, chondrocytes, adipocytes, and pro-remodeling cells such as myofibroblasts and macrophages. These data indicate that AdvSca1 cells may be a targetable cell population to influence the outcomes of pathologic vascular remodeling. Important questions remain regarding the origins of AdvSca1 cells and the essential signaling mechanisms and microenvironmental factors that regulate both maintenance of their stem-like, progenitor phenotype and their differentiation into lineage-specified cell types. Adding complexity to the story, recent data indicate that the collective population of adventitial progenitor cells is likely composed of several smaller, lineage-restricted subpopulations which are not fully defined by their transcriptomic profile and differentiation capabilities. The aim of this review is to outline the heterogeneity of Sca1+ adventitial progenitor cells, summarize their role in vascular homeostasis and remodeling, and comment on their translational relevance in humans.
Collapse
Affiliation(s)
- Austin J Jolly
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Sizhao Lu
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Keith A Strand
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Allison M Dubner
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Marie F Mutryn
- Department of Medicine, Division of Renal Diseases and Hypertension
| | - Raphael A Nemenoff
- Department of Medicine, Division of Renal Diseases and Hypertension.,School of Medicine,Consortium for Fibrosis Research and Translation
| | - Mark W Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101.,Departments of Pediatrics and Pathology, University of Washington, Seattle, WA, 98195
| | | | - Mary C M Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension.,School of Medicine,Consortium for Fibrosis Research and Translation.,Cardiovascular Pulmonary Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| |
Collapse
|
24
|
Vazquez-Padron RI, Martinez L, Duque JC, Salman LH, Tabbara M. The anatomical sources of neointimal cells in the arteriovenous fistula. J Vasc Access 2021; 24:99-106. [PMID: 33960241 PMCID: PMC8958841 DOI: 10.1177/11297298211011875] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Neointimal cells are an elusive population with ambiguous origins, functions, and states of differentiation. Expansion of the venous intima in arteriovenous fistula (AVF) is one of the most prominent remodeling processes in the wall after access creation. However, most of the current knowledge about neointimal cells in AVFs comes from extrapolations from the arterial neointima in non-AVF systems. Understanding the origin of neointimal cells in fistulas may have important implications for the design and effective delivery of therapies aimed to decrease intimal hyperplasia (IH). In addition, a broader knowledge of cellular dynamics during postoperative remodeling of the AVF may help clarify other transformation processes in the wall that combined with IH determine the successful remodeling or failure of the access. In this review, we discuss the possible anatomical sources of neointimal cells in AVFs and their relative contribution to intimal expansion.
Collapse
Affiliation(s)
- Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Juan C Duque
- Katz Family Division of Nephrology, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Loay H Salman
- Division of Nephrology, Albany Medical College, Albany, NY, USA
| | - Marwan Tabbara
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
25
|
Parreau S, Vedrenne N, Regent A, Richard L, Sindou P, Mouthon L, Fauchais AL, Jauberteau MO, Ly KH. An immunohistochemical analysis of fibroblasts in giant cell arteritis. Ann Diagn Pathol 2021; 52:151728. [PMID: 33798926 DOI: 10.1016/j.anndiagpath.2021.151728] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 02/26/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Giant cell arteritis (GCA) is a systemic vasculitis of large and medium vessels characterized by an inflammatory arterial infiltrate. GCA begins in the adventitia and leads to vascular remodeling by promoting proliferation of myofibroblasts in the intima. The morphology of the fibroblasts in the adventitia in GCA is unclear. Access to temporal artery biopsies allows morphological studies and evaluation of the microenvironment of the arterial wall. We evaluated the distribution of vascular fibroblasts and of markers of their activation in GCA. METHODS Formalin-fixed paraffin-embedded tissue sections from 29 patients with GCA and 36 controls were examined. Immunohistochemistry was performed for CD90, vimentin, desmin, alpha-smooth muscle actin (ASMA), prolyl-4-hydroxylase (P4H), and myosin to evaluate the distribution of fibroblasts within the intima, media, and adventitia. RESULTS Temporal arteries from patients with GCA showed increased levels of CD90, vimentin, and ASMA in the adventitia and intima compared to the controls. Desmin was expressed only in the media in both groups. P4H was expressed similarly in the adventitia and intima in the two groups. Adventitial and intimal CD90+ cells co-expressed P4H, ASMA, and myosin at a high level in GCA. CONCLUSION The results suggest a role for adventitial fibroblasts in GCA. Inhibiting the differentiation of adventitial fibroblasts to myofibroblasts has therapeutic potential for GCA.
Collapse
Affiliation(s)
- Simon Parreau
- Department of Internal Medicine, Dupuytren Hospital, Limoges, France; EA3842-CaPTuR, Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance thérapeutique, Faculty of Medicine, Limoges, France.
| | - Nicolas Vedrenne
- EA3842-CaPTuR, Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance thérapeutique, Faculty of Medicine, Limoges, France
| | - Alexis Regent
- Department of Internal Medicine, Cochin Hospital, Paris, France
| | | | - Philippe Sindou
- EA3842-CaPTuR, Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance thérapeutique, Faculty of Medicine, Limoges, France
| | - Luc Mouthon
- Department of Internal Medicine, Cochin Hospital, Paris, France
| | - Anne-Laure Fauchais
- Department of Internal Medicine, Dupuytren Hospital, Limoges, France; EA3842-CaPTuR, Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance thérapeutique, Faculty of Medicine, Limoges, France
| | - Marie-Odile Jauberteau
- EA3842-CaPTuR, Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance thérapeutique, Faculty of Medicine, Limoges, France
| | - Kim-Heang Ly
- Department of Internal Medicine, Dupuytren Hospital, Limoges, France; EA3842-CaPTuR, Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance thérapeutique, Faculty of Medicine, Limoges, France
| |
Collapse
|
26
|
Liu J, Chen L, Huang J, Guo S, Zhu D, Gao P. Transient Receptor Potential Melastatin 7 Promotes Vascular Adventitial Fibroblasts Phenotypic Transformation and Inflammatory Reaction Induced by Mechanical Stretching Stress via p38 MAPK/JNK Pathway. J Vasc Res 2021; 58:108-120. [PMID: 33494094 DOI: 10.1159/000512595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/23/2020] [Indexed: 11/19/2022] Open
Abstract
Remodeling of the arteries is one of the pathological bases of hypertension. We have previously shown that transient receptor potential melastatin 7 (TRPM7) aggravates the vascular adventitial remodeling caused by pressure overload in the transverse aortic constriction (TAC) model. In this study, we sought to explore the functional expression and downstream signaling of TRPM7 in vascular adventitial fibroblasts (AFs) stimulated by mechanical stretching stress (MSS). The expression of TRPM7 was upregulated with a concomitant translocation to the cytoplasm in the AFs stimulated with 20% MSS. Meanwhile, the expression of α-smooth muscle actin (α-SMA), a marker of transformation from AFs to myofibroblasts (MFs) was also increased. Moreover, AF-conditioned medium caused a significant migration of macrophages after treatment with MSS and contained high levels of monocyte chemotactic protein-1 (MCP-1), interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-α (TNF-α). Pharmacological and RNA interference approaches using the TRPM7 inhibitor 2-aminoethoxydiphenyl borate (2-APB) and specific anti-TRPM7 small interfering RNA (si-RNA-TRPM7) abrogated these changes significantly. Further exploration uncloaked that inhibition of TRPM7 reduced the phosphorylation of p38 MAP kinase (p38MAPK) and c-Jun N-terminal kinase (JNK) in the AFs stimulated with MSS. Furthermore, inhibition of the phosphorylation of p38MAPK or JNK could also alleviate the MSS-induced expression of α-SMA and secretion of inflammatory factors. These observations indicate that activated TRPM7 participates in the phenotypic transformation and inflammatory action of AFs in response to MSS through the p38MAPK/JNK pathway and suggest that TRPM7 may be a potential therapeutic target for vascular remodeling caused by hemodynamic changes in hypertension.
Collapse
Affiliation(s)
- Jiachen Liu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Laijiang Chen
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Zhejiang, Ningbo, China
| | - Jun Huang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Guo
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,
| | - Dingliang Zhu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pingjin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Bruijn LE, van den Akker BEWM, van Rhijn CM, Hamming JF, Lindeman JHN. Extreme Diversity of the Human Vascular Mesenchymal Cell Landscape. J Am Heart Assoc 2020; 9:e017094. [PMID: 33190596 PMCID: PMC7763765 DOI: 10.1161/jaha.120.017094] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Background Human mesenchymal cells are culprit factors in vascular (patho)physiology and are hallmarked by phenotypic and functional heterogeneity. At present, they are subdivided by classic umbrella terms, such as "fibroblasts," "myofibroblasts," "smooth muscle cells," "fibrocytes," "mesangial cells," and "pericytes." However, a discriminative marker-based subclassification has to date not been established. Methods and Results As a first effort toward a classification scheme, a systematic literature search was performed to identify the most commonly used phenotypical and functional protein markers for characterizing and classifying vascular mesenchymal cell subpopulation(s). We next applied immunohistochemistry and immunofluorescence to inventory the expression pattern of identified markers on human aorta specimens representing early, intermediate, and end stages of human atherosclerotic disease. Included markers comprise markers for mesenchymal lineage (vimentin, FSP-1 [fibroblast-specific protein-1]/S100A4, cluster of differentiation (CD) 90/thymocyte differentiation antigen 1, and FAP [fibroblast activation protein]), contractile/non-contractile phenotype (α-smooth muscle actin, smooth muscle myosin heavy chain, and nonmuscle myosin heavy chain), and auxiliary contractile markers (h1-Calponin, h-Caldesmon, Desmin, SM22α [smooth muscle protein 22α], non-muscle myosin heavy chain, smooth muscle myosin heavy chain, Smoothelin-B, α-Tropomyosin, and Telokin) or adhesion proteins (Paxillin and Vinculin). Vimentin classified as the most inclusive lineage marker. Subset markers did not separate along classic lines of smooth muscle cell, myofibroblast, or fibroblast, but showed clear temporal and spatial diversity. Strong indications were found for presence of stem cells/Endothelial-to-Mesenchymal cell Transition and fibrocytes in specific aspects of the human atherosclerotic process. Conclusions This systematic evaluation shows a highly diverse and dynamic landscape for the human vascular mesenchymal cell population that is not captured by the classic nomenclature. Our observations stress the need for a consensus multiparameter subclass designation along the lines of the cluster of differentiation classification for leucocytes.
Collapse
Affiliation(s)
- Laura E. Bruijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | | | - Connie M. van Rhijn
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jaap F. Hamming
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| | - Jan H. N. Lindeman
- Division of Vascular SurgeryDepartment of SurgeryLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
28
|
Fernández-Colino A, Jockenhoevel S. Advances in Engineering Venous Valves: The Pursuit of a Definite Solution for Chronic Venous Disease. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:253-265. [PMID: 32967586 DOI: 10.1089/ten.teb.2020.0131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Native venous valves enable proper return of blood to the heart. Under pathological conditions (e.g., chronic venous insufficiency), venous valves malfunction and fail to prevent backward flow. Clinically, this can result in painful swelling, varicose veins, edema, and skin ulcerations leading to a chronic wound situation. Surgical correction of venous valves has proven to drastically reduce these symptoms. However, the absence of intact leaflets in many patients limits the applicability of this strategy. In this context, the development of venous valve replacements represents an appealing approach. Despite acceptable results in animal models, no venous valve has succeeded in clinical trials, and so far no single prosthetic venous valve is commercially available. This calls for advanced materials and fabrication approaches to develop clinically relevant venous valves able to restore natural flow conditions in the venous circulation. In this study, we critically discuss the approaches attempted in the last years, and we highlight the potential of tissue engineering to offer new avenues for valve fabrication. Impact statement Venous valves prosthesis offer the potential to restore normal venous flow, and to improve the prospect of patients that suffer from chronic venous disease. Current venous valve replacements are associated with poor outcomes. A deeper understanding of the approaches attempted so far is essential to establish the next steps toward valve development, and importantly, tissue engineering constitutes a unique toolbox to advance in this quest.
Collapse
Affiliation(s)
- Alicia Fernández-Colino
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany.,AMIBM-Aachen-Maastricht-Institute for Biobased Materials, Maastricht University, Geleen, Netherlands
| |
Collapse
|
29
|
Jeong Y, Yao Y, Yim EKF. Current understanding of intimal hyperplasia and effect of compliance in synthetic small diameter vascular grafts. Biomater Sci 2020; 8:4383-4395. [PMID: 32643723 PMCID: PMC7452756 DOI: 10.1039/d0bm00226g] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite much effort, synthetic small diameter vascular grafts still face limited success due to vascular wall thickening known as intimal hyperplasia (IH). Compliance mismatch between graft and native vessels has been proposed to be one of a key mechanical factors of synthetic vascular grafts that could contribute to the formation of IH. While many methods have been developed to determine compliance both in vivo and in vitro, the effects of compliance mismatch still remain uncertain. This review aims to explain the biomechanical factors that are responsible for the formation and development of IH and their relationship with compliance mismatch. Furthermore, this review will address the current methods used to measure compliance both in vitro and in vivo. Lastly, current limitations in understanding the connection between the compliance of vascular grafts and the role it plays in the development and progression of IH will be discussed.
Collapse
Affiliation(s)
- YeJin Jeong
- Department of Chemical engineering, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada.
| | | | | |
Collapse
|
30
|
Wang L, Kikuchi S, Schmidt TA, Hoofnagle M, Wight TN, Azuma N, Tang GL, Sobel M, Velamoor GR, Mokadam NA, Kenagy RD. Inhibitory Effects of PRG4 on Migration and Proliferation of Human Venous Cells. J Surg Res 2020; 253:53-62. [PMID: 32320897 DOI: 10.1016/j.jss.2020.03.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/22/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Proteoglycan 4 (PRG4; lubricin) is a member of two gene co-expression network modules associated with human vein graft failure. However, little is known about PRG4 and the vascular system. Therefore, we have investigated the effects of recombinant human PRG4 (rhPRG4) on cell migration and proliferation in human veins. METHODS Effects of rhPRG4 on cell migration, proliferation, and neointima formation were determined in human venous tissue and cultured venous smooth muscle cells (SMCs), adventitial cells, and endothelial cells. Expression of PRG4 by cultured human saphenous veins, failed vein grafts, and varicose veins was determined by immunostaining or Western blotting. RESULTS Limited expression of PRG4 in fresh saphenous veins was dramatically increased around medial SMCs after culture ex vivo. rhPRG4 inhibited the migration of cultured SMCs, adventitial cells, and endothelial cells, as well as the proliferation of endothelial cells. rhPRG4 also inhibited the migration of SMCs and adventitial cells from tissue explants, but there was no effect on cell proliferation or neointima formation in ex vivo whole veins. Finally, PRG4 was largely absent in two examples of venous pathology, that is, failed human vein grafts and varicose veins. CONCLUSIONS Although rhPRG4 can inhibit the migration of venous SMCs, endothelial cells, and adventitial cells, and the proliferation of endothelial cells, PRG4 was only increased around medial SMCs in veins after ex vivo culture. PRG4 was not observed around medial SMCs in failed human vein grafts and varicose veins, suggesting the possibility that a failure of PRG4 upregulation may promote these pathologies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Vascular Surgery, First Hospital of China Medical University, Shenyang, China
| | - Shinsuke Kikuchi
- Department of Vascular Surgery, Asahikawa Medical University, Asahikawa, Japan
| | | | - Max Hoofnagle
- Department of Surgery, University of Washington, Seattle, Washington
| | - Thomas N Wight
- USA Matrix Biology Program, Benaroya Research Institute, Seattle, Washington
| | - Nobuyoshi Azuma
- Department of Vascular Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Gale L Tang
- Department of Surgery, University of Washington, Seattle, Washington; Center for Cardiovascular Biology and Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Michael Sobel
- Department of Surgery, University of Washington, Seattle, Washington
| | - Gautum R Velamoor
- Department of Surgery, Virginia Mason Medical Center, Seattle, Washington
| | - Nahush A Mokadam
- Department of Surgery, University of Washington, Seattle, Washington
| | - Richard D Kenagy
- Department of Surgery, University of Washington, Seattle, Washington; Center for Cardiovascular Biology and Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
31
|
Chang L, Garcia-Barrio MT, Chen YE. Perivascular Adipose Tissue Regulates Vascular Function by Targeting Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2020; 40:1094-1109. [PMID: 32188271 DOI: 10.1161/atvbaha.120.312464] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adipose tissues are present at multiple locations in the body. Most blood vessels are surrounded with adipose tissue which is referred to as perivascular adipose tissue (PVAT). Similarly to adipose tissues at other locations, PVAT harbors many types of cells which produce and secrete adipokines and other undetermined factors which locally modulate PVAT metabolism and vascular function. Uncoupling protein-1, which is considered as a brown fat marker, is also expressed in PVAT of rodents and humans. Thus, compared with other adipose tissues in the visceral area, PVAT displays brown-like characteristics. PVAT shows a distinct function in the cardiovascular system compared with adipose tissues in other depots which are not adjacent to the vascular tree. Growing and extensive studies have demonstrated that presence of normal PVAT is required to maintain the vasculature in a functional status. However, excessive accumulation of dysfunctional PVAT leads to vascular disorders, partially through alteration of its secretome which, in turn, affects vascular smooth muscle cells and endothelial cells. In this review, we highlight the cross talk between PVAT and vascular smooth muscle cells and its roles in vascular remodeling and blood pressure regulation.
Collapse
Affiliation(s)
- Lin Chang
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor
| | - Minerva T Garcia-Barrio
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor
| | - Y Eugene Chen
- From the Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical School, Ann Arbor
| |
Collapse
|
32
|
Age-Dependent and -Independent Effects of Perivascular Adipose Tissue and Its Paracrine Activities during Neointima Formation. Int J Mol Sci 2019; 21:ijms21010282. [PMID: 31906225 PMCID: PMC6981748 DOI: 10.3390/ijms21010282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/15/2019] [Accepted: 12/29/2019] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular risk factors may act by modulating the composition and function of the adventitia. Here we examine how age affects perivascular adipose tissue (PVAT) and its paracrine activities during neointima formation. Aortic tissue and PVAT or primary aortic smooth muscle cells from male C57BL/6JRj mice aged 52 weeks (“middle-aged”) were compared to tissue or cells from mice aged 16 weeks (“adult”). Vascular injury was induced at the carotid artery using 10% ferric chloride. Carotid arteries from the middle-aged mice exhibited smooth muscle de-differentiation and elevated senescence marker expression, and vascular injury further aggravated media and adventitia thickening. Perivascular transplantation of PVAT had no effect on these parameters, but age-independently reduced neointima formation and lumen stenosis. Quantitative PCR analysis revealed a blunted increase in senescence-associated proinflammatory changes in perivascular tissue compared to visceral adipose tissue and higher expression of mediators attenuating neointima formation. Elevated levels of protein inhibitor of activated STAT1 (PIAS1) and lower expression of STAT1- or NFκB-regulated genes involved in adipocyte differentiation, inflammation, and apoptosis/senescence were present in mouse PVAT, whereas PIAS1 was reduced in the PVAT of patients with atherosclerotic vessel disease. Our findings suggest that age affects adipose tissue and its paracrine vascular activities in a depot-specific manner. PIAS1 may mediate the age-independent vasculoprotective effects of perivascular fat.
Collapse
|
33
|
Wadey K, Lopes J, Bendeck M, George S. Role of smooth muscle cells in coronary artery bypass grafting failure. Cardiovasc Res 2019; 114:601-610. [PMID: 29373656 DOI: 10.1093/cvr/cvy021] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/22/2018] [Indexed: 01/30/2023] Open
Abstract
Atherosclerosis is the underlying pathology of many cardiovascular diseases. The formation and rupture of atherosclerotic plaques in the coronary arteries results in angina and myocardial infarction. Venous coronary artery bypass grafts are designed to reduce the consequences of atherosclerosis in the coronary arteries by diverting blood flow around the atherosclerotic plaques. However, vein grafts suffer a high failure rate due to intimal thickening that occurs as a result of vascular cell injury and activation and can act as 'a soil' for subsequent atherosclerotic plaque formation. A clinically-proven method for the reduction of vein graft intimal thickening and subsequent major adverse clinical events is currently not available. Consequently, a greater understanding of the underlying mechanisms of intimal thickening may be beneficial for the design of future therapies for vein graft failure. Vein grafting induces inflammation and endothelial cell damage and dysfunction, that promotes vascular smooth muscle cell (VSMC) migration, and proliferation. Injury to the wall of the vein as a result of grafting leads to the production of chemoattractants, remodelling of the extracellular matrix and cell-cell contacts; which all contribute to the induction of VSMC migration and proliferation. This review focuses on the role of altered behaviour of VSMCs in the vein graft and some of the factors which critically lead to intimal thickening that pre-disposes the vein graft to further atherosclerosis and re-occurrence of symptoms in the patient.
Collapse
Affiliation(s)
- Kerry Wadey
- Bristol Medical School, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Joshua Lopes
- Translational Biology and Engineering Program, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Michelle Bendeck
- Translational Biology and Engineering Program, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Sarah George
- Bristol Medical School, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| |
Collapse
|
34
|
Intimal regeneration after coronary endarterectomy and onlay grafting in coronary artery bypass grafting. Gen Thorac Cardiovasc Surg 2019; 67:677-683. [PMID: 30790237 DOI: 10.1007/s11748-019-01083-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 02/13/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVES Coronary onlay grafting, with or without endarterectomy, has been widely used for the treatment of diffuse lesions. Recent studies have demonstrated excellent long-term patency and favorable remodeling of onlay anastomosis; however, the underlying mechanisms remain unknown. Here, we describe the mechanism of intimal regeneration based on postmortem pathological evaluation of a patient who had undergone onlay grafting with coronary endarterectomy. METHODS The onlay anastomosis was analyzed using a combination of immunohistological stainings, namely, H&E, vimentin, α-SMA, factor VIII, and Ki-67, to identify the source and mechanism of intimal regeneration after onlay grafting with endarterectomy. RESULTS Our results suggest that the regenerated endothelium derives from the smooth muscle cells of the endarterectomized media of the coronary artery and that it circumferentially covers the internal lumen of the arterial graft. CONCLUSIONS Intimal regeneration, derived from the smooth muscle cells of the endarterectomized coronary artery that proliferate toward the graft lumen, may be a key mechanism that underlies the observed favorable remodeling after onlay grafting during coronary endarterectomy.
Collapse
|
35
|
Mahmoud M, Evans IM, Mehta V, Pellet-Many C, Paliashvili K, Zachary I. Smooth muscle cell-specific knockout of neuropilin-1 impairs postnatal lung development and pathological vascular smooth muscle cell accumulation. Am J Physiol Cell Physiol 2019; 316:C424-C433. [PMID: 30649916 PMCID: PMC6457104 DOI: 10.1152/ajpcell.00405.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropilin 1 (NRP1) is important for neuronal and cardiovascular development due to its role in conveying class 3 semaphorin and vascular endothelial growth factor signaling, respectively. NRP1 is expressed in smooth muscle cells (SMCs) and mediates their migration and proliferation in cell culture and is implicated in pathological SMC remodeling in vivo. To address the importance of Nrp1 for SMC function during development, we generated conditional inducible Nrp1 SMC-specific knockout mice. Induction of early postnatal SMC-specific Nrp1 knockout led to pulmonary hemorrhage associated with defects in alveogenesis and revealed a specific requirement for Nrp1 in myofibroblast recruitment to the alveolar septae and PDGF-AA-induced migration in vitro. Furthermore, SMC-specific Nrp1 knockout inhibited PDGF-BB-stimulated SMC outgrowth ex vivo in aortic ring assays and reduced pathological arterial neointima formation in vivo. In contrast, we observed little significant effect of SMC-specific Nrp1 knockout on neonatal retinal vascularization. Our results point to a requirement of Nrp1 in vascular smooth muscle and myofibroblast function in vivo, which may have relevance for postnatal lung development and for pathologies characterized by excessive SMC and/or myofibroblast proliferation.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Ian M Evans
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Vedanta Mehta
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Caroline Pellet-Many
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Ketevan Paliashvili
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| | - Ian Zachary
- Centre for Cardiovascular Biology and Medicine, BHF Laboratories, Division of Medicine, University College London , London , United Kingdom
| |
Collapse
|
36
|
Wight TN. A role for proteoglycans in vascular disease. Matrix Biol 2018; 71-72:396-420. [PMID: 29499356 PMCID: PMC6110991 DOI: 10.1016/j.matbio.2018.02.019] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/15/2022]
Abstract
The content of proteoglycans (PGs) is low in the extracellular matrix (ECM) of vascular tissue, but increases dramatically in all phases of vascular disease. Early studies demonstrated that glycosaminoglycans (GAGs) including chondroitin sulfate (CS), dermatan sulfate (DS), keratan sulfate (KS) and heparan sulfate (HS) accumulate in vascular lesions in both humans and in animal models in areas of the vasculature that are susceptible to disease initiation (such as at branch points) and are frequently coincident with lipid deposits. Later studies showed the GAGs were covalently attached to specific types of core proteins that accumulate in vascular lesions. These molecules include versican (CSPG), biglycan and decorin (DS/CSPGs), lumican and fibromodulin (KSPGs) and perlecan (HSPG), although other types of PGs are present, but in lesser quantities. While the overall molecular design of these macromolecules is similar, there is tremendous structural diversity among the different PG families creating multiple forms that have selective roles in critical events that form the basis of vascular disease. PGs interact with a variety of different molecules involved in disease pathogenesis. For example, PGs bind and trap serum components that accumulate in vascular lesions such as lipoproteins, amyloid, calcium, and clotting factors. PGs interact with other ECM components and regulate, in part, ECM assembly and turnover. PGs interact with cells within the lesion and alter the phenotypes of both resident cells and cells that invade the lesion from the circulation. A number of therapeutic strategies have been developed to target specific PGs involved in key pathways that promote vascular disease. This review will provide a historical perspective of this field of research and then highlight some of the evidence that defines the involvement of PGs and their roles in the pathogenesis of vascular disease.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, United States.
| |
Collapse
|
37
|
Sobel M, Kikuchi S, Chen L, Tang GL, Wight TN, Kenagy RD. Clinical factors that influence the cellular responses of saphenous veins used for arterial bypass. J Vasc Surg 2018; 68:165S-176S.e6. [PMID: 29914830 DOI: 10.1016/j.jvs.2018.03.436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 03/22/2018] [Indexed: 02/01/2023]
Abstract
OBJECTIVE When an autogenous vein is harvested and used for arterial bypass, it suffers physical and biologic injuries that may set in motion the cellular processes that lead to wall thickening, fibrosis, stenosis, and ultimately graft failure. Whereas the injurious effects of surgical preparation of the vein conduit have been extensively studied, little is known about the influence of the clinical environment of the donor leg from which the vein is obtained. METHODS We studied the cellular responses of fresh saphenous vein samples obtained before implantation in 46 patients undergoing elective lower extremity bypass surgery. Using an ex vivo model of response to injury, we quantified the outgrowth of cells from explants of the adventitial and medial layers of the vein. We correlated this cellular outgrowth with the clinical characteristics of the patients, including the Wound, Ischemia, and foot Infection classification of the donor leg for ischemia, wounds, and infection as well as smoking and diabetes. RESULTS Cellular outgrowth was significantly faster and more robust from the adventitial layer than from the medial layer. The factors of leg ischemia (P < .001), smoking (P = .042), and leg infection (P = .045) were associated with impaired overall outgrowth from the adventitial tissue on multivariable analysis. Only ischemia (P = .046) was associated with impaired outgrowth of smooth muscle cells (SMCs) from the medial tissue. Co-culture of adventitial cells and SMCs propagated from vein explants revealed that adventitial cells significantly inhibited the growth of SMCs, whereas SMCs promoted the growth of adventitial cells. The AA genotype of the -838C>A p27 polymorphism (previously associated with superior graft patency) enhanced these effects, whereas the factor of smoking attenuated adventitial cell inhibition of SMC growth. Comparing gene expression, the cells cultured from the media overexpress Kyoto Encyclopedia of Genes and Genomes pathways associated with inflammation and infection, whereas those from the adventitia overexpress gene families associated with development and stem/progenitor cell maintenance. CONCLUSIONS The adverse clinical environment of the leg may influence the biologic behavior of the cells in the vein wall, especially the adventitial cells. Chronic ischemia was the most significant factor that retards adventitial cell outgrowth. The cells arising from the vein adventitia may be key players in determining a healthy adaptive or a pathologic response to the injuries associated with vein grafting.
Collapse
Affiliation(s)
- Michael Sobel
- Division of Vascular Surgery, VA Puget Sound Health Care System, Seattle, Wash; Division of Vascular Surgery, University of Washington, Seattle, Wash
| | - Shinsuke Kikuchi
- Department of Vascular Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Lihua Chen
- Division of Vascular Surgery, University of Washington, Seattle, Wash
| | - Gale L Tang
- Division of Vascular Surgery, VA Puget Sound Health Care System, Seattle, Wash; Division of Vascular Surgery, University of Washington, Seattle, Wash
| | - Tom N Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, Wash
| | - Richard D Kenagy
- Division of Vascular Surgery, University of Washington, Seattle, Wash.
| |
Collapse
|
38
|
Design and development of a robust photo-responsive block copolymer framework for tunable nucleic acid delivery and efficient gene silencing. Polym J 2018. [DOI: 10.1038/s41428-018-0077-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
39
|
Han X, Wu A, Wang J, Chang H, Zhao Y, Zhang Y, Mao Y, Lou L, Gao Y, Zhang D, Li T, Yang T, Wang L, Feng C, Zhao M. Activation and Migration of Adventitial Fibroblasts Contributes to Vascular Remodeling. Anat Rec (Hoboken) 2018; 301:1216-1223. [PMID: 29406614 DOI: 10.1002/ar.23793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/03/2017] [Accepted: 12/07/2017] [Indexed: 12/26/2022]
Abstract
The rat carotid artery balloon injury model was used to prove the activation and migration of adventitial fibroblasts. We found that at day 7 after injury, adventitial fibroblasts proliferated, transformed into myofibroblasts under transmission electron microscopy in the model group. Simultaneously, we proved that the adventitial cells migrated to the media and intima on seventh day after injury by directly labeled the adventitial cells by the in vivo gene transfer technique. Moreover, we captured the precise moment when the adventitial fibroblasts migrated from the adventitia to the media through the external elastic plate under transmission electron microscope. This study provides direct evidences that adventitial fibroblasts activate and migrate to the media and intima, then actively take part in revascularization. Anat Rec, 301:1216-1223, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaowan Han
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Aiming Wu
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Jie Wang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China.,Huayuanlu Community Health Service Center, Beijing 100088, China
| | - Hong Chang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Yizhou Zhao
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Yan Zhang
- Department of Ultrapathology of the Neurosurgical Institute Affiliated Bayi Brain Hospital, Army General Hospital of PLA, Beijing 100700, China
| | - Yingqiu Mao
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lixia Lou
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Yonghong Gao
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China
| | - Dongmei Zhang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Tong Li
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Tao Yang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Lei Wang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Cuiling Feng
- Peking University People's Hospital, Beijing, China
| | - Mingjing Zhao
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| |
Collapse
|
40
|
Deng Y, Lin C, Zhou HJ, Min W. Smooth muscle cell differentiation: Mechanisms and models for vascular diseases. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s11515-017-1473-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Souilhol C, Harmsen MC, Evans PC, Krenning G. Endothelial–mesenchymal transition in atherosclerosis. Cardiovasc Res 2018; 114:565-577. [DOI: 10.1093/cvr/cvx253] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Celine Souilhol
- Department of Infection, Immunity & Cardiovascular Disease (IICD), Faculty of Medicine, Dentistry & Health, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| | - Martin C Harmsen
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ Groningen, The Netherlands
| | - Paul C Evans
- Department of Infection, Immunity & Cardiovascular Disease (IICD), Faculty of Medicine, Dentistry & Health, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ Groningen, The Netherlands
| |
Collapse
|
42
|
Fitzpatrick E, Han X, Liu W, Corcoran E, Burtenshaw D, Morrow D, Helt JC, Cahill PA, Redmond EM. Alcohol Reduces Arterial Remodeling by Inhibiting Sonic Hedgehog-Stimulated Stem Cell Antigen-1 Positive Progenitor Stem Cell Expansion. Alcohol Clin Exp Res 2017; 41:2051-2065. [PMID: 28921619 DOI: 10.1111/acer.13499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/11/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cell and molecular mechanisms mediating the cardiovascular effects of alcohol are not fully understood. Our aim was to determine the effect of moderate ethanol (EtOH) on sonic hedgehog (SHh) signaling in regulating possible stem cell antigen-1 positive (Sca1+ ) progenitor stem cell involvement during pathologic arterial remodeling. METHODS Partial ligation or sham operation of the left carotid artery was performed in transgenic Sca1-enhanced green fluorescent protein (eGFP) mice gavaged with or without "daily moderate" EtOH. RESULTS The EtOH group had reduced adventitial thickening and less neointimal formation, compared to ligated controls. There was expansion of eGFP-expressing (i.e., Sca1+ ) cells in remodeled vessels postligation (day 14), especially in the neo intima. EtOH treatment reduced the number of Sca1+ cells in ligated vessel cross-sections concomitant with diminished remodeling, compared to control ligated vessels. Moreover, EtOH attenuated SHh signaling in injured carotids as determined by immunohistochemical analysis of the target genes patched 1 and Gli2, and RT-PCR of whole-vessel Gli2 mRNA levels. Intraperitoneal injection of ligated Sca1-eGFP mice with the SHh signaling inhibitor cyclopamine diminished SHh target gene expression, reduced the number of Sca1+ cells, and ameliorated carotid remodeling. EtOH treatment of purified Sca1+ adventitial progenitor stem cells in vitro inhibited SHh signaling, and their rSHh-induced differentiation to vascular smooth muscle cells. CONCLUSIONS EtOH reduces SHh-responsive Sca1+ progenitor cell myogenic differentiation/expansion in vitro and during arterial remodeling in response to ligation injury in vivo. Regulation of vascular Sca1+ progenitor cells in this way may be an important novel mechanism contributing to alcohol's cardiovascular protective effects.
Collapse
Affiliation(s)
- Emma Fitzpatrick
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Xu Han
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Weimin Liu
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Eoin Corcoran
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Denise Burtenshaw
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - David Morrow
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Jay-Christian Helt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| | - Paul A Cahill
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Eileen M Redmond
- Department of Surgery, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
43
|
Greco CT, Akins RE, Epps TH, Sullivan MO. Attenuation of Maladaptive Responses in Aortic Adventitial Fibroblasts through Stimuli-Triggered siRNA Release from Lipid-Polymer Nanocomplexes. ADVANCED BIOSYSTEMS 2017; 1:1700099. [PMID: 29392169 PMCID: PMC5788321 DOI: 10.1002/adbi.201700099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lipid-siRNA assemblies are modified with photo-responsive polymers to enable spatiotemporally-controlled silencing of interleukin 1 beta (IL1β) and cadherin 11 (CDH11), two genes that are essential drivers of maladaptive responses in human aortic adventitial fibroblasts (AoAFs). These hybrid nanocomplexes address the critical challenge of locally mitigating fibrotic actions that lead to the high rates of vascular graft failures. In particular, the lipid-polymer formulations provide potent silencing of IL1β and CDH11 that is precisely modulated by a photo-release stimulus. Moreover, a dynamic modeling framework is used to design a multi-dose siRNA regimen that sustains knockdown of both genes over clinically-relevant timescales. Multi-dose suppression illuminates a cooperative role for IL1β and CDH11 in pathogenic adventitial remodeling and is directly linked to desirable functional outcomes. Specifically, myofibroblast differentiation and cellular proliferation, two of the primary hallmarks of fibrosis, are significantly attenuated by IL1β silencing. Meanwhile, the effects of CDH11 siRNA treatment on differentiation become more pronounced at higher cell densities characteristic of constrictive adventitial remodeling in vivo. Thus, this work offers a unique formulation design for photo-responsive gene suppression in human primary cells and establishes a new dosing method to satisfy the critical need for local attenuation of fibrotic responses in the adventitium surrounding vascular grafts.
Collapse
Affiliation(s)
- Chad T Greco
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Robert E Akins
- Department of Biomedical Research, Nemours - Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Thomas H Epps
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
44
|
Kuwabara JT, Tallquist MD. Tracking Adventitial Fibroblast Contribution to Disease: A Review of Current Methods to Identify Resident Fibroblasts. Arterioscler Thromb Vasc Biol 2017; 37:1598-1607. [PMID: 28705796 DOI: 10.1161/atvbaha.117.308199] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/23/2017] [Indexed: 01/18/2023]
Abstract
Cells present in the adventitia, or outermost layer of the blood vessel, contribute to the progression of vascular diseases, such as atherosclerosis, hypertension, and aortic dissection. The adventitial fibroblast of the aorta is the prototypic perivascular fibroblast, but the adventitia is composed of multiple distinct cell populations. Therefore, methods for uniquely identifying the fibroblast are critical for a better understanding of how these cells contribute to disease processes. A popular method for distinguishing adventitial cell types relies on the use of genetic tools in the mouse to trace and manipulate these cells. Because lineage tracing relying on Cre-recombinase expressing mice is used more frequently in studies of vascular disease, it is important to outline the advantages and limitations of these genetic tools. The purpose of this article is to provide an overview of the various genetic tools available in the mouse for the study of resident adventitial fibroblasts.
Collapse
Affiliation(s)
- Jill T Kuwabara
- From the Center for Cardiovascular Research, University of Hawaii, Honolulu
| | | |
Collapse
|
45
|
Perivascular adipose tissue: epiphenomenon or local risk factor? Int J Obes (Lond) 2017; 41:1311-1323. [PMID: 28529328 DOI: 10.1038/ijo.2017.121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 04/24/2017] [Accepted: 05/01/2017] [Indexed: 02/08/2023]
|
46
|
Scott RA, Kharkar PM, Kiick KL, Akins RE. Aortic adventitial fibroblast sensitivity to mitogen activated protein kinase inhibitors depends on substrate stiffness. Biomaterials 2017; 137:1-10. [PMID: 28527302 DOI: 10.1016/j.biomaterials.2017.05.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/25/2017] [Accepted: 05/07/2017] [Indexed: 11/18/2022]
Abstract
Adventitial fibroblasts (AFs) are key determinants of arterial function and critical mediators of arterial disease progression. The effects of altered stiffness, particularly those observed across individuals during normal vascular function, and the mechanisms by which AFs respond to altered stiffness, are not well understood. To study the effects of matrix stiffness on AF phenotype, cytokine production, and the regulatory pathways utilized to interpret basic cell-matrix interactions, human aortic AFs were grown in 5%, 7.5%, and 10% (w/v%) PEG-based hydrogels with Young's moduli of 1.2, 3.3, and 9.6 kPa, respectively. In 5% gels, AFs had higher proliferation rates, elevated monocyte chemoattractant protein-1 secretion, and enhanced monocyte recruitment. Significantly more AFs were α-smooth muscle actin positive in 7.5% gels, indicating myofibroblast development. AFs in 10% gels had low proliferation rates but produced high levels of interleukin-6 and vascular endothelial growth factor-A. Importantly, these modulus-dependent changes in AF phenotype were accompanied by alterations in the mitogen-activated protein kinase (MAPK) pathways contributing to the production of cytokines. These data indicate that complex cell regulatory changes occur with altered tissue stiffness and suggest that therapeutics affecting MAPK pathways may have altered effects on AFs depending on substrate stiffness.
Collapse
Affiliation(s)
- Rebecca A Scott
- Nemours - Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA; Delaware Biotechnology Institute, Newark, DE 19711, USA
| | - Prathamesh M Kharkar
- Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA
| | - Kristi L Kiick
- Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA; Delaware Biotechnology Institute, Newark, DE 19711, USA
| | - Robert E Akins
- Nemours - Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Materials Science & Engineering, University of Delaware, Newark, DE 19716, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
47
|
Mylonaki I, Allémann É, Saucy F, Haefliger JA, Delie F, Jordan O. Perivascular medical devices and drug delivery systems: Making the right choices. Biomaterials 2017; 128:56-68. [PMID: 28288349 DOI: 10.1016/j.biomaterials.2017.02.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/13/2017] [Accepted: 02/26/2017] [Indexed: 12/31/2022]
Abstract
Perivascular medical devices and perivascular drug delivery systems are conceived for local application around a blood vessel during open vascular surgery. These systems provide mechanical support and/or pharmacological activity for the prevention of intimal hyperplasia following vessel injury. Despite abundant reports in the literature and numerous clinical trials, no efficient perivascular treatment is available. In this review, the existing perivascular medical devices and perivascular drug delivery systems, such as polymeric gels, meshes, sheaths, wraps, matrices, and metal meshes, are jointly evaluated. The key criteria for the design of an ideal perivascular system are identified. Perivascular treatments should have mechanical specifications that ensure system localization, prolonged retention and adequate vascular constriction. From the data gathered, it appears that a drug is necessary to increase the efficacy of these systems. As such, the release kinetics of pharmacological agents should match the development of the pathology. A successful perivascular system must combine these optimized pharmacological and mechanical properties to be efficient.
Collapse
Affiliation(s)
- Ioanna Mylonaki
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, rue Michel Servet 1, CH-1211 Geneva 4, Switzerland
| | - Éric Allémann
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, rue Michel Servet 1, CH-1211 Geneva 4, Switzerland
| | - François Saucy
- Department of Vascular Surgery, Lausanne University Hospital, rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Jacques-Antoine Haefliger
- Department of Vascular Surgery, Lausanne University Hospital, rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - Florence Delie
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, rue Michel Servet 1, CH-1211 Geneva 4, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, rue Michel Servet 1, CH-1211 Geneva 4, Switzerland.
| |
Collapse
|
48
|
Gerbay A, Terreaux J, Cerisier A, Vola M, Isaaz K. Impact of very high pressure stent deployment on angiographic and long-term clinical outcomes in true coronary bifurcation lesions treated by the mini-crush stent technique: A single center experience. Indian Heart J 2017; 69:32-36. [PMID: 28228303 PMCID: PMC5318982 DOI: 10.1016/j.ihj.2016.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/28/2016] [Accepted: 05/21/2016] [Indexed: 01/21/2023] Open
Abstract
Background Percutaneous coronary intervention (PCI) for bifurcation lesions (BL) using 2 stents technique is known to be associated with high rates of procedural failure especially on the side branch (SB) mainly due to stent incomplete apposition. Stent deployment at very high pressure (SDHP) may lead to better stent expansion and apposition. However, SDHP may also be at the origin of deeper wall injury resulting into major cardiac adverse events. No data are available on evaluation of SDHP in BL treated by a mini-crush stent technique. Methods One hundred and thirteen consecutive patients underwent PCI for BL (Medina 1, 1, 1) using a mini-crush stent technique with SDHP defined as ≥20 atm. An angiographic follow-up was performed at 6 month and clinical follow-up was obtained at a median of 3 years. Results Stent deployment mean pressures were 20 ± 1.4 atm (range 20–25) in the main vessel (MV) and 20 ± 1.5 atm (range 20–25) in SB. Simultaneous final kissing balloon was used in 92% of cases. PCI was successful in 100%. Angiographic follow-up was obtained in 83% of patients. Restenosis rate was 13% (12% restenosis in the SB) with only one case (0.8%) of SB probable thrombosis. Another case of late stent thrombosis occurred at a 3 years clinical follow-up. Conclusion Compared with previously published studies in which stents were deployed at lower pressure, SDHP does not increase the restenosis rate in BL using mini-crush stent technique but seems to reduce the rate of stent thrombosis.
Collapse
Affiliation(s)
- Antoine Gerbay
- Division of Cardiology, University of Saint Etienne, Saint Etienne, France.
| | - Jeremy Terreaux
- Division of Cardiology, University of Saint Etienne, Saint Etienne, France
| | - Alexis Cerisier
- Division of Cardiology, University of Saint Etienne, Saint Etienne, France
| | - Marco Vola
- Division of Cardiology, University of Saint Etienne, Saint Etienne, France
| | - Karl Isaaz
- Division of Cardiology, University of Saint Etienne, Saint Etienne, France
| |
Collapse
|
49
|
Boire TC, Balikov DA, Lee Y, Guth CM, Cheung-Flynn J, Sung HJ. Biomaterial-Based Approaches to Address Vein Graft and Hemodialysis Access Failures. Macromol Rapid Commun 2016; 37:1860-1880. [PMID: 27673474 PMCID: PMC5156561 DOI: 10.1002/marc.201600412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/15/2016] [Indexed: 12/19/2022]
Abstract
Veins used as grafts in heart bypass or as access points in hemodialysis exhibit high failure rates, thereby causing significant morbidity and mortality for patients. Interventional or revisional surgeries required to correct these failures have been met with limited success and exorbitant costs, particularly for the US Centers for Medicare & Medicaid Services. Vein stenosis or occlusion leading to failure is primarily the result of neointimal hyperplasia. Systemic therapies have achieved little long-term success, indicating the need for more localized, sustained, biomaterial-based solutions. Numerous studies have demonstrated the ability of external stents to reduce neointimal hyperplasia. However, successful results from animal models have failed to translate to the clinic thus far, and no external stent is currently approved for use in the US to prevent vein graft or hemodialysis access failures. This review discusses current progress in the field, design considerations, and future perspectives for biomaterial-based external stents. More comparative studies iteratively modulating biomaterial and biomaterial-drug approaches are critical in addressing mechanistic knowledge gaps associated with external stent application to the arteriovenous environment. Addressing these gaps will ultimately lead to more viable solutions that prevent vein graft and hemodialysis access failures.
Collapse
Affiliation(s)
- Timothy C Boire
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
| | - Daniel A Balikov
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
| | - Yunki Lee
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
| | - Christy M Guth
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37235, USA
| | - Joyce Cheung-Flynn
- Division of Vascular Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, 37235, USA
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, TN, USA
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul, 120-752, Republic of Korea
| |
Collapse
|
50
|
Chandiwal A, Balasubramanian V, Baldwin ZK, Conte MS, Schwartz LB. Gene Therapy for the Extension of Vein Graft Patency: A Review. Vasc Endovascular Surg 2016; 39:1-14. [PMID: 15696243 DOI: 10.1177/153857440503900101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The mainstay of treatment for long-segment small-vessel chronic occlusive disease not amenable to endovascular intervention remains surgical bypass grafting using autologous vein. The procedure is largely successful and the immediate operative results almost always favorable. However, the lifespan of a given vein graft is highly variable, and less than 50% will remain primarily patent after 5 years. The slow process of graft malfunction is a result of the vein's chronic maladaptive response to the systemic arterial environment, its primary component being the uncontrolled proliferation of vascular smooth muscle cells (SMCs). It has recently been suggested that this response might be attenuated through pre-implantation genetic modification of the vein, so-called gene therapy for the extension of vein graft patency. Gene therapy seems particularly well suited for the prevention or postponement of vein graft failure since: (1) the stimulation of SMC proliferation appears to largely be an early and transient process, matching the kinetics of current gene transfer technology; (2) most veins are relatively normal and free of disease at the time of bypass allowing for effective gene transfer using a variety of systems; and (3) the target tissue is directly accessible during operation because manipulation and irrigation of the vein is part of the normal workflow of the surgical procedure. This review briefly summarizes the current knowledge of the incidence and basic mechanisms of vein graft failure, the vector systems and molecular targets that have been proposed as possible pre-treatments, the results of experimental genetic modification of vein grafts, and the few available clinical studies of gene therapy for vascular proliferative disorders.
Collapse
Affiliation(s)
- Amito Chandiwal
- Section of Vascular Surgery, Department of Surgery, University of Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|