1
|
Guo X, Liu S, Wu X, Yang R, Ren Q, Zhou Y, Shi K, Yuan L, Zhang N, Liu S. Alleviating vascular calcification with Bushen Huoxue formula in rats with chronic kidney disease by inhibiting the PTEN/PI3K/AKT signaling pathway through exosomal microRNA-32. J Pharm Pharmacol 2024:rgae120. [PMID: 39440885 DOI: 10.1093/jpp/rgae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 08/29/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Vascular calcification (VC) significantly raises cardiovascular mortality in chronic kidney disease (CKD) patients. VC is characterized by the phenotypic transformation of vascular smooth muscle cells (VSMCs) to osteoblast-like cells, mediated by exosomes derived from calcified VSMCs and the exosomal microRNAs (miRNA) which may trigger some signals to recipient VSMCs. Bushen Huoxue (BSHX) formula has demonstrated its clinical efficacy in CKD and its protective role in CKD-VC rats has also been observed. However, little is known about its underlying mechanism. METHODS To establish a VC model, aortic VSMCs from rats were induced to osteogenic differentiation by high-level phosphate (HP) in vitro. The expression of exosome and calcification makers were analyzed by western blot, including CD9, CD63, α-SMA, BMP-2, and Runx2, respectively. Differential expression of exosomal miRNAs in normal and HP-induced VSMCs were identified by using whole miRNA microarray technology. GO and KEGG analyses were performed to determine the significant enrichment of functions and signaling pathways in the target genes. In vivo, the CKD-VC rat model was established by administering adenine gavage combined with a high phosphorus diet. The rats were divided into normal control, model, low-dose BSHX, medium-dose BSHX, high-dose BSHX groups, and sevelamer groups. The blood biochemical parameters were measured. Renal histopathology and aortic calcification were observed. Western blot detected the levels of the calcification markers. Quantitative real-time PCR (qPCR) assay detected exosomal microRNA-32 (miR-32) mRNA expression in the aorta, the most differentially expressed exosomal miRNA previously identified. Phosphatase and tensin homolog located on chromosome ten (PTEN)/phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT) signaling pathway components were also tested by western blot. RESULTS Exosomal miRNA-32 and PI3K/AKT signaling pathways were highly differentially expressed between normal and HP-induced VSMCs. In vivo, BSHX improved blood biochemical parameters, renal histopathology, and aortic calcification in CKD-VC rats. BSHX increased the expression level of α-SMA and decreased the level of BMP-2 and Runx2. BSHX also lowered the expression level of exosomal miR-32 mRNA, enhanced PTEN expression, therefore, reduced p-PI3K and p-AKT levels in the aorta. CONCLUSION BSHX alleviated VC in CKD rats by downregulating exosomal miR-32 expression in the aorta, thereby promoting PTEN expression and inhibiting the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xingyun Guo
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Fever Outpatient Clinic, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Shiwei Liu
- Department of Nephrology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Xiaoyi Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Ronglu Yang
- Department of Traditional Chinese Medicine, The First Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Qiuyue Ren
- Department of Endocrinology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450003, China
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100007, China
| | - Kaifeng Shi
- Department of Nephrology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Lisha Yuan
- Department of Nephrology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ning Zhang
- Department of Nephrology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Shiyi Liu
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
2
|
Liu B, Xu G, Li H, Lu G, Ning N, Tang Q. Excessive collagen fiber deposition in idiopathic scrotal calcinosis: a case report. BMC Urol 2024; 24:212. [PMID: 39363259 PMCID: PMC11448437 DOI: 10.1186/s12894-024-01601-w] [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: 05/08/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Idiopathic scrotal calcinosis (ISC) is a manifestation of idiopathic calcinosis cutis, and its etiology is still unknown. CASE PRESENTATION We report a 36-year-old patient manifested multiple gradually increasing yellowish-white scrotal nodules with occasional itching and stinging in the past 6 years and was successfully cured via surgical excision. The laboratory test combined with pathological analysis confirmed the diagnosis of ISC. Like pathological calcinosis in other soft tissues, a large amount of collagen fiber deposition was observed around the calcification nodule, suggesting that abnormal collagen fiber deposition might be an important factor leading to idiopathic calcinosis in the scrotum. Moreover, koilocytes, which indicate human papillomavirus (HPV) infection, were also detected around calcified nodules, indicating the potential pathogenic role of HPV infection in ISC. CONCLUSIONS Here, we report that ISC shows abnormal excessive deposition of collagen fibers around calcified nodules, which may be a vital factor contributing to the disease. Furthermore, combined with the literature review, a new pathogenic mechanism of ISC is proposed, and the site specificity of scrotal calcinosis is explained, providing a basis for further exploration of the pathogenic mechanism of ISC.
Collapse
Affiliation(s)
- Bo Liu
- Department of Human Anatomy, Histology and Embryology, Fourth Military Medical University, Xi'an, 710032, China
- Department of Urology, Central Theater Air Force Hospital, Datong, 037000, China
| | - Gongquan Xu
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Hao Li
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Guocheng Lu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Ning Ning
- Department of Pathology, Northwest Women's and Children's Hospital, Xi'an, 710061, Shaanxi Province, China.
| | - Qisheng Tang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
3
|
Son JH, Park JK, Bang JH, Kim D, Moon I, Kong MG, Park HW, Choi HO, Seo HS, Cho YH, Chang HS, Suh J. Exosomal miRNAs Differentiate Chronic Total Occlusion from Acute Myocardial Infarction. Int J Mol Sci 2024; 25:10223. [PMID: 39337705 PMCID: PMC11432064 DOI: 10.3390/ijms251810223] [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: 08/30/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Although coronary artery occlusion can have a negative effect on the myocardium, chronic total occlusion (CTO) exhibits different clinical features from those of acute myocardial infarction (AMI). In this study, we identify the differential associations of exosomal miRNAs with CTO and AMI. Exosomes were isolated from the plasma obtained from coronary arteries of patients undergoing percutaneous coronary intervention to treat CTO (n = 29) and AMI (n = 24), followed by small RNA sequencing, target gene predictions, and functional enrichment analyses. Promising miRNA markers were validated using real-time PCR in 35 CTO, 35 AMI, and 10 normal subjects. A total of 205 miRNAs were detected in all subjects, and 20 and 12 miRNAs were upregulated and downregulated in CTO compared to AMI patients, respectively (|fold change| > 4, FDR q < 0.05). The target genes of miRNAs that were higher in CTO patients were associated with "regulation of cell cycle phase transition", "cell growth", and "apoptosis". The target genes of miRNAs that were lower in CTO patients were enriched in terms such as "muscle cell differentiation", "response to oxygen levels", and "artery morphogenesis". On qRT-PCR analysis, the expression levels of miR-9-5p and miR-127-3p were significantly different between CTO and AMI patients. The miRNA expression levels accurately distinguished CTO from AMI patients with 79% specificity and 97% sensitivity. The miRNA contents of plasma exosomes were significantly different between CTO and AMI patients. The miRNAs may play important roles in CTO and AMI.
Collapse
Affiliation(s)
- Ji-Hye Son
- Department of Interdisciplinary Program in Biomedical Science Major, Graduate School, Soonchunhyang University, Asan 31538, Republic of Korea; (J.-H.S.); (J.-H.B.)
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea
| | - Jeong Kyu Park
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Ji-Hong Bang
- Department of Interdisciplinary Program in Biomedical Science Major, Graduate School, Soonchunhyang University, Asan 31538, Republic of Korea; (J.-H.S.); (J.-H.B.)
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea
| | - Dongeon Kim
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Inki Moon
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Min Gyu Kong
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Hyun-Woo Park
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Hyung-Oh Choi
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Hye-Sun Seo
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Yoon Haeng Cho
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| | - Hun Soo Chang
- Department of Interdisciplinary Program in Biomedical Science Major, Graduate School, Soonchunhyang University, Asan 31538, Republic of Korea; (J.-H.S.); (J.-H.B.)
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 33151, Republic of Korea
| | - Jon Suh
- Divisions of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea; (J.K.P.); (D.K.); (I.M.); (M.G.K.); (H.-W.P.); (H.-O.C.); (H.-S.S.); (Y.H.C.)
| |
Collapse
|
4
|
Clift CL, Blaser MC, Gerrits W, Turner ME, Sonawane A, Pham T, Andresen JL, Fenton OS, Grolman JM, Campedelli A, Buffolo F, Schoen FJ, Hjortnaes J, Muehlschlegel JD, Mooney DJ, Aikawa M, Singh SA, Langer R, Aikawa E. Intracellular proteomics and extracellular vesiculomics as a metric of disease recapitulation in 3D-bioprinted aortic valve arrays. SCIENCE ADVANCES 2024; 10:eadj9793. [PMID: 38416823 PMCID: PMC10901368 DOI: 10.1126/sciadv.adj9793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
In calcific aortic valve disease (CAVD), mechanosensitive valvular cells respond to fibrosis- and calcification-induced tissue stiffening, further driving pathophysiology. No pharmacotherapeutics are available to treat CAVD because of the paucity of (i) appropriate experimental models that recapitulate this complex environment and (ii) benchmarking novel engineered aortic valve (AV)-model performance. We established a biomaterial-based CAVD model mimicking the biomechanics of the human AV disease-prone fibrosa layer, three-dimensional (3D)-bioprinted into 96-well arrays. Liquid chromatography-tandem mass spectrometry analyses probed the cellular proteome and vesiculome to compare the 3D-bioprinted model versus traditional 2D monoculture, against human CAVD tissue. The 3D-bioprinted model highly recapitulated the CAVD cellular proteome (94% versus 70% of 2D proteins). Integration of cellular and vesicular datasets identified known and unknown proteins ubiquitous to AV calcification. This study explores how 2D versus 3D-bioengineered systems recapitulate unique aspects of human disease, positions multiomics as a technique for the evaluation of high throughput-based bioengineered model systems, and potentiates future drug discovery.
Collapse
Affiliation(s)
- Cassandra L Clift
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark C Blaser
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Willem Gerrits
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mandy E Turner
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Abhijeet Sonawane
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tan Pham
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jason L Andresen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Owen S Fenton
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joshua M Grolman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
- Materials Science and Engineering, The Technion-Israel Institute of Technology, Haifa, Israel
| | - Alesandra Campedelli
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Fabrizio Buffolo
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Internal Medicine and Hypertension Unite, Department of Medical Sciences, University of Torin, Turin, Italy
| | - Frederick J Schoen
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jesper Hjortnaes
- Department of Cardiothoracic Surgery, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Masanori Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sasha A Singh
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Onnis C, Virmani R, Kawai K, Nardi V, Lerman A, Cademartiri F, Scicolone R, Boi A, Congiu T, Faa G, Libby P, Saba L. Coronary Artery Calcification: Current Concepts and Clinical Implications. Circulation 2024; 149:251-266. [PMID: 38227718 PMCID: PMC10794033 DOI: 10.1161/circulationaha.123.065657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Coronary artery calcification (CAC) accompanies the development of advanced atherosclerosis. Its role in atherosclerosis holds great interest because the presence and burden of coronary calcification provide direct evidence of the presence and extent of coronary artery disease; furthermore, CAC predicts future events independently of concomitant conventional cardiovascular risk factors and to a greater extent than any other noninvasive biomarker of this disease. Nevertheless, the relationship between CAC and the susceptibility of a plaque to provoke a thrombotic event remains incompletely understood. This review summarizes the current understanding and literature on CAC. It outlines the pathophysiology of CAC and reviews laboratory, histopathological, and genetic studies, as well as imaging findings, to characterize different types of calcification and to elucidate their implications. Some patterns of calcification such as microcalcification portend increased risk of rupture and cardiovascular events and may improve prognosis assessment noninvasively. However, contemporary computed tomography cannot assess early microcalcification. Limited spatial resolution and blooming artifacts may hinder estimation of degree of coronary artery stenosis. Technical advances such as photon counting detectors and combination with nuclear approaches (eg, NaF imaging) promise to improve the performance of cardiac computed tomography. These innovations may speed achieving the ultimate goal of providing noninvasively specific and clinically actionable information.
Collapse
Affiliation(s)
- Carlotta Onnis
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari – Polo di Monserrato s.s. 554 Monserrato (Cagliari) 09045, ITALY
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, 19 Firstfield Road, Gaithersburg, MD
| | - Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, 19 Firstfield Road, Gaithersburg, MD
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | | | - Roberta Scicolone
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari – Polo di Monserrato s.s. 554 Monserrato (Cagliari) 09045, ITALY
| | - Alberto Boi
- Department of Cardiology, Azienda Ospedaliera Brotzu, Cagliari Italy
| | - Terenzio Congiu
- Department of Pathology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari – Ospedale San Giovanni di Dio (Cagliari) 09100 ITALY
| | - Gavino Faa
- Department of Pathology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari – Ospedale San Giovanni di Dio (Cagliari) 09100 ITALY
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), di Cagliari – Polo di Monserrato s.s. 554 Monserrato (Cagliari) 09045, ITALY
| |
Collapse
|
6
|
Liu Y, An Y, Li G, Wang S. Regulatory mechanism of macrophage polarization based on Hippo pathway. Front Immunol 2023; 14:1279591. [PMID: 38090595 PMCID: PMC10715437 DOI: 10.3389/fimmu.2023.1279591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Macrophages are found to infiltrate and migrate in a large number of Tumor-associated macrophages (TMEs) and other macrophages in the microenvironment of tumors and related diseases, and undergo phenotypic changes in response to a variety of cytokines, mainly including the primary phenotype M2 and the anti-tumor phenotype M1. The Hippo signaling pathway affects the development of cancer and other diseases through various biological processes, such as inhibition of cell growth. In this review, we focus on immune cells within the microenvironment of tumors and other diseases, and the role of the Hippo pathway in tumors on macrophage polarization in the tumor microenvironment (TME) and other diseases.
Collapse
Affiliation(s)
- Yuanqing Liu
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yina An
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Gebin Li
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shuaiyu Wang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Center of Research and Innovation of Chinese Traditional Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
7
|
Su G, Zhang D, Li T, Pei T, Yang J, Tu S, Liu S, Ren J, Zhang Y, Duan M, Yang X, Shen Y, Zhou C, Xie J, Liu X. Annexin A5 derived from matrix vesicles protects against osteoporotic bone loss via mineralization. Bone Res 2023; 11:60. [PMID: 37940665 PMCID: PMC10632518 DOI: 10.1038/s41413-023-00290-9] [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: 04/24/2023] [Revised: 07/23/2023] [Accepted: 08/31/2023] [Indexed: 11/10/2023] Open
Abstract
Matrix vesicles (MVs) have shown strong effects in diseases such as vascular ectopic calcification and pathological calcified osteoarthritis and in wound repair of the skeletal system due to their membranous vesicle characteristics and abundant calcium and phosphorus content. However, the role of MVs in the progression of osteoporosis is poorly understood. Here, we report that annexin A5, an important component of the matrix vesicle membrane, plays a vital role in bone matrix homeostasis in the deterioration of osteoporosis. We first identified annexin A5 from adherent MVs but not dissociative MVs of osteoblasts and found that it could be sharply decreased in the bone matrix during the occurrence of osteoporosis based on ovariectomized mice. We then confirmed its potential in mediating the mineralization of the precursor osteoblast lineage via its initial binding with collagen type I to achieve MV adhesion and the subsequent activation of cellular autophagy. Finally, we proved its protective role in resisting bone loss by applying it to osteoporotic mice. Taken together, these data revealed the importance of annexin A5, originating from adherent MVs of osteoblasts, in bone matrix remodeling of osteoporosis and provided a new strategy for the treatment and intervention of bone loss.
Collapse
Affiliation(s)
- Guanyue Su
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Tiantian Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Tong Pei
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jie Yang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Shasha Tu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Sijun Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jie Ren
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yaojia Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xinrui Yang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, Liu J. Vascular calcification: from the perspective of crosstalk. MOLECULAR BIOMEDICINE 2023; 4:35. [PMID: 37851172 PMCID: PMC10584806 DOI: 10.1186/s43556-023-00146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
Vascular calcification (VC) is highly correlated with cardiovascular disease morbidity and mortality, but anti-VC treatment remains an area to be tackled due to the ill-defined molecular mechanisms. Regardless of the type of VC, it does not depend on a single cell but involves multi-cells/organs to form a complex cellular communication network through the vascular microenvironment to participate in the occurrence and development of VC. Therefore, focusing only on the direct effect of pathological factors on vascular smooth muscle cells (VSMCs) tends to overlook the combined effect of other cells and VSMCs, including VSMCs-VSMCs, ECs-VMSCs, Macrophages-VSMCs, etc. Extracellular vesicles (EVs) are a collective term for tiny vesicles with a membrane structure that are actively secreted by cells, and almost all cells secrete EVs. EVs docked on the surface of receptor cells can directly mediate signal transduction or transfer their contents into the cell to elicit a functional response from the receptor cells. They have been proven to participate in the VC process and have also shown attractive therapeutic prospects. Based on the advantages of EVs and the ability to be detected in body fluids, they may become a novel therapeutic agent, drug delivery vehicle, diagnostic and prognostic biomarker, and potential therapeutic target in the future. This review focuses on the new insight into VC molecular mechanisms from the perspective of crosstalk, summarizes how multi-cells/organs interactions communicate via EVs to regulate VC and the emerging potential of EVs as therapeutic methods in VC. We also summarize preclinical experiments on crosstalk-based and the current state of clinical studies on VC-related measures.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qian Chen
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Xie
- Department of Orthopaedics, Movement System Injury and Repair Research Centre, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
9
|
Huang C, Han X, Yang L, Song W, Zhang H, Zhu X, Huang G, Xu J. Exosomal miR-129 and miR-342 derived from intermittent hypoxia-stimulated vascular smooth muscle cells inhibit the eIF2α/ATF4 axis from preventing calcified aortic valvular disease. J Cell Commun Signal 2023:10.1007/s12079-023-00785-4. [PMID: 37812275 DOI: 10.1007/s12079-023-00785-4] [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: 04/20/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
This study aims to elucidate the role of miR-129/miR-342 loaded in exosomes derived from vascular smooth muscle cells (VSMCs) stimulated by intermittent hypoxia in calcified aortic valvular disease (CAVD). Bioinformatics analysis was conducted to identify differentially expressed miRs in VSMCs-derived exosomes and CAVD samples, and their potential target genes were predicted. VSMCs were exposed to intermittent hypoxia to induce stimulation, followed by isolation of exosomes. Valvular interstitial cells (VICs) were cultured in vitro to investigate the impact of miR-129/miR-342 on VICs' osteogenic differentiation and aortic valve calcification with eIF2α. A CAVD mouse model was established using ApoE knockout mice for in vivo validation. In CAVD samples, miR-129 and miR-342 were downregulated, while eIF2α and ATF4 were upregulated. miR-129 and miR-342 exhibited inhibitory effects on eIF2α through targeted regulation. Exosomes released from intermittently hypoxia-stimulated VSMCs contained miR-129 and miR-342. Overexpression of miR-129 and miR-342, or silencing ATF4, suppressed VICs' osteogenic differentiation and aortic valve calcification, which could be rescued by overexpressed eIF2α. Collectively, intermittent hypoxia stimulation of VSMCs leads to the secretion of exosomes that activate the miR-129/miR-342 dual pathway, thereby inhibiting the eIF2α/ATF4 axis and attenuating VICs' osteogenic differentiation and CAVD progression.
Collapse
Affiliation(s)
- Chen Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Xu Han
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Linjie Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Wei Song
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Hualu Zhang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaohua Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Gongcheng Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Jing Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
10
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
11
|
Wal P, Rathore S, Aziz N, Singh YK, Gupta A. Aortic stenosis: a review on acquired pathogenesis and ominous combination with diabetes mellitus. Egypt Heart J 2023; 75:26. [PMID: 37027109 PMCID: PMC10082141 DOI: 10.1186/s43044-023-00345-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Aortic stenosis (AS) is a progressive disease, with no pharmacological treatment. The prevalence of diabetes mellitus (DM) among AS patients is higher than in the general population. DM significantly increases the risk of AS development and progression from mild to severe. The interplay between AS and DM's mechanism is not entirely known yet. MAIN BODY The increased accumulation of advanced glycation end products (AGEs) was linked to increased valvular oxidative stress, inflammation, expression of coagulation factors, and signs of calcification, according to an analysis of aortic stenotic valves. It is interesting to note that in diabetic AS patients, valvular inflammation did not correlate with serum glucose levels but rather only with long-term glycemic management markers like glycated haemoglobin and fructosamine. Transcatheter aortic valve replacement, which has been shown to be safer than surgical aortic valve replacement, is advantageous for AS patients who also have concurrent diabetes. Additionally, novel anti-diabetic medications have been proposed to lower the risk of AS development in DM patients, including sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonist that target reduction of AGEs-mediated oxidative stress. CONCLUSIONS There are little data on the effects of hyperglycemia on valvular calcification, but understanding the interactions between them is essential to develop a successful treatment strategy to stop or at least slow the progression of AS in DM patients. There is a link among AS and DM and that DM negatively impacts the quality of life and longevity of AS patients. The sole successful treatment, despite ongoing efforts to find new therapeutic modalities, involves aortic valve replacement. More research is required to find methods that can slow the advancement of these conditions, enhancing the prognosis and course of people with AS and DM.
Collapse
Affiliation(s)
- Pranay Wal
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur, UP, 209305, India.
| | - Shruti Rathore
- LCIT School of Pharmacy, Bilaspur, Chhattisgarh, 495220, India
| | - Namra Aziz
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur, UP, 209305, India
| | - Yash Kumar Singh
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur, UP, 209305, India
| | - Arpit Gupta
- Pranveer Singh Institute of Technology (Pharmacy), Bhauti, Kanpur, UP, 209305, India
| |
Collapse
|
12
|
Hosen MR, Goody PR, Zietzer A, Xiang X, Niepmann ST, Sedaghat A, Tiyerili V, Chennupati R, Moore JB, Boon RA, Uchida S, Sinning JM, Zimmer S, Latz E, Werner N, Nickenig G, Jansen F. Circulating MicroRNA-122-5p Is Associated With a Lack of Improvement in Left Ventricular Function After Transcatheter Aortic Valve Replacement and Regulates Viability of Cardiomyocytes Through Extracellular Vesicles. Circulation 2022; 146:1836-1854. [PMID: 35862223 DOI: 10.1161/circulationaha.122.060258] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/14/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Transcatheter aortic valve replacement (TAVR) is a well-established treatment option for high- and intermediate-risk patients with severe symptomatic aortic valve stenosis. A majority of patients exhibit improvements in left ventricular ejection fraction (LVEF) after TAVR in response to TAVR-associated afterload reduction. However, a specific role for circulating microRNAs (miRNAs) in the improvement of cardiac function for patients after TAVR has not yet been investigated. Here, we profiled the differential expression of miRNAs in circulating extracellular vesicles (EVs) in patients after TAVR and, in particular, the novel role of circulating miR-122-5p in cardiomyocytes. METHODS Circulating EV-associated miRNAs were investigated by use of an unbiased Taqman-based human miRNA array. Several EV miRNAs (miR-122-5p, miR-26a, miR-192, miR-483-5p, miR-720, miR-885-5p, and miR-1274) were significantly deregulated in patients with aortic valve stenosis at day 7 after TAVR compared with the preprocedural levels in patients without LVEF improvement. The higher levels of miR-122-5p were negatively correlated with LVEF improvement at both day 7 (r=-0.264 and P=0.015) and 6 months (r=-0.328 and P=0.0018) after TAVR. RESULTS Using of patient-derived samples and a murine aortic valve stenosis model, we observed that the expression of miR-122-5p correlates negatively with cardiac function, which is associated with LVEF. Mice with graded wire injury-induced aortic valve stenosis demonstrated a higher level of miR-122-5p, which was related to cardiomyocyte dysfunction. Murine ex vivo experiments revealed that miR-122-5p is highly enriched in endothelial cells compared with cardiomyocytes. Coculture experiments, copy-number analysis, and fluorescence microscopy with Cy3-labeled miR-122-5p demonstrated that miR-122-5p can be shuttled through large EVs from endothelial cells into cardiomyocytes. Gain- and loss-of-function experiments suggested that EV-mediated shuttling of miR-122-5p increases the level of miR-122-5p in recipient cardiomyocytes. Mechanistically, mass spectrometry, miRNA pulldown, electrophoretic mobility shift assay, and RNA immunoprecipitation experiments confirmed that miR-122-5p interacts with the RNA-binding protein hnRNPU (heterogeneous nuclear ribonucleoprotein U) in a sequence-specific manner to encapsulate miR-122-5p into large EVs. On shuttling, miR-122-5p reduces the expression of the antiapoptotic gene BCL2 by binding to its 3' untranslated region to inhibit its translation, thereby decreasing the viability of target cardiomyocytes. CONCLUSIONS Increased levels of circulating proapoptotic EV-incorporated miR-122-5p are associated with reduced LVEF after TAVR. EV shuttling of miR-122-5p regulates the viability and apoptosis of cardiomyocytes in a BCL2-dependent manner.
Collapse
Affiliation(s)
- Mohammed Rabiul Hosen
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Philip Roger Goody
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Andreas Zietzer
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Xu Xiang
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
- Department of International Medical Center, Affiliated Hospital of Qingdao University, Shinan, Qingdao, Shandong, China (X.X.)
| | - Sven Thomas Niepmann
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Alexander Sedaghat
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Vedat Tiyerili
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Ramesh Chennupati
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Germany (R.C.)
| | - Joseph B Moore
- Christina Lee Brown Environment Institute, Department of Medicine, University of Louisville, KY (J.B.M.)
- Diabetes and Obesity Center, Louisville, KY (J.B.M.)
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Germany (R.A.B.)
- Center for Cardiovascular Research (DZHK), Partner Site-Rhein-Main, Frankfurt am Main, Germany (R.A.B.)
- Department of Physiology, VU University Medical Center, Amsterdam, the Netherlands (R.A.B.)
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark (S.U.)
| | - Jan-Malte Sinning
- Department of Internal Medicine-III-Cardiology, St. Vinzenz Hospital, Cologne, Germany (J.-M.S.)
| | - Sebastian Zimmer
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Eicke Latz
- Institute of Innate Immunity (E.L.), University Hospital Bonn, Venusberg-Campus Germany
| | - Nikos Werner
- Department of Internal Medicine/ Cardiology, Krankenhaus der Barmherzigen Brüder Trier, Germany (N.W.)
| | - Georg Nickenig
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| | - Felix Jansen
- Heart Center Bonn, Department of Internal Medicine II (M.R.H., P.R.G., A.Z., X.X., S.T.N., A.S., V.T., S.Z., G.N., F.J.), University Hospital Bonn, Venusberg-Campus Germany
| |
Collapse
|
13
|
Xia H, Gao M, Chen J, Huang G, Xiang X, Wang Y, Huang Z, Li Y, Su S, Zhao Z, Zeng Q, Ruan Y. M1 macrophage-derived extracellular vesicle containing tsRNA-5006c promotes osteogenic differentiation of aortic valve interstitial cells through regulating mitophagy. PeerJ 2022; 10:e14307. [PMID: 36518291 PMCID: PMC9744173 DOI: 10.7717/peerj.14307] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background Osteogenic differentiation of aortic valve interstitial cells (AVICs) plays a key role in the calcific aortic valve disease progression. Extracellular vesicles (EVs)-derived from M1-polarized macrophages (M1-EVs) orchestrated intercellular communication by delivering non-coding RNAs such as tRNA-derived small RNAs (tsRNAs) is crucial for cardiovascular disease. However, the role and mechanism of M1-EVs tsRNAs in osteogenic differentiation of AVICs remains largely unclear. Methods M1-EVs and PBS treated-RAW 264.7 cell-derived EVs (NC-EVs) were incubated with AVICs and subjected to small RNA sequencing. Candidate tsRNA in M1-EVs was silenced to explore their effects on AVIC osteogenic differentiation and mitophagy. Results DiI-labeled M1-EVs were internalized by AVICs, resulting in significantly increased calcium nodule formation and expression of osteogenesis-related genes in AVICs, including RUNX2, BMP2, osteopontin, and SPP1, compared with NC-EVs. Small RNA sequencing revealed that 17 tsRNAs were significantly up-regulated such as tsRNA-5006c, while 28 tsRNAs were significantly down-regulated in M1-EVs compared with NC-EVs. Intriguingly, tsRNA-5006c-deleted M1-EVs treatment significantly reduced calcium nodule formation and expression of osteogenesis-related genes in AVICs relative to control group. Moreover, target genes of tsRNA-5006c were mainly involved in autophagy-related signaling pathways, such as MAPK, Ras, Wnt, and Hippo signaling pathway. Hallmarks of mitophagy activation in AVICs including mitophagosome formation, TMRM fluorescence, expression of LC3-II, BINP3, and PGC1α, were significantly elevated in the M1-EVs group compared with NC-EVs group, whereas M1-EVs tsRNA-5006c inhibitor led to a significant reduction in these indicators. Conclusion M1-EVs carried tsRNA-5006c regulates AVIC osteogenic differentiation from the perspective of mitophagy, and we provide a new target for the prevention and treatment of aortic valve calcification.
Collapse
Affiliation(s)
- Hao Xia
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingjian Gao
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Chen
- Department of Cardiology, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, China
| | - Guanshen Huang
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiuting Xiang
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuyan Wang
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhaohui Huang
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongchun Li
- Department of traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuang Su
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zewei Zhao
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qingchun Zeng
- Department of Cardiology, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, China
| | - Yunjun Ruan
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Role of Collagen in Vascular Calcification. J Cardiovasc Pharmacol 2022; 80:769-778. [PMID: 35998017 DOI: 10.1097/fjc.0000000000001359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/03/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Vascular calcification is a pathological process characterized by ectopic calcification of the vascular wall. Medial calcifications are most often associated with kidney disease, diabetes, hypertension, and advanced age. Intimal calcifications are associated with atherosclerosis. Collagen can regulate mineralization by binding to apatite minerals and promoting their deposition, binding to collagen receptors to initiate signal transduction, and inducing cell transdifferentiation. In the process of vascular calcification, type I collagen is not only the scaffold for mineral deposition but also a signal entity, guiding the distribution, aggregation, and nucleation of vesicles and promoting the transformation of vascular smooth muscle cells into osteochondral-like cells. In recent years, collagen has been shown to affect vascular calcification through collagen disc-domain receptors, matrix vesicles, and transdifferentiation of vascular smooth muscle cells.
Collapse
|
15
|
Turner ME, Bartoli‐Leonard F, Aikawa E. Small particles with large impact: Insights into the unresolved roles of innate immunity in extracellular vesicle‐mediated cardiovascular calcification. Immunol Rev 2022; 312:20-37. [DOI: 10.1111/imr.13134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mandy E Turner
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Francesca Bartoli‐Leonard
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine Department of Medicine Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
- Division of Cardiovascular Medicine Department of Medicine Center for Excellence in Vascular Biology Brigham and Women's Hospital Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
16
|
Hisey CL, Artuyants A, Guo G, Chang V, Reshef G, Middleditch M, Jacob B, Chamley LW, Blenkiron C. Investigating the consistency of extracellular vesicle production from breast cancer subtypes using CELLine adherent bioreactors. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e60. [PMID: 38938775 PMCID: PMC11080891 DOI: 10.1002/jex2.60] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicle (EV) research has grown rapidly in recent years, largely due to the potential use of EVs as liquid biopsy biomarkers or therapeutics. However, in-depth characterisation and validation of EVs produced using conventional in vitro cultures can be challenging due to the large area of cell monolayers and volumes of culture media required. To overcome this obstacle, multiple bioreactor designs have been tested for EV production with varying success, but the consistency of EVs produced over time in these systems has not been reported previously. In this study, we demonstrate that several breast cancer cell lines of different subtypes can be cultured simultaneously in space, resource, and time efficient manner using CELLine AD 1000 systems, allowing the consistent production of vast amounts of EVs for downstream experimentation. We report an improved workflow used for inoculating, maintaining, and monitoring the bioreactors, their EV production, and the characterisation of the EVs produced. Lastly, our proteomic analyses of the EVs produced throughout the lifetime of the bioreactors show that core EV-associated proteins are relatively consistent, with few minor variations over time, but that tracking the production of EVs is a convenient method to indirectly monitor the bioreactor and consistency of the yielded EVs. These findings will aid future studies requiring the simultaneous production of large amounts of EVs from several cell lines of different subtypes of a disease and other EV biomanufacturing applications.
Collapse
Affiliation(s)
- Colin L. Hisey
- Hub for Extracellular Vesicle InvestigationsUniversity of AucklandAucklandNew Zealand
- Department of Obstetrics and GynaecologyUniversity of AucklandAucklandNew Zealand
| | - Anastasiia Artuyants
- Hub for Extracellular Vesicle InvestigationsUniversity of AucklandAucklandNew Zealand
- Auckland Cancer Society Research CentreUniversity of AucklandAucklandNew Zealand
| | - George Guo
- Department of PhysiologySchool of Medical SciencesUniversity of AucklandAucklandNew Zealand
| | - Vanessa Chang
- Hub for Extracellular Vesicle InvestigationsUniversity of AucklandAucklandNew Zealand
- Department of Obstetrics and GynaecologyUniversity of AucklandAucklandNew Zealand
| | - Gabrielle Reshef
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | | | - Bincy Jacob
- School of Biological SciencesUniversity of AucklandAucklandNew Zealand
| | - Lawrence W. Chamley
- Hub for Extracellular Vesicle InvestigationsUniversity of AucklandAucklandNew Zealand
- Department of Obstetrics and GynaecologyUniversity of AucklandAucklandNew Zealand
| | - Cherie Blenkiron
- Hub for Extracellular Vesicle InvestigationsUniversity of AucklandAucklandNew Zealand
- Auckland Cancer Society Research CentreUniversity of AucklandAucklandNew Zealand
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
17
|
Pavlic A, Bahram Sangani N, Kerins J, Nicolaes G, Schurgers L, Reutelingsperger C. Vascular Smooth Muscle Cell Neutral Sphingomyelinase 2 in the Release of Exosomes and Vascular Calcification. Int J Mol Sci 2022; 23:ijms23169178. [PMID: 36012444 PMCID: PMC9409231 DOI: 10.3390/ijms23169178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/12/2022] Open
Abstract
Vascular calcification (VC) is the pathological precipitation of calcium salts in the walls of blood vessels. It is a risk factor for cardiovascular events and their associated mortality. VC can be observed in a variety of cardiovascular diseases and is most prominent in diseases that are associated with dysregulated mineral homeostasis such as in chronic kidney disease. Local factors and mechanisms underlying VC are still incompletely understood, but it is appreciated that VC is a multifactorial process in which vascular smooth muscle cells (VSMCs) play an important role. VSMCs participate in VC by releasing extracellular vesicles (EVs), the extent, composition, and propensity to calcify of which depend on VSMC phenotype and microenvironment. Currently, no targeted therapy is available to treat VC. In-depth knowledge of molecular players of EV release and the understanding of their mechanisms constitute a vital foundation for the design of pharmacological treatments to combat VC effectively. This review highlights our current knowledge of VSMCs in VC and focuses on the biogenesis of exosomes and the role of the neutral Sphingomyelinase 2 (nSMase2).
Collapse
Affiliation(s)
- Angelina Pavlic
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Nasim Bahram Sangani
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Johanna Kerins
- University College Maastricht, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Gerry Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Chris Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-388-1533
| |
Collapse
|
18
|
Atkins SK, Sonawane AR, Brouwhuis R, Barrientos J, Ha A, Rogers M, Tanaka T, Okui T, Kuraoka S, Singh SA, Aikawa M, Aikawa E. Induced pluripotent stem cell-derived smooth muscle cells to study cardiovascular calcification. Front Cardiovasc Med 2022; 9:925777. [PMID: 35958427 PMCID: PMC9357895 DOI: 10.3389/fcvm.2022.925777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular calcification is the lead predictor of cardiovascular events and the top cause of morbidity and mortality worldwide. To date, only invasive surgical options are available to treat cardiovascular calcification despite the growing understanding of underlying pathological mechanisms. Key players in vascular calcification are vascular smooth muscle cells (SMCs), which transform into calcifying SMCs and secrete mineralizing extracellular vesicles that form microcalcifications, subsequently increasing plaque instability and consequential plaque rupture. There is an increasing, practical need for a large scale and inexhaustible source of functional SMCs. Here we describe an induced pluripotent stem cell (iPSC)-derived model of SMCs by differentiating iPSCs toward SMCs to study the pathogenesis of vascular calcification. Specifically, we characterize the proteome during iPSC differentiation to better understand the cellular dynamics during this process. First, we differentiated human iPSCs toward an induced-SMC (iSMC) phenotype in a 10-day protocol. The success of iSMC differentiation was demonstrated through morphological analysis, immunofluorescent staining, flow cytometry, and proteomics characterization. Proteomics was performed throughout the entire differentiation time course to provide a robust, well-defined starting and ending cell population. Proteomics data verified iPSC differentiation to iSMCs, and functional enrichment of proteins on different days showed the key pathways changing during iSMC development. Proteomics comparison with primary human SMCs showed a high correlation with iSMCs. After iSMC differentiation, we initiated calcification in the iSMCs by culturing the cells in osteogenic media for 17 days. Calcification was verified using Alizarin Red S staining and proteomics data analysis. This study presents an inexhaustible source of functional vascular SMCs and calcifying vascular SMCs to create an in vitro model of vascular calcification in osteogenic conditions, with high potential for future applications in cardiovascular calcification research.
Collapse
Affiliation(s)
- Samantha K. Atkins
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Abhijeet R. Sonawane
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Abhijeet R. Sonawane,
| | - Romi Brouwhuis
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Johana Barrientos
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Anna Ha
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Maximillian Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Takeshi Tanaka
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Takehito Okui
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Shiori Kuraoka
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Elena Aikawa,
| |
Collapse
|
19
|
Extracellular Vesicles, Inflammation, and Cardiovascular Disease. Cells 2022; 11:cells11142229. [PMID: 35883672 PMCID: PMC9320258 DOI: 10.3390/cells11142229] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. The underlying mechanisms of most cardiovascular disorders involve innate and adaptive immune responses, and extracellular vesicles are implicated in both. In this review, we describe the mechanistic role of extracellular vesicles at the intersection of inflammatory processes and cardiovascular disease. Our discussion focuses on atherosclerosis, myocardial ischemia and ischemic heart disease, heart failure, aortic aneurysms, and valvular pathology.
Collapse
|
20
|
He R, Chou C, Chen L, Stoller M, Kang M, Ho SP. Insights Into Pulp Biomineralization in Human Teeth. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.883336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
IntroductionMineralized pulp (MP) compromises tooth function and its causation is unknown. The hypothesis of this study is that pulp mineralization is associated with pulpal tissue adaptation, increased mineral densities, and decreased permeabilities of tubular dentin and cementum. Methods will include correlative spatial mapping of physicochemical and biochemical characteristics of pulp, and contextualize these properties within the dentin-pulp complex (DPC) to reveal the inherent vunerabilities of pulp.MethodsSpecimens (N = 25) were scanned using micro X-ray computed tomography (micro-XCT) to visualize MP and measure mineral density (MD). Elemental spatial maps of MP were acquired using synchrotron X-ray fluorescence microprobe (μXRF) and energy dispersive X-ray spectroscopy (EDX). Extracted pulp tissues were sectioned for immunolabelling and the sections were imaged using a light microscope. Microscale morphologies and nanoscale ultrastructures of MP were imaged using scanning electron (SEM) and scanning transmission electron microscopy (STEM) techniques.ResultsHeterogeneous distribution of MD from 200 to 2,200 mg/cc, and an average MD of 892 (±407) mg/cc were observed. Highly mineralized pulp with increased number of occluded tubules, reduced pore diameter in cementum, and decreased connectivity in lateral channels were observed. H&E, trichrome, and von Kossa staining showed lower cell and collagen densities, and mineralized regions in pulp. The biomolecules osteopontin (OPN), osteocalcin (OCN), osterix (OSX), and bone sialoprotein (BSP) were immunolocalized around PGP 9.5 positive neurovascular bundles in MP. SEM and STEM revealed a wide range of nano/micro particulates in dentin tubules and spherulitic mineral aggregates in the collagen with intrafibrillar mineral surrounding neurovascular bundles. EDX and μXRF showed elevated counts of Ca, P, Mg, and Zn inside pulp and at the dentin-pulp interface (DPI) in the DPC.ConclusionColocalization of physical and chemical, and biomolecular compositions in MP suggest primary and secondary biomineralization pathways in pulp and dentin at a tissue level, and altered fluid dynamics at an organ level. Elevated counts of Zn at the mineralizing front in MP indicated its role in pulp biomineralization. These observations underpin the inherent mechano- and chemo-responsiveness of the neurovascular DPC and help elucidate the clinical subtleties related to pulpitis, dentin-bridge, and pulp stone formation.
Collapse
|
21
|
Man K, Joukhdar H, Manz XD, Brunet MY, Jiang LH, Rnjak-Kovacina J, Yang XB. Bone tissue engineering using 3D silk scaffolds and human dental pulp stromal cells epigenetic reprogrammed with the selective histone deacetylase inhibitor MI192. Cell Tissue Res 2022; 388:565-581. [PMID: 35362831 PMCID: PMC9110470 DOI: 10.1007/s00441-022-03613-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/11/2022] [Indexed: 11/30/2022]
Abstract
Epigenetics plays a critical role in regulating mesenchymal stem cells’ (MSCs) fate for tissue repair and regeneration. There is increasing evidence that the inhibition of histone deacetylase (HDAC) isoform 3 can enhance MSC osteogenesis. This study investigated the potential of using a selective HDAC2 and 3 inhibitor, MI192, to promote human dental pulp stromal cells (hDPSCs) bone-like tissue formation in vitro and in vivo within porous Bombyx Mori silk scaffolds. Both 2 and 5 wt% silk scaffolds were fabricated and characterised. The 5 wt% scaffolds possess thicker internal lamellae, reduced scaffold swelling and degradation rates, whilst increased compressive modulus in comparison to the 2 wt% silk scaffold. MI192 pre-treatment of hDPSCs on 5 wt% silk scaffold significantly enhanced hDPSCs alkaline phosphatase activity (ALP). The expression of osteoblast-related genes (RUNX2, ALP, Col1a, OCN) was significantly upregulated in the MI192 pre-treated cells. Histological analysis confirmed that the MI192 pre-treated hDPSCs-silk scaffold constructs promoted bone extracellular matrix (ALP, Col1a, OCN) deposition and mineralisation compared to the untreated group. Following 6 weeks of subcutaneous implantation in nude mice, the MI192 pre-treated hDPSCs-silk scaffold constructs enhanced the vascularisation and extracellular matrix mineralisation compared to untreated control. In conclusion, these findings demonstrate the potential of using epigenetic reprogramming and silk scaffolds to promote hDPSCs bone formation efficacy, which provides evidence for clinical translation of this technology for bone augmentation.
Collapse
Affiliation(s)
- Kenny Man
- Biomaterials & Tissue Engineering Group, School of Dentistry, University of Leeds, WTBB, St. James's University Hospital, Leeds, LS97TF, UK.,School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Habib Joukhdar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Xue D Manz
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia.,Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Centre, Amsterdam, The Netherlands
| | - Mathieu Y Brunet
- School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Xuebin B Yang
- Biomaterials & Tissue Engineering Group, School of Dentistry, University of Leeds, WTBB, St. James's University Hospital, Leeds, LS97TF, UK.
| |
Collapse
|
22
|
Qiang L, Hu J, Tian M, Li Y, Ren C, Deng Y, Jiang Y. Extracellular vesicles from helicobacter pylori-infected cells and helicobacter pylori outer membrane vesicles in atherosclerosis. Helicobacter 2022; 27:e12877. [PMID: 35099837 DOI: 10.1111/hel.12877] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/26/2021] [Accepted: 01/11/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND The role of H. pylori infection has been reported in various extragastric diseases, particularly, the correlation between H. pylori and atherosclerosis (AS) have received lots of attention. Some scholars demonstrated that the presence of H. pylori-specific DNA in the sclerotic plaques of atheromatous patients provides biological evidences, with indicating that H. pylori infection is a potential factor of AS. However, the underlying mechanism of H. pylori or their products cross the epithelial barriers to enter the blood circulation remains unclear. Recent studies have shown that the extracellular vesicles (EVs) derived from H. pylori-infected gastric epithelial cells encapsulated H. pylori virulence factor cytotoxin-associated gene A (CagA) and existed in the blood samples of patients or mice, which indicating that they can carry CagA into the blood circulation. Based on these findings, some researchers proposed a hypothesis that H. pylori is involved in the pathogenesis of AS via EVs-based mechanisms. In addition, outer membrane vesicles (OMVs) serve as transport vehicles to deliver H. pylori virulence factors to epithelial cells. It is necessary to discuss the role of H. pylori OMVs in the development of AS. OBJECTIVES This review will focus on the correlation between H. pylori infection and AS and tried to unveil the possible role of EVs from H. pylori-infected cells and H. pylori OMVs in the pathogenesis of AS, with a view to providing help in refining our knowledge in this aspect. METHODS All of information included in this review was retrieved from published studies on H. pylori infection in AS. RESULTS H. pylori infection may be an atherosclerotic risk factor and drives researchers to reevaluate the role of H. pylori in the pathogenesis of AS. Some findings proposed a new hypothesis that H. pylori may be involved in the pathogenesis of AS through EVs-based mechanisms. Besides EVs from H. pylori-infected cells, whether H. pylori OMVs may play some role in the pathogenesis of AS is still remain unclear. CONCLUSION Existing epidemiological and clinical evidence had shown that there is a possible association between H. pylori and AS. However, except for the larger randomized controlled trials, more basic research about EVs from H. pylori-infected cells and H. pylori OMVs is the need of the hour to unveil the possible role of H. pylori infection in the pathogenesis of AS.
Collapse
Affiliation(s)
- Liming Qiang
- Department of Gastroenterology, West China-Guang'an Hospital, Sichuan University, Guang'an, China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Mingyuan Tian
- Department of Endocrinology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Li
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Chao Ren
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yi Deng
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yuan Jiang
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
23
|
Coppola A, Vigorito C, Lombari P, Martínez YG, Borriello M, Trepiccione F, Ingrosso D, Perna AF. Uremic Toxin Lanthionine Induces Endothelial Cell Mineralization In Vitro. Biomedicines 2022; 10:biomedicines10020444. [PMID: 35203651 PMCID: PMC8962276 DOI: 10.3390/biomedicines10020444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Vascular calcification (VC) is a pathological event caused by the unusual deposition of minerals in the vascular system, representing the leading cause of cardiovascular mortality in chronic kidney disease (CKD). In CKD, the deregulation of calcium and phosphate metabolism, along with the effect of several uremic toxins, act as key processes conveying altered mineralization. In this work, we tested the ability of lanthionine, a novel uremic toxin, to promote calcification in human endothelial cell cultures (Ea.hy926). We evaluated the effects of lanthionine, at a concentration similar to that actually detected in CKD patients, alone and under pro-calcifying culture conditions using calcium and phosphate. In pro-calcific culture conditions, lanthionine increased both the intracellular and extracellular calcium content and induced the expression of Bone Morphogenetic Protein 2 (BMP2) and RUNX Family Transcription Factor 2 (RUNX2). Lanthionine treatment, in pro-calcifying conditions, raised levels of tissue-nonspecific alkaline phosphatase (ALPL), whose expression also overlapped with Dickkopf WNT Signaling Pathway Inhibitor 1 (DKK1) gene expression, suggesting a possible role of the latter gene in the activation of ALPL. In addition, treatment with lanthionine alone or in combination with calcium and phosphate reduced Inorganic Pyrophosphate Transport Regulator (ANKH) gene expression, a protective factor toward the mineralizing process. Moreover, lanthionine in a pro-calcifying condition induced the activation of ERK1/2, which is not associated with an increase in DKK1 protein levels. Our data underscored a link between mineral disease and the alterations of sulfur amino acid metabolisms at a cell and molecular level. These results set the basis for the understanding of the link between uremic toxins and mineral-bone disorder during CKD progression.
Collapse
Affiliation(s)
- Annapaola Coppola
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
| | - Carmela Vigorito
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Patrizia Lombari
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Yuselys García Martínez
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Margherita Borriello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
| | - Francesco Trepiccione
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Diego Ingrosso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
- Correspondence: (D.I.); (A.F.P.)
| | - Alessandra F. Perna
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
- Correspondence: (D.I.); (A.F.P.)
| |
Collapse
|
24
|
Li T, Yu H, Zhang D, Feng T, Miao M, Li J, Liu X. Matrix Vesicles as a Therapeutic Target for Vascular Calcification. Front Cell Dev Biol 2022; 10:825622. [PMID: 35127686 PMCID: PMC8814528 DOI: 10.3389/fcell.2022.825622] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/04/2022] [Indexed: 01/01/2023] Open
Abstract
Vascular calcification (VC) is linked to an increased risk of heart disease, stroke, and atherosclerotic plaque rupture. It is a cell-active process regulated by vascular cells rather than pure passive calcium (Ca) deposition. In recent years, extracellular vesicles (EVs) have attracted extensive attention because of their essential role in the process of VC. Matrix vesicles (MVs), one type of EVs, are especially critical in extracellular matrix mineralization and the early stages of the development of VC. Vascular smooth muscle cells (VSMCs) have the potential to undergo phenotypic transformation and to serve as a nucleation site for hydroxyapatite crystals upon extracellular stimulation. However, it is not clear what underlying mechanism that MVs drive the VSMCs phenotype switching and to result in calcification. This article aims to review the detailed role of MVs in the progression of VC and compare the difference with other major drivers of calcification, including aging, uremia, mechanical stress, oxidative stress, and inflammation. We will also bring attention to the novel findings in the isolation and characterization of MVs, and the therapeutic application of MVs in VC.
Collapse
Affiliation(s)
- Tiantian Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hongchi Yu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Tang Feng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Michael Miao
- Division of Oral & Craniofacial Health Sciences, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, United States
| | - Jianwei Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jianwei Li, ; Xiaoheng Liu,
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Jianwei Li, ; Xiaoheng Liu,
| |
Collapse
|
25
|
Vascular Pathobiology: Atherosclerosis and Large Vessel Disease. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
26
|
Ramos AP, Sebinelli HG, Ciancaglini P, Rosato N, Mebarek S, Buchet R, Millán JL, Bottini M. The functional role of soluble proteins acquired by extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e34. [PMID: 38938684 PMCID: PMC11080634 DOI: 10.1002/jex2.34] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed nanosized particles released by all cell types during physiological as well as pathophysiological processes to carry out diverse biological functions, including acting as sources of cellular dumping, signalosomes and mineralisation nanoreactors. The ability of EVs to perform specific biological functions is due to their biochemical machinery. Among the components of the EVs' biochemical machinery, surface proteins are of critical functional significance as they mediate the interactions of EVs with components of the extracellular milieu, the extracellular matrix and neighbouring cells. Surface proteins are thought to be native, that is, pre-assembled on the EVs' surface by the parent cells before the vesicles are released. However, numerous pieces of evidence have suggested that soluble proteins are acquired by the EVs' surface from the extracellular milieu and further modulate the biological functions of EVs during innate and adaptive immune responses, autoimmune disorders, complement activation, coagulation, viral infection and biomineralisation. Herein, we will describe the methods currently used to identify the EVs' surface proteins and discuss recent knowledge on the functional relevance of the soluble proteins acquired by EVs.
Collapse
Affiliation(s)
- Ana Paula Ramos
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Heitor Gobbi Sebinelli
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Pietro Ciancaglini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Nicola Rosato
- Dipartimento di Medicina SperimentaleUniversita’ di Roma “Tor Vergata”RomeItaly
| | - Saida Mebarek
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | - Rene Buchet
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | | | - Massimo Bottini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
- Sanford Burnham PrebysLa JollaCaliforniaUSA
| |
Collapse
|
27
|
Sun XJ, Liu NF. Diabetic mellitus, vascular calcification and hypoxia: A complex and neglected tripartite relationship. Cell Signal 2021; 91:110219. [PMID: 34921978 DOI: 10.1016/j.cellsig.2021.110219] [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: 09/25/2021] [Revised: 12/11/2021] [Accepted: 12/11/2021] [Indexed: 11/15/2022]
Abstract
DM (diabetic mellitus) and its common vascular complications VC (vascular calcification), are increasingly harmful to human health. In recent years, the research on the relationship between DM and VC is also deepening. Hypoxia, as one of the pathogenic factors of many disease models, is also closely related to the occurrence of DM and VC. There are some studies on the role of hypoxia in the pathogenesis of DM and VC respectively, but no one has made an in-depth summary of the systematic connection between hypoxia, DM and VC. Therefore, what we want to review in this article are the relationship between DM, VC and hypoxia, respectively, as well as the role of hypoxia in the development of DM and VC, which has little concern but is a novel and potentially target that may provide some new ideas for the prevention and treatment of DM, VC, especially diabetic VC.
Collapse
Affiliation(s)
- Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China.
| |
Collapse
|
28
|
Bartoli-Leonard F, Zimmer J, Aikawa E. Innate and adaptive immunity: the understudied driving force of heart valve disease. Cardiovasc Res 2021; 117:2506-2524. [PMID: 34432007 PMCID: PMC8783388 DOI: 10.1093/cvr/cvab273] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Calcific aortic valve disease (CAVD), and its clinical manifestation that is calcific aortic valve stenosis, is the leading cause for valve disease within the developed world, with no current pharmacological treatment available to delay or halt its progression. Characterized by progressive fibrotic remodelling and subsequent pathogenic mineralization of the valve leaflets, valve disease affects 2.5% of the western population, thus highlighting the need for urgent intervention. Whilst the pathobiology of valve disease is complex, involving genetic factors, lipid infiltration, and oxidative damage, the immune system is now being accepted to play a crucial role in pathogenesis and disease continuation. No longer considered a passive degenerative disease, CAVD is understood to be an active inflammatory process, involving a multitude of pro-inflammatory mechanisms, with both the adaptive and the innate immune system underpinning these complex mechanisms. Within the valve, 15% of cells evolve from haemopoietic origin, and this number greatly expands following inflammation, as macrophages, T lymphocytes, B lymphocytes, and innate immune cells infiltrate the valve, promoting further inflammation. Whether chronic immune infiltration or pathogenic clonal expansion of immune cells within the valve or a combination of the two is responsible for disease progression, it is clear that greater understanding of the immune systems role in valve disease is required to inform future treatment strategies for control of CAVD development.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jonas Zimmer
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
29
|
High levels of osteoprotegerin are associated with coronary artery calcification in patients suspected of a chronic coronary syndrome. Sci Rep 2021; 11:18946. [PMID: 34556709 PMCID: PMC8460823 DOI: 10.1038/s41598-021-98177-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/03/2021] [Indexed: 11/08/2022] Open
Abstract
Plasma osteoprotegerin (OPG) and vascular smooth muscle cell (VSMC) derived extracellular vesicles (EVs) are important regulators in the process of vascular calcification (VC). In population studies, high levels of OPG are associated with events. In animal studies, however, high OPG levels result in reduction of VC. VSMC-derived EVs are assumed to be responsible for OPG transport and VC but this role has not been studied. For this, we investigated the association between OPG in plasma and circulating EVs with coronary artery calcium (CAC) as surrogate for VC in symptomatic patients. We retrospectively assessed 742 patients undergoing myocardial perfusion imaging (MPI). CAC scores were determined on the MPI-CT images using a previously developed automated algorithm. Levels of OPG were quantified in plasma and two EV-subpopulations (LDL and TEX), using an electrochemiluminescence immunoassay. Circulating levels of OPG were independently associated with CAC scores in plasma; OR 1.39 (95% CI 1.17–1.65), and both EV populations; EV-LDL; OR 1.51 (95% CI 1.27–1.80) and EV-TEX; OR 1.21 (95% CI 1.02–1.42). High levels of OPG in plasma were independently associated with CAC scores in this symptomatic patient cohort. High levels of EV-derived OPG showed the same positive association with CAC scores, suggesting that EV-derived OPG mirrors the same pathophysiological process as plasma OPG.
Collapse
|
30
|
Lanzer P, Hannan FM, Lanzer JD, Janzen J, Raggi P, Furniss D, Schuchardt M, Thakker R, Fok PW, Saez-Rodriguez J, Millan A, Sato Y, Ferraresi R, Virmani R, St Hilaire C. Medial Arterial Calcification: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 78:1145-1165. [PMID: 34503684 PMCID: PMC8439554 DOI: 10.1016/j.jacc.2021.06.049] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 01/07/2023]
Abstract
Medial arterial calcification (MAC) is a chronic systemic vascular disorder distinct from atherosclerosis that is frequently but not always associated with diabetes mellitus, chronic kidney disease, and aging. MAC is also a part of more complex phenotypes in numerous less common diseases. The hallmarks of MAC include disseminated and progressive precipitation of calcium phosphate within the medial layer, a prolonged and clinically silent course, and compromise of hemodynamics associated with chronic limb-threatening ischemia. MAC increases the risk of complications during vascular interventions and mitigates their outcomes. With the exception of rare monogenetic defects affecting adenosine triphosphate metabolism, MAC pathogenesis remains unknown, and causal therapy is not available. Implementation of genetics and omics-based approaches in research recognizing the critical importance of calcium phosphate thermodynamics holds promise to unravel MAC molecular pathogenesis and to provide guidance for therapy. The current state of knowledge concerning MAC is reviewed, and future perspectives are outlined.
Collapse
Affiliation(s)
- Peter Lanzer
- Middle German Heart Center-Bitterfeld, Bitterfeld-Wolfen Health Care Center, Bitterfeld, Germany.
| | - Fadil M Hannan
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Jan D Lanzer
- Institute for Computational Biomedicine, Bioquant, Faculty of Medicine, Heidelberg University, Heidelberg, Germany; Department of Internal Medicine II, Heidelberg University Hospital, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Heidelberg, Germany
| | | | - Paolo Raggi
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Dominic Furniss
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Mirjam Schuchardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Rajesh Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pak-Wing Fok
- Department of Mathematical Sciences, University of Delaware, Newark, Delaware, USA
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Bioquant, Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Angel Millan
- Institute of Materials Science, University of Zaragoza, Zaragoza, Spain
| | - Yu Sato
- CVPath Institute, Gaithersburg, Maryland, USA
| | | | | | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Calcific aortic stenosis (CAVS) is the most common form of valvular heart disease in developed countries, increasing in prevalence with the aging population. Surgical or transcatheter aortic valve replacement is the only treatment available for CAVS. However, these interventions are typically reserved for severe symptomatic aortic stenosis (AS). The purpose of this review is to summarize the recent literature in uncovering the underlying pathophysiology of CAVS in the setting of lipoprotein (a) [Lp(a)] and emerging therapies targeting Lp(a) which may help halt disease progression in CAVS. RECENT FINDINGS Pathophysiologic, epidemiological, and genetic studies over the past two decades have provided strong evidence that Lp(a) is an important mediator of calcific aortic valvular disease (CAVD). Studies suggest that Lp(a) is a key carrier of pro-calcifying oxidized phospholipids (OxPL). The metabolism of OxPL results in a pro-inflammatory state and subsequent valvular thickening and mineralization through pro-osteogenic signaling. The identification of Lp(a) as a causal mediator of CAVD has allowed for opportunities for emerging therapeutic agents which may slow the progression of CAVD (Fig. 1JOURNAL/cocar/04.03/00001573-202109000-00007/figure1/v/2021-08-04T080204Z/r/image-jpeg). SUMMARY This review summarizes the current knowledge on the association of Lp(a) with CAVD and ongoing studies of potential Lp(a)-lowering therapies. Based on the rate-limiting and causal role of Lp(a) in progression of CAVS, these therapies may represent novel pharmacotherapies in AS and inform the developing role of Lp(a) in the clinical management of CAVD.
Collapse
|
32
|
Marsh SR, Pridham KJ, Jourdan J, Gourdie RG. Novel Protocols for Scalable Production of High Quality Purified Small Extracellular Vesicles from Bovine Milk. Nanotheranostics 2021; 5:488-498. [PMID: 34367882 PMCID: PMC8342262 DOI: 10.7150/ntno.62213] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022] Open
Abstract
Extracellular Vesicles (EVs) are cell-secreted nanovesicles that have unique potential for encapsulating and targeting "difficult-to-drug" therapeutic cargos. Milk provides an enriched source of EVs, and of particular interest to the drug delivery field, small EVs. Small EVs are distinguished from large EVs by membrane components, biogenesis mechanism and downstream functionality - in particular, small EVs are primarily composed of exosomes, which show high stability in vivo and naturally function in the targeted delivery of biological materials to cells. Moreover, bovine milk is abundantly produced by the dairy industry, widely consumed, and generally well tolerated by humans. Importantly, there is evidence that milk exosomes and small EVs are efficiently taken up into the circulation from the gut, providing the opportunity for their use in administration of therapeutics such as microRNAs or peptides not typically available via an oral route. Unfortunately, present methods for isolation do not efficiently separate EVs from milk proteins, resulting in contamination that is not desirable in a clinical-grade therapeutic. Herein, we present novel EV purification methods focused on optimized timing and levels of temperature and divalent cation chelation. Incorporation of these solubilization steps into centrifugation- and tangential flow filtration-based methods provide large amounts of purified small EVs at ultra-dense concentrations, which are substantially free from contaminating milk proteins. Remarkably, these ultra-dense isolates equal 10 to 15% of the starting volume of milk indicating a prodigious rate of small EV production by mammary glands. Our approach enables gentle, scalable production of ultrastructurally and functionally intact small EVs from milk, providing a path to their industrial scale purification for oral delivery of therapeutic biologics and small drugs.
Collapse
Affiliation(s)
- Spencer R. Marsh
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Kevin J. Pridham
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Jane Jourdan
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Robert G. Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carillion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, 24061, USA
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA, 24016, USA
- Faculty of Health Science, Virginia Tech, Blacksburg, VA, 24061, USA
| |
Collapse
|
33
|
Al Naem M, Bourebaba L, Kucharczyk K, Röcken M, Marycz K. Therapeutic mesenchymal stromal stem cells: Isolation, characterization and role in equine regenerative medicine and metabolic disorders. Stem Cell Rev Rep 2021; 16:301-322. [PMID: 31797146 DOI: 10.1007/s12015-019-09932-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSC) have become a popular treatment modality in equine orthopaedics. Regenerative therapies are especially interesting for pathologies like complicated tendinopathies of the distal limb, osteoarthritis, osteochondritis dissecans (OCD) and more recently metabolic disorders. Main sources for MSC harvesting in the horse are bone marrow, adipose tissue and umbilical cord blood. While the acquisition of umbilical cord blood is fairly easy and non-invasive, extraction of bone marrow and adipose tissue requires more invasive techniques. Characterization of the stem cells as a result of any isolation method, is also a crucial step for the confirmation of the cells' stemness properties; thus, three main characteristics must be fulfilled by these cells, namely: adherence, expression of a series of well-defined differentiation clusters as well as pluripotency. EVs, resulting from the paracrine action of MSCs, also play a key role in the therapeutic mechanisms mediated by stem cells; MSC-EVs are thus largely implicated in the regulation of proliferation, maturation, polarization and migration of various target cells. Evidence that EVs alone represent a complex network 0involving different soluble factors and could then reflect biophysical characteristics of parent cells has fuelled the importance of developing highly specific techniques for their isolation and analysis. All these aspects related to the functional and technical understanding of MSCs will be discussed and summarized in this review.
Collapse
Affiliation(s)
- Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland.,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kucharczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany. .,Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,International Institute of Translational Medicine, Jesionowa, 11, Malin, 55-114, Wisznia Mała, Poland.
| |
Collapse
|
34
|
Zhang L, Yao J, Yao Y, Boström KI. Contributions of the Endothelium to Vascular Calcification. Front Cell Dev Biol 2021; 9:620882. [PMID: 34079793 PMCID: PMC8165270 DOI: 10.3389/fcell.2021.620882] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 04/06/2021] [Indexed: 01/14/2023] Open
Abstract
Vascular calcification (VC) increases morbidity and mortality and constitutes a significant obstacle during percutaneous interventions and surgeries. On a cellular and molecular level, VC is a highly regulated process that involves abnormal cell transitions and osteogenic differentiation, re-purposing of signaling pathways normally used in bone, and even formation of osteoclast-like cells. Endothelial cells have been shown to contribute to VC through a variety of means. This includes direct contributions of osteoprogenitor cells generated through endothelial-mesenchymal transitions in activated endothelium, with subsequent migration into the vessel wall. The endothelium also secretes pro-osteogenic growth factors, such as bone morphogenetic proteins, inflammatory mediators and cytokines in conditions like hyperlipidemia, diabetes, and renal failure. High phosphate levels caused by renal disease have deleterious effects on the endothelium, and induction of tissue non-specific alkaline phosphatase adds to the calcific process. Furthermore, endothelial activation promotes proteolytic destruction of the internal elastic lamina that serves, among other things, as a stabilizer of the endothelium. Appropriate bone mineralization is highly dependent on active angiogenesis, but it is unclear whether the same relationship exists in VC. Through its location facing the vascular lumen, the endothelium is the first to encounter circulating factor and bone marrow-derived cells that might contribute to osteoclast-like versus osteoblast-like cells in the vascular wall. In the same way, the endothelium may be the easiest target to reach with treatments aimed at limiting calcification. This review provides a brief summary of the contributions of the endothelium to VC as we currently know them.
Collapse
Affiliation(s)
- Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- UCLA Molecular Biology Institute, Los Angeles, CA, United States
| | - Kristina I. Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
35
|
Freise C, Querfeld U, Ludwig A, Hamm B, Schnorr J, Taupitz M. Uraemic extracellular vesicles augment osteogenic transdifferentiation of vascular smooth muscle cells via enhanced AKT signalling and PiT-1 expression. J Cell Mol Med 2021; 25:5602-5614. [PMID: 33960650 PMCID: PMC8184672 DOI: 10.1111/jcmm.16572] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EV) function as messengers between endothelial cells (EC) and vascular smooth muscle cells (VSMC). Since chronic kidney disease (CKD) increases the risk for vascular calcifications, we investigated whether EV derived from uraemic milieu‐stimulated EC and derived from uraemic rats impact the osteogenic transdifferentiation/calcification of VSMC. For that purpose, human EC were treated with urea and indoxyl sulphate or left untreated. Experimental uraemia in rats was induced by adenine feeding. ‘Uraemic’ and control EV (EVUR; EVCTRL) were isolated from supernatants and plasma by using an exosome isolation reagent. Rat VSMC were treated with a pro‐calcifying medium (CM) with or without EV supplementation. Gene expressions, miRNA contents and protein expressions were determined by qPCR and Western blots, respectively. Calcifications were determined by colorimetric assays. Delivery of miRNA inhibitors/mimics to EV and siRNA to VSMC was achieved via transfection. EVCTRL and EVUR differed in size and miRNA contents. Contrary to EVCTRL, EC‐ and plasma‐derived EVUR significantly increased the pro‐calcifying effects of CM, including altered gene expressions of osterix, runx2, osteocalcin and SM22α. Further, EVUR enhanced the protein expression of the phosphate transporter PiT‐1 in VSMC and induced a phosphorylation of AKT and ERK. Knock down of PiT‐1 and individual inhibition of AKT and ERK signalling in VSMC blocked the pro‐calcifying effects of EVUR. Similar effects were achieved by inhibition of miR‐221/‐222 and mimicking of miR‐143/‐145 in EVUR. In conclusion, EVUR might represent an additional puzzle piece of the complex pathophysiology of vascular calcifications in CKD.
Collapse
Affiliation(s)
- Christian Freise
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Uwe Querfeld
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Antje Ludwig
- Department of Cardiology and Angiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bernd Hamm
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jörg Schnorr
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Matthias Taupitz
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
36
|
Abstract
Calcific aortic valve disease sits at the confluence of multiple world-wide epidemics of aging, obesity, diabetes, and renal dysfunction, and its prevalence is expected to nearly triple over the next 3 decades. This is of particularly dire clinical relevance, as calcific aortic valve disease can progress rapidly to aortic stenosis, heart failure, and eventually premature death. Unlike in atherosclerosis, and despite the heavy clinical toll, to date, no pharmacotherapy has proven effective to halt calcific aortic valve disease progression, with invasive and costly aortic valve replacement representing the only treatment option currently available. This substantial gap in care is largely because of our still-limited understanding of both normal aortic valve biology and the key regulatory mechanisms that drive disease initiation and progression. Drug discovery is further hampered by the inherent intricacy of the valvular microenvironment: a unique anatomic structure, a complex mixture of dynamic biomechanical forces, and diverse and multipotent cell populations collectively contributing to this currently intractable problem. One promising and rapidly evolving tactic is the application of multiomics approaches to fully define disease pathogenesis. Herein, we summarize the application of (epi)genomics, transcriptomics, proteomics, and metabolomics to the study of valvular heart disease. We also discuss recent forays toward the omics-based characterization of valvular (patho)biology at single-cell resolution; these efforts promise to shed new light on cellular heterogeneity in healthy and diseased valvular tissues and represent the potential to efficaciously target and treat key cell subpopulations. Last, we discuss systems biology- and network medicine-based strategies to extract meaning, mechanisms, and prioritized drug targets from multiomics datasets.
Collapse
Affiliation(s)
- Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
- Heart Division, Royal Brompton & Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW This review encompasses the main novelties regarding nonimmune mechanisms implicated in the pathogenesis of idiopathic inflammatory myopathies (IIM). RECENT FINDINGS In recent years, growing data support a role for endoplasmic-reticulum (ER) stress as a propagator of muscular damage, together with the release of interferon type I and reactive oxygen species in hypoxemic muscle fibers. Other studies evaluating the relationship between autophagy and Toll-like receptors (TLRs) in IIM subtypes have shown increased TLR3 and TLR4 expression in fibers of IIM patients and colocalization with LC3, an autophagy marker, submitting autophagy as a likely player in IIM pathogenesis. Most novel evidences concern the potential role of denervation of the neuromuscular junction in IIM, possibly connected to hyperexpression of MHC-I, and trafficking of extracellular vesicles, which may represent a connection between nonimmune and immune-mediated mechanisms of muscle inflammation and damage. SUMMARY Nonimmune mechanisms contribute to the pathogenesis of IIM, likely cooperating with immune-mediated inflammation. Consistent data were released for ER stress, autophagy, mitochondrial dysfunction and hypoxia; in addition to, neuromuscular denervation and extracellular vesicles have been proposed as thoughtful links between muscle inflammation, damage and atrophy. Further understanding of nonimmune abnormalities and potential reversible pathways is needed to improve the management of IIM.
Collapse
|
38
|
Sahoo S, Adamiak M, Mathiyalagan P, Kenneweg F, Kafert-Kasting S, Thum T. Therapeutic and Diagnostic Translation of Extracellular Vesicles in Cardiovascular Diseases: Roadmap to the Clinic. Circulation 2021; 143:1426-1449. [PMID: 33819075 PMCID: PMC8021236 DOI: 10.1161/circulationaha.120.049254] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Exosomes are small membrane-bound vesicles of endocytic origin that are actively secreted. The potential of exosomes as effective communicators of biological signaling in myocardial function has previously been investigated, and a recent explosion in exosome research not only underscores their significance in cardiac physiology and pathology, but also draws attention to methodological limitations of studying these extracellular vesicles. In this review, we discuss recent advances and challenges in exosome research with an emphasis on scientific innovations in isolation, identification, and characterization methodologies, and we provide a comprehensive summary of web-based resources available in the field. Importantly, we focus on the biology and function of exosomes, highlighting their fundamental role in cardiovascular pathophysiology to further support potential applications of exosomes as biomarkers and therapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Marta Adamiak
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Prabhu Mathiyalagan
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York (S.S., M.A., P.M.)
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
| | - Sabine Kafert-Kasting
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany (S.K-K., T.T.)
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) (F.K., S.K-K., T.T.), Hannover Medical School, Germany
- REBIRTH Center for Translational Regenerative Medicine (T.T.), Hannover Medical School, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany (S.K-K., T.T.)
| |
Collapse
|
39
|
Canet-Soulas E, Bessueille L, Mechtouff L, Magne D. The Elusive Origin of Atherosclerotic Plaque Calcification. Front Cell Dev Biol 2021; 9:622736. [PMID: 33768090 PMCID: PMC7985066 DOI: 10.3389/fcell.2021.622736] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
It has been known for decades or even centuries that arteries calcify as they age. Vascular calcification probably affects all adults, since virtually all have atherosclerotic plaques: an accumulation of lipids, inflammatory cells, necrotic debris, and calcium phosphate crystals. A high vascular calcium score is associated with a high cardiovascular mortality risk, and relatively recent data suggest that even microcalcifications that form in early plaques may destabilize plaques and trigger a cardiovascular event. If the cellular and molecular mechanisms of plaque calcification have been relatively well characterized in mice, human plaques appear to calcify through different mechanisms that remain obscure. In this context, we will first review articles reporting the location and features of early calcifications in human plaques and then review the articles that explored the mechanisms though which human and mouse plaques calcify.
Collapse
Affiliation(s)
- Emmanuelle Canet-Soulas
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Laurence Bessueille
- ICBMS, CNRS, INSA Lyon, CPE, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Laura Mechtouff
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Stroke Department, Hospices Civils de Lyon, Lyon, France
| | - David Magne
- ICBMS, CNRS, INSA Lyon, CPE, University of Lyon, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
40
|
Bäck M, Michel JB. From organic and inorganic phosphates to valvular and vascular calcifications. Cardiovasc Res 2021; 117:2016-2029. [PMID: 33576771 PMCID: PMC8318101 DOI: 10.1093/cvr/cvab038] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/26/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Calcification of the arterial wall and valves is an important part of the pathophysiological process of peripheral and coronary atherosclerosis, aortic stenosis, ageing, diabetes, and chronic kidney disease. This review aims to better understand how extracellular phosphates and their ability to be retained as calcium phosphates on the extracellular matrix initiate the mineralization process of arteries and valves. In this context, the physiological process of bone mineralization remains a human model for pathological soft tissue mineralization. Soluble (ionized) calcium precipitation occurs on extracellular phosphates; either with inorganic or on exposed organic phosphates. Organic phosphates are classified as either structural (phospholipids, nucleic acids) or energetic (corresponding to phosphoryl transfer activities). Extracellular phosphates promote a phenotypic shift in vascular smooth muscle and valvular interstitial cells towards an osteoblast gene expression pattern, which provokes the active phase of mineralization. A line of defense systems protects arterial and valvular tissue calcifications. Given the major roles of phosphate in soft tissue calcification, phosphate mimetics, and/or prevention of phosphate dissipation represent novel potential therapeutic approaches for arterial and valvular calcification.
Collapse
Affiliation(s)
- Magnus Bäck
- Division of Valvular and Coronary Disease, Department of Cardiology, Karolinska University Hospital, 141 86 Stockholm, Sweden.,Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,University of Lorraine, Nancy University Hospital, INSERM U1116, Nancy, France
| | | |
Collapse
|
41
|
Furmanik M, van Gorp R, Whitehead M, Ahmad S, Bordoloi J, Kapustin A, Schurgers LJ, Shanahan CM. Endoplasmic Reticulum Stress Mediates Vascular Smooth Muscle Cell Calcification via Increased Release of Grp78 (Glucose-Regulated Protein, 78 kDa)-Loaded Extracellular Vesicles. Arterioscler Thromb Vasc Biol 2021; 41:898-914. [PMID: 33297752 PMCID: PMC7837691 DOI: 10.1161/atvbaha.120.315506] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/25/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Vascular calcification is common among aging populations and mediated by vascular smooth muscle cells (VSMCs). The endoplasmic reticulum (ER) is involved in protein folding and ER stress has been implicated in bone mineralization. The role of ER stress in VSMC-mediated calcification is less clear. Approach and Results: mRNA expression of the ER stress markers PERK (PKR (protein kinase RNA)-like ER kinase), ATF (activating transcription factor) 4, ATF6, and Grp78 (glucose-regulated protein, 78 kDa) was detectable in human vessels with levels of PERK decreased in calcified plaques compared to healthy vessels. Protein deposition of Grp78/Grp94 was increased in the matrix of calcified arteries. Induction of ER stress accelerated human primary VSMC-mediated calcification, elevated expression of some osteogenic markers (Runx2 [RUNX family transcription factor 2], OSX [Osterix], ALP [alkaline phosphatse], BSP [bone sialoprotein], and OPG [osteoprotegerin]), and decreased expression of SMC markers. ER stress potentiated extracellular vesicle (EV) release via SMPD3 (sphingomyelin phosphodiesterase 3). EVs from ER stress-treated VSMCs showed increased Grp78 levels and calcification. Electron microscopy confirmed the presence of Grp78/Grp94 in EVs. siRNA (short interfering RNA) knock-down of Grp78 decreased calcification. Warfarin-induced Grp78 and ATF4 expression in rat aortas and VSMCs and increased calcification in an ER stress-dependent manner via increased EV release. CONCLUSIONS ER stress induces vascular calcification by increasing release of Grp78-loaded EVs. Our results reveal a novel mechanism of action of warfarin, involving increased EV release via the PERK-ATF4 pathway, contributing to calcification. This study is the first to show that warfarin induces ER stress and to link ER stress to cargo loading of EVs.
Collapse
MESH Headings
- Activating Transcription Factor 4/genetics
- Activating Transcription Factor 4/metabolism
- Adolescent
- Adult
- Aged
- Animals
- Cells, Cultured
- Disease Models, Animal
- Endoplasmic Reticulum Chaperone BiP
- Endoplasmic Reticulum Stress/drug effects
- Extracellular Vesicles/drug effects
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/pathology
- Female
- Gene Expression Regulation
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/metabolism
- Humans
- Male
- Middle Aged
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rats, Sprague-Dawley
- Signal Transduction
- Vascular Calcification/chemically induced
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Warfarin/toxicity
- Young Adult
- eIF-2 Kinase/genetics
- eIF-2 Kinase/metabolism
- Rats
Collapse
Affiliation(s)
- Malgorzata Furmanik
- Department of Biochemistry, Cardiovascular Research Institute Maastricht CARIM, Maastricht University, the Netherlands (M.F., R.v.G., L.J.S.)
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (M.F., M.W., S.A., J.B., A.K., C.M.S.)
| | - Rick van Gorp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht CARIM, Maastricht University, the Netherlands (M.F., R.v.G., L.J.S.)
| | - Meredith Whitehead
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (M.F., M.W., S.A., J.B., A.K., C.M.S.)
| | - Sadia Ahmad
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (M.F., M.W., S.A., J.B., A.K., C.M.S.)
| | - Jayanta Bordoloi
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (M.F., M.W., S.A., J.B., A.K., C.M.S.)
| | - Alexander Kapustin
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (M.F., M.W., S.A., J.B., A.K., C.M.S.)
| | - Leon J. Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht CARIM, Maastricht University, the Netherlands (M.F., R.v.G., L.J.S.)
| | - Catherine M. Shanahan
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (M.F., M.W., S.A., J.B., A.K., C.M.S.)
| |
Collapse
|
42
|
Extracellular Matrix in Calcific Aortic Valve Disease: Architecture, Dynamic and Perspectives. Int J Mol Sci 2021; 22:ijms22020913. [PMID: 33477599 PMCID: PMC7831300 DOI: 10.3390/ijms22020913] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
Calcific Aortic Valve Disease (CAVD) is the most common valvular heart disease in developed countries and in the ageing population. It is strongly correlated to median age, affecting up to 13% of the population over the age of 65. Pathophysiological analysis indicates CAVD as a result of an active and degenerative disease, starting with sclerosis and chronic inflammation and then leaflet calcification, which ultimately can account for aortic stenosis. Although CAVD has been firstly recognized as a passive event mostly resulting from a degenerative aging process, much evidences suggests that calcification arises from different active processes, involving both aortic valve-resident cells (valve endothelial cells, valve interstitial cells, mesenchymal stem cells, innate immunity cells) and circulating cells (circulating mesenchymal cells, immunity cells). Moreover, a role for the cell-derived "matrix vesicles" and extracellular matrix (ECM) components has also been recognized. The aim of this work is to review the cellular and molecular alterations occurring in aortic valve during CAVD pathogenesis, focusing on the role of ECM in the natural course of the disease.
Collapse
|
43
|
Aikawa E, Blaser MC. 2020 Jeffrey M. Hoeg Award Lecture: Calcifying Extracellular Vesicles as Building Blocks of Microcalcifications in Cardiovascular Disorders. Arterioscler Thromb Vasc Biol 2021; 41:117-127. [PMID: 33115271 PMCID: PMC7832175 DOI: 10.1161/atvbaha.120.314704] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022]
Abstract
Cardiovascular calcification is an insidious form of ectopic tissue mineralization that presents as a frequent comorbidity of atherosclerosis, aortic valve stenosis, diabetes, renal failure, and chronic inflammation. Calcification of the vasculature and heart valves contributes to mortality in these diseases. An inability to clinically image or detect early microcalcification coupled with an utter lack of pharmaceutical therapies capable of inhibiting or regressing entrenched and detectable macrocalcification has led to a prominent and deadly gap in care for a growing portion of our rapidly aging population. Recognition of this mounting concern has arisen over the past decade and led to a series of revolutionary works that has begun to pull back the curtain on the pathogenesis, mechanistic basis, and causative drivers of cardiovascular calcification. Central to this progress is the discovery that calcifying extracellular vesicles act as active precursors of cardiovascular microcalcification in diverse vascular beds. More recently, the omics revolution has resulted in the collection and quantification of vast amounts of molecular-level data. As the field has become poised to leverage these resources for drug discovery, new means of deriving relevant biological insights from these rich and complex datasets have come into focus through the careful application of systems biology and network medicine approaches. As we look onward toward the next decade, we envision a growing need to standardize approaches to study this complex and multifaceted clinical problem and expect that a push to translate mechanistic findings into therapeutics will begin to finally provide relief for those impacted by this disease.
Collapse
Affiliation(s)
- Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Yaker L, Kamel S, Ausseil J, Boullier A. Effects of Chronic Kidney Disease and Uremic Toxins on Extracellular Vesicle Biology. Toxins (Basel) 2020; 12:toxins12120811. [PMID: 33371311 PMCID: PMC7767379 DOI: 10.3390/toxins12120811] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/02/2020] [Accepted: 12/16/2020] [Indexed: 12/28/2022] Open
Abstract
Vascular calcification (VC) is a cardiovascular complication associated with a high mortality rate, especially in patients with diabetes, atherosclerosis or chronic kidney disease (CKD). In CKD patients, VC is associated with the accumulation of uremic toxins, such as indoxyl sulphate or inorganic phosphate, which can have a major impact in vascular remodeling. During VC, vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and secrete extracellular vesicles (EVs) that are heterogeneous in terms of their origin and composition. Under physiological conditions, EVs are involved in cell-cell communication and the maintenance of cellular homeostasis. They contain high levels of calcification inhibitors, such as fetuin-A and matrix Gla protein. Under pathological conditions (and particularly in the presence of uremic toxins), the secreted EVs acquire a pro-calcifying profile and thereby act as nucleating foci for the crystallization of hydroxyapatite and the propagation of calcification. Here, we review the most recent findings on the EVs’ pathophysiological role in VC, the impact of uremic toxins on EV biogenesis and functions, the use of EVs as diagnostic biomarkers and the EVs’ therapeutic potential in CKD.
Collapse
Affiliation(s)
- Linda Yaker
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Avenue de la Croix Jourdain, F-80054 Amiens, France; (L.Y.); (S.K.)
| | - Saïd Kamel
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Avenue de la Croix Jourdain, F-80054 Amiens, France; (L.Y.); (S.K.)
- Laboratoire de Biochimie CHU Amiens-Picardie, Avenue de la Croix Jourdain, F-80054 Amiens, France
| | - Jérôme Ausseil
- INSERM UMR1043, CNRS UMR5282, University of Toulouse III, F-31024 Toulouse, France;
- CHU PURPAN—Institut Fédératif de Biologie, Laboratoire de Biochimie, Avenue de Grande Bretagne, F-31059 Toulouse, France
| | - Agnès Boullier
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Avenue de la Croix Jourdain, F-80054 Amiens, France; (L.Y.); (S.K.)
- Laboratoire de Biochimie CHU Amiens-Picardie, Avenue de la Croix Jourdain, F-80054 Amiens, France
- Correspondence: ; Tel.: +33-322087019
| |
Collapse
|
45
|
Biology and Biomechanics of the Heart Valve Extracellular Matrix. J Cardiovasc Dev Dis 2020; 7:jcdd7040057. [PMID: 33339213 PMCID: PMC7765611 DOI: 10.3390/jcdd7040057] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023] Open
Abstract
Heart valves are dynamic structures that, in the average human, open and close over 100,000 times per day, and 3 × 109 times per lifetime to maintain unidirectional blood flow. Efficient, coordinated movement of the valve structures during the cardiac cycle is mediated by the intricate and sophisticated network of extracellular matrix (ECM) components that provide the necessary biomechanical properties to meet these mechanical demands. Organized in layers that accommodate passive functional movements of the valve leaflets, heart valve ECM is synthesized during embryonic development, and remodeled and maintained by resident cells throughout life. The failure of ECM organization compromises biomechanical function, and may lead to obstruction or leaking, which if left untreated can lead to heart failure. At present, effective treatment for heart valve dysfunction is limited and frequently ends with surgical repair or replacement, which comes with insuperable complications for many high-risk patients including aged and pediatric populations. Therefore, there is a critical need to fully appreciate the pathobiology of biomechanical valve failure in order to develop better, alternative therapies. To date, the majority of studies have focused on delineating valve disease mechanisms at the cellular level, namely the interstitial and endothelial lineages. However, less focus has been on the ECM, shown previously in other systems, to be a promising mechanism-inspired therapeutic target. Here, we highlight and review the biology and biomechanical contributions of key components of the heart valve ECM. Furthermore, we discuss how human diseases, including connective tissue disorders lead to aberrations in the abundance, organization and quality of these matrix proteins, resulting in instability of the valve infrastructure and gross functional impairment.
Collapse
|
46
|
Mansour A, Darwiche W, Yaker L, Da Nascimento S, Gomila C, Rossi C, Jung V, Sonnet P, Kamel S, Guerrera IC, Boullier A, Ausseil J. GFOGER Peptide Modifies the Protein Content of Extracellular Vesicles and Inhibits Vascular Calcification. Front Cell Dev Biol 2020; 8:589761. [PMID: 33330469 PMCID: PMC7734313 DOI: 10.3389/fcell.2020.589761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/26/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Vascular calcification (VC) is an active process during which vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and release extracellular vesicles (EVs). In turn, the EVs serve as calcification foci via interaction with type 1 collagen (COL1). We recently showed that a specific, six-amino-acid repeat (GFOGER) in the sequence of COL1 was involved in the latter's interaction with integrins expressed on EVs. Our main objective was to test the GFOGER ability to inhibit VC. APPROACH We synthesized the GFOGER peptide and tested its ability to inhibit the inorganic phosphate (Pi)-induced calcification of VSMCs and aortic rings. Using mass spectrometry, we studied GFOGER's effect on the protein composition of EVs released from Pi-treated VSMCs. RESULTS Calcification of mouse VSMCs (MOVAS-1 cells), primary human VSMCs, and rat aortic rings was lower in the presence of GFOGER than with Pi alone (with relative decreases of 66, 58, and 91%, respectively; p < 0.001 for all) (no effect was observed with the scramble peptide GOERFG). A comparative proteomic analysis of EVs released from MOVAS-1 cells in the presence or absence of Pi highlighted significant differences in EVs' protein content. Interestingly, the expression of some of the EVs' proteins involved in the calcification process (such as osteogenic markers, TANK-binding kinase 1, and casein kinase II) was diminished in the presence of GFOGER peptide (data are available via ProteomeXchange with identifier PXD018169∗). The decrease of osteogenic marker expression observed in the presence of GFOGER was confirmed by q-RT-PCR analysis. CONCLUSION GFOGER peptide reduces vascular calcification by modifying the protein content of the subsequently released EVs, in particular by decreasing osteogenicswitching in VSMCs.
Collapse
Affiliation(s)
- Ali Mansour
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Walaa Darwiche
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Linda Yaker
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Sophie Da Nascimento
- AGIR, UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens, France
| | - Cathy Gomila
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Claire Rossi
- Alliance Sorbonne Université, Université de Technologie de Compiègne, UMR7025 CNRS Enzyme and Cell Engineering Laboratory, Compiègne, France
| | - Vincent Jung
- Plateforme protéomique Necker, Faculté de Médecine Paris Descartes, Université de Paris – Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Pascal Sonnet
- AGIR, UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens, France
| | - Saïd Kamel
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
- Laboratoire de Biochimie, Centre Hospitalier Universitaire d’ Amiens, Amiens, France
| | - Ida Chiara Guerrera
- Plateforme protéomique Necker, Faculté de Médecine Paris Descartes, Université de Paris – Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Agnès Boullier
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
- Laboratoire de Biochimie, Centre Hospitalier Universitaire d’ Amiens, Amiens, France
| | - Jérôme Ausseil
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR1043 – CNRS UMR5282 – Université Toulouse III, Toulouse, France
- CHU Toulouse – Institut Fédératif de Biologie, Laboratoire de Biochimie, Toulouse, France
| |
Collapse
|
47
|
Huang A, Guo G, Yu Y, Yao L. The roles of collagen in chronic kidney disease and vascular calcification. J Mol Med (Berl) 2020; 99:75-92. [PMID: 33236192 DOI: 10.1007/s00109-020-02014-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 01/16/2023]
Abstract
The extracellular matrix component collagen is widely expressed in human tissues and participates in various cellular biological processes. The collagen amount generally remains stable due to intricate regulatory networks, but abnormalities can lead to several diseases. During the development of renal fibrosis and vascular calcification, the expression of collagen is significantly increased, which promotes phenotypic changes in intrinsic renal cells and vascular smooth muscle cells, thereby exacerbating disease progression. Reversing the overexpression of collagen substantially prevents or slows renal fibrosis and vascular calcification in a wide range of animal models, suggesting a novel target for treating patients with these diseases. Stem cell therapy seems to be an effective strategy to alleviate these two conditions. However, recent findings indicate that the natural pore structure of collagen fibers is sufficient to induce the inappropriate differentiation of stem cells and thereby exacerbate renal fibrosis and vascular calcification. A comprehensive understanding of the role of collagen in these diseases and its effect on stem cell biology will assist in improving the unmet requirements for treating patients with kidney disease.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, 110000, China
| | - Guangying Guo
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, 110000, China
| | - Yanqiu Yu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, 110013, China. .,Shenyang Engineering Technology R&D Center of Cell Therapy Co. LTD., Shenyang, 110169, China.
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, 110000, China.
| |
Collapse
|
48
|
Donato M, Ferri N, Lupo MG, Faggin E, Rattazzi M. Current Evidence and Future Perspectives on Pharmacological Treatment of Calcific Aortic Valve Stenosis. Int J Mol Sci 2020; 21:ijms21218263. [PMID: 33158204 PMCID: PMC7663524 DOI: 10.3390/ijms21218263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS), the most common heart valve disease, is characterized by the slow progressive fibro-calcific remodeling of the valve leaflets, leading to progressive obstruction to the blood flow. CAVS is an increasing health care burden and the development of an effective medical treatment is a major medical need. To date, no effective pharmacological therapies have proven to halt or delay its progression to the severe symptomatic stage and aortic valve replacement represents the only available option to improve clinical outcomes and to increase survival. In the present report, the current knowledge and latest advances in the medical management of patients with CAVS are summarized, placing emphasis on lipid-lowering agents, vasoactive drugs, and anti-calcific treatments. In addition, novel potential therapeutic targets recently identified and currently under investigation are reported.
Collapse
Affiliation(s)
- Maristella Donato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Elisabetta Faggin
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
| | - Marcello Rattazzi
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
- Correspondence: ; Tel.: +39-0498-211-867 or +39-0422-322-207
| |
Collapse
|
49
|
Duan M, Zhao WL, Zhou L, Novák P, Zhu X, Yin K. Omics research in vascular calcification. Clin Chim Acta 2020; 511:319-328. [PMID: 33096035 DOI: 10.1016/j.cca.2020.10.022] [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] [Received: 08/31/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC), the pathological process of hydroxyapatite mineral deposition in the vascular system, is closely associated with aging, atherosclerotic plaque formation, cardiovascular disease (CVD) and diabetes mellitus (DM). Studies have shown that VC is related to cellular phenotypic changes, extracellular vesicles, disordered calcium and phosphate homeostasis, and an imbalance between inducers and inhibitors of VC. Unfortunately, there is currently no effective preventive or targeted treatment for pathologic condition. The rapid evolution of omics technology (genomics, epigenomics, transcriptomics, proteomics and metabolomics) has provided a novel approach for elucidation of pathophysiologic mechanisms in general and those associated with VC specifically. Here, we review articles published over the last twenty years and focus on the current state, challenges, limitations and future of omics in VC research and clinical practice. Highlighting potential targets based on omics technology will improve our understanding of this pathologic condition and assist in the development of potential treatment options for VC related disease.
Collapse
Affiliation(s)
- Meng Duan
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Wen-Li Zhao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| |
Collapse
|
50
|
Duan M, Zhao WL, Zhou L, Novák P, Zhu X, Yin K. Omics research in vascular calcification. Clin Chim Acta 2020; 511:198-207. [PMID: 33096032 DOI: 10.1016/j.cca.2020.10.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
Vascular calcification (VC), the pathological process of hydroxyapatite mineral deposition in the vascular system, is closely associated with aging, atherosclerotic plaque formation, cardiovascular disease (CVD) and diabetes mellitus (DM). Studies have shown that VC is related to cellular phenotypic changes, extracellular vesicles, disordered calcium phosphate homeostasis and an imbalance between inducers and inhibitors of VC. Unfortunately, there is currently no effective preventive or targeted treatment for this disorder. Recently, the evolution of omics technology (genomics, epigenomics, transcriptomics, proteomics and metabolomics) has paved the way for elucidation of complex biochemical processes and, as such, may provide new insight on VC. Accordingly, we conducted a review of articles published over the last twenty years and herein focus on current and future potential of omics technology in clarifying mechanisms of this disease process. Identification of new biomarkers will provide additional tools in characterizing this pathology and will further assist in the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Meng Duan
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Wen-Li Zhao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| |
Collapse
|