1
|
Gheorghe SR, Crăciun AM, Ilyés T, Tisa IB, Sur L, Lupan I, Samasca G, Silaghi CN. Converging Mechanisms of Vascular and Cartilaginous Calcification. BIOLOGY 2024; 13:565. [PMID: 39194503 DOI: 10.3390/biology13080565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Physiological calcification occurs in bones and epiphyseal cartilage as they grow, whereas ectopic calcification occurs in blood vessels, cartilage, and soft tissues. Although it was formerly thought to be a passive and degenerative process associated with aging, ectopic calcification has been identified as an active cell-mediated process resembling osteogenesis, and an increasing number of studies have provided evidence for this paradigm shift. A significant association between vascular calcification and cardiovascular risk has been demonstrated by various studies, which have shown that arterial calcification has predictive value for future coronary events. With respect to cartilaginous calcification, calcium phosphate or hydroxyapatite crystals can form asymptomatic deposits in joints or periarticular tissues, contributing to the pathophysiology of osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, tendinitis, and bursitis. The risk factors and sequence of events that initiate ectopic calcification, as well as the mechanisms that prevent the development of this pathology, are still topics of debate. Consequently, in this review, we focus on the nexus of the mechanisms underlying vascular and cartilaginous calcifications, trying to circumscribe the similarities and disparities between them to provide more clarity in this regard.
Collapse
Affiliation(s)
- Simona R Gheorghe
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandra M Crăciun
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Tamás Ilyés
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Badiu Tisa
- Department of Pediatrics III, Iuliu Hatieganu University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania
| | - Lucia Sur
- Department of Pediatrics I, Iuliu Hatieganu University of Medicine and Pharmacy, 400370 Cluj-Napoca, Romania
| | - Iulia Lupan
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Ciprian N Silaghi
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
2
|
Zhou R, Li R, Ding Q, Zhang Y, Yang H, Han Y, Liu C, Liu J, Wang S. OPN silencing reduces hypoxic pulmonary hypertension via PI3K-AKT-induced protective autophagy. Sci Rep 2024; 14:8670. [PMID: 38622371 PMCID: PMC11018812 DOI: 10.1038/s41598-024-59367-y] [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: 10/08/2023] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is a pulmonary vascular disease primarily characterized by progressive pulmonary vascular remodeling in a hypoxic environment, posing a significant clinical challenge. Leveraging data from the Gene Expression Omnibus (GEO) and human autophagy-specific databases, osteopontin (OPN) emerged as a differentially expressed gene, upregulated in cardiovascular diseases such as pulmonary arterial hypertension (PAH). Despite this association, the precise mechanism by which OPN regulates autophagy in HPH remains unclear, prompting the focus of this study. Through biosignature analysis, we observed significant alterations in the PI3K-AKT signaling pathway in PAH-associated autophagy. Subsequently, we utilized an animal model of OPNfl/fl-TAGLN-Cre mice and PASMCs with OPN shRNA to validate these findings. Our results revealed right ventricular hypertrophy and elevated mean pulmonary arterial pressure (mPAP) in hypoxic pulmonary hypertension model mice. Notably, these effects were attenuated in conditionally deleted OPN-knockout mice or OPN-silenced hypoxic PASMCs. Furthermore, hypoxic PASMCs with OPN shRNA exhibited increased autophagy compared to those in hypoxia alone. Consistent findings from in vivo and in vitro experiments indicated that OPN inhibition during hypoxia reduced PI3K expression while increasing LC3B and Beclin1 expression. Similarly, PASMCs exposed to hypoxia and PI3K inhibitors had higher expression levels of LC3B and Beclin1 and suppressed AKT expression. Based on these findings, our study suggests that OPNfl/fl-TAGLN-Cre effectively alleviates HPH, potentially through OPN-mediated inhibition of autophagy, thereby promoting PASMCs proliferation via the PI3K-AKT signaling pathway. Consequently, OPN emerges as a novel therapeutic target for HPH.
Collapse
Affiliation(s)
- Rui Zhou
- Qinghai University Medical Department, Xining, 810016, China
| | - Ran Li
- Zhengzhou Medical and Health Vocational College, Zhengzhou, 452385, China
| | - Qi Ding
- Pathology Department of Tianjin Huanghe Hospital, Tianjin, 300110, China
| | - Yuwei Zhang
- Department of Public Health, School of Medical, Qinghai University, Xining, 810016, China
| | - Hui Yang
- Qinghai University Medical Department, Xining, 810016, China
| | - Ying Han
- Qinghai University Medical Department, Xining, 810016, China
| | - Chuanchuan Liu
- Key Laboratory of Hydatid Disease, Qinghai University, Xining, 810001, China
| | - Jie Liu
- Qinghai University Medical Department, Xining, 810016, China
| | - Shenglan Wang
- Qinghai University Medical Department, Xining, 810016, China.
| |
Collapse
|
3
|
Kumar K, Rawat P, Kaur S, Singh N, Yadav HN, Singh D, Jaggi AS, Sethi D. Unveiling Wide Spectrum Therapeutic Implications and Signaling Mechanisms of Valsartan in Diverse Disorders: A Comprehensive Review. Curr Drug Res Rev 2024; 16:268-288. [PMID: 37461345 DOI: 10.2174/2589977515666230717120828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/27/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2024]
Abstract
Valsartan is an orally active non-peptide angiotensin receptor antagonist, an effective and well-tolerated anti-hypertensive drug. Besides its antihypertensive action, it has clinical implications in many other disorders, like heart failure (HF), arrhythmia, chronic kidney disease (CKD), diabetic complications (DM), atherosclerosis, etc. Besides angiotensin receptor blocking activity, valsartan reduces circulating levels of biochemical markers, such as hs-CRP, which is responsible for its anti-inflammatory and anti-oxidant activity. Moreover, valsartan also acts by inhibiting or inducing various signalling pathways, such as inducing autophagy via the AKT/mTOR/S6K pathway or inhibiting the TLR/NF-kB pathway. The current review exhaustively discusses the therapeutic implications of valsartan with specific emphasis on the mechanism of action in various disorders. The article provides a detailed spectrum of the therapeutic profile of valsartan and will likely be very useful to researchers working in the relevant research areas.
Collapse
Affiliation(s)
- Kuldeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Pooja Rawat
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Simrat Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Dhandeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Dimple Sethi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| |
Collapse
|
4
|
Fastrès A, Roels E, Tutunaru AC, Bolen G, Merveille A, Day MJ, Garigliany M, Antoine N, Clercx C. Osteopontin and fibronectin in lung tissue, serum, and bronchoalveolar lavage fluid of dogs with idiopathic pulmonary fibrosis and control dogs. J Vet Intern Med 2023; 37:2468-2477. [PMID: 37853926 PMCID: PMC10658509 DOI: 10.1111/jvim.16870] [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: 05/30/2023] [Accepted: 09/08/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) affects West Highland white terriers (WHWTs). Osteopontin (SPP1) and fibronectin (FN1) are associated with human IPF and are overexpressed by bronchoalveolar lavage fluid (BALF) macrophages in dogs with IPF. OBJECTIVE To investigate the value of these proteins as biomarkers of IPF. ANIMALS West Highland white terriers (WHWTs) with IPF, control WHWTs, and terriers. METHODS Cross-sectional observational study. Immunohistochemistry was used to localize SPP1 and FN1 in lung tissue. Serum and BALF SPP1 and FN1 concentrations were measured using canine ELISA kits and compared between groups. RESULTS Osteopontin stained ciliated epithelial cells, smooth muscular cells, and macrophages of all included dogs, and type-II pneumocytes and extracellular matrix of all 12 diseased WHWTs, 4/6 control WHWTs, and none of the 3 terriers. Osteopontin serum concentration was higher in diseased WHWTs (n = 22; 2.15 ng/mL [0.74-5.30]) compared with control WHWTs (n = 13; 0.63 ng/mL [0.41-1.63]; P = .005) and terriers (n = 15; 0.31 ng/mL [0.19-0.51]; P < .0001), and in control WHWTs compared with terriers (P = .005). Osteopontin BALF concentrations were higher in diseased (0.27 ng/mL [0.14-0.43]) and control WHWTs (0.25 ng/mL [0.14-0.40]), compared with terriers (0.02 ng/mL [0.01-0.08]; P < .0001 and P = .003, respectively). Fibronectin (FN1) serum concentrations were lower in diseased dogs (1.03 ng/mL [0.35-1.48]) and control WHWTs (0.61 ng/mL [0.24-0.65]) compared with terriers (2.72 ng/mL [0.15-5.21]; P < .0001 and P = .0001, respectively). There was no difference in FN1 immunostaining and FN1 BALF concentrations between groups. CONCLUSIONS Results suggest that SPP1 is involved in pathogenesis of IPF and could predispose that breed to the disease. Osteopontin serum concentration could serve as a diagnostic biomarker of IPF.
Collapse
Affiliation(s)
- Aline Fastrès
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Elodie Roels
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Alexandru C. Tutunaru
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Géraldine Bolen
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Anne‐Christine Merveille
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Michael J. Day
- School of Veterinary SciencesUniversity of BristolLangfordUnited Kingdom
| | - Mutien‐Marie Garigliany
- Department of Morphology and Pathology, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Nadine Antoine
- Department of Morphology and Pathology, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| | - Cécile Clercx
- Department of Clinical Sciences, FARAH, Faculty of Veterinary MedicineUniversity of LiègeLiègeBelgium
| |
Collapse
|
5
|
Mamazhakypov A, Maripov A, Sarybaev AS, Schermuly RT, Sydykov A. Osteopontin in Pulmonary Hypertension. Biomedicines 2023; 11:biomedicines11051385. [PMID: 37239056 DOI: 10.3390/biomedicines11051385] [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: 03/28/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Pulmonary hypertension (PH) is a pathological condition with multifactorial etiology, which is characterized by elevated pulmonary arterial pressure and pulmonary vascular remodeling. The underlying pathogenetic mechanisms remain poorly understood. Accumulating clinical evidence suggests that circulating osteopontin may serve as a biomarker of PH progression, severity, and prognosis, as well as an indicator of maladaptive right ventricular remodeling and dysfunction. Moreover, preclinical studies in rodent models have implicated osteopontin in PH pathogenesis. Osteopontin modulates a plethora of cellular processes within the pulmonary vasculature, including cell proliferation, migration, apoptosis, extracellular matrix synthesis, and inflammation via binding to various receptors such as integrins and CD44. In this article, we provide a comprehensive overview of the current understanding of osteopontin regulation and its impact on pulmonary vascular remodeling, as well as consider research issues required for the development of therapeutics targeting osteopontin as a potential strategy for the management of PH.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Abdirashit Maripov
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Akpay S Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Akylbek Sydykov
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| |
Collapse
|
6
|
Harris B, Saleem S, Cook N, Searle E. Targeting hypoxia in solid and haematological malignancies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:318. [PMID: 36320041 PMCID: PMC9628170 DOI: 10.1186/s13046-022-02522-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
Abstract
Tumour hypoxia is a known and extensively researched phenomenon that occurs in both solid and haematological malignancies. As cancer cells proliferate, demand for oxygen can outstrip supply reducing tumour oxygenation. In solid tumours this is contributed to by disorganized blood vessel development. Tumour hypoxia is associated with resistance to treatment, more aggressive disease behaviour and an increased likelihood of metastatic progression. It can be measured using both invasive and non-invasive methods to varying degrees of accuracy. The presence of hypoxia stimulates a complex cellular network of downstream factors including Hypoxia Inducible Factor 1 (HIF1), C-X-C motif chemokine 4 (CXCR4) and Hypoxia‐inducible glycolytic enzyme hexokinase‐2 (HK2) amongst many others. They work by affecting different mechanisms including influencing angiogenesis, treatment resistance, immune surveillance and the ability to metastasize all of which contribute to a more aggressive disease pattern. Tumour hypoxia has been correlated with poorer outcomes and worse prognosis in patients. The correlation between hypoxic microenvironments and poor prognosis has led to an interest in trying to therapeutically target this phenomenon. Various methods have been used to target hypoxic microenvironments. Hypoxia-activated prodrugs (HAPs) are drugs that are only activated within hypoxic environments and these agents have been subject to investigation in several clinical trials. Drugs that target downstream factors of hypoxic environments including HIF inhibitors, mammalian target of rapamycin (mTOR) inhibitors and vascular endothelial growth factor (anti-VEGF) therapies are also in development and being used in combination in clinical trials. Despite promising pre-clinical data, clinical trials of hypoxia targeting strategies have proven challenging. Further understanding of the effect of hypoxia and related molecular mechanisms in human rather than animal models is required to guide novel therapeutic strategies and future trial design. This review will discuss the currently available methods of hypoxia targeting and assessments that may be considered in planning future clinical trials. It will also outline key trials to date in both the solid and haemato-oncology treatment spheres and discuss the limitations that may have impacted on clinical success to date.
Collapse
Affiliation(s)
- Bill Harris
- grid.412917.80000 0004 0430 9259Experimental Cancer Medicine Team, Christie NHS Foundation Trust, Manchester, UK
| | - Sana Saleem
- grid.412917.80000 0004 0430 9259Haematology Department, Christie NHS Foundation Trust, Manchester, UK
| | - Natalie Cook
- grid.412917.80000 0004 0430 9259Experimental Cancer Medicine Team, Christie NHS Foundation Trust, Manchester, UK ,grid.5379.80000000121662407Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Emma Searle
- grid.412917.80000 0004 0430 9259Haematology Department, Christie NHS Foundation Trust, Manchester, UK ,grid.5379.80000000121662407Division of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
7
|
p38 MAPK priming boosts VSMC proliferation and arteriogenesis by promoting PGC1α-dependent mitochondrial dynamics. Sci Rep 2022; 12:5938. [PMID: 35396524 PMCID: PMC8994030 DOI: 10.1038/s41598-022-09757-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/28/2022] [Indexed: 12/05/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) proliferation is essential for arteriogenesis to restore blood flow after artery occlusion, but the mechanisms underlying this response remain unclear. Based on our previous findings showing increased VSMC proliferation in the neonatal aorta of mice lacking the protease MT4-MMP, we aimed at discovering new players in this process. We demonstrate that MT4-MMP absence boosted VSMC proliferation in vitro in response to PDGF-BB in a cell-autonomous manner through enhanced p38 MAPK activity. Increased phospho-p38 in basal MT4-MMP-null VSMCs augmented the rate of mitochondrial degradation by promoting mitochondrial morphological changes through the co-activator PGC1α as demonstrated in PGC1α−/− VSMCs. We tested the in vivo implications of this pathway in a novel conditional mouse line for selective MT4-MMP deletion in VSMCs and in mice pre-treated with the p38 MAPK activator anisomycin. Priming of p38 MAPK activity in vivo by the absence of the protease MT4-MMP or by anisomycin treatment led to enhanced arteriogenesis and improved flow recovery after femoral artery occlusion. These findings may open new therapeutic opportunities for peripheral vascular diseases.
Collapse
|
8
|
The value of plasma hypoxia markers for predicting imaging-based hypoxia in patients with head-and-neck cancers undergoing definitive chemoradiation. Clin Transl Radiat Oncol 2022; 33:120-127. [PMID: 35243023 PMCID: PMC8881198 DOI: 10.1016/j.ctro.2022.02.008] [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: 12/16/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Higher osteopontin plasma levels correlate with more hypoxic tumors at baseline. Increased baseline osteopontin levels are associated with residual tumor hypoxia. Absent early hypoxia response is linked with higher VEGF and CTGF levels in week 5. Plasma hypoxic markers may serve as biomarkers favoring radiotherapy personalization.
Background Methods Results Conclusion
Collapse
|
9
|
Grossmannova K, Barathova M, Belvoncikova P, Lauko V, Csaderova L, Tomka J, Dulka T, Pastorek J, Madaric J. Hypoxia Marker Carbonic Anhydrase IX Is Present in Abdominal Aortic Aneurysm Tissue and Plasma. Int J Mol Sci 2022; 23:ijms23020879. [PMID: 35055064 PMCID: PMC8778372 DOI: 10.3390/ijms23020879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/05/2023] Open
Abstract
Abdominal aortic aneurysms (AAA) are a significant cause of premature deaths worldwide. Since there is no specific treatment for reducing AAA progression, it is crucial to understand the pathogenesis leading to aneurysm wall weakening/remodeling and identify new proteins involved in this process which could subsequently serve as novel therapeutic targets. In this study, we analyzed the presence of the hypoxia-related proteins carbonic anhydrase IX (CA IX), hypoxia-inducible factor 1α (HIF-1α), and AKT as the key molecule in the phosphoinositide-3-kinase pathway in the AAA wall. Additionally, we used a blood-based assay to examine soluble CA IX (s-CA IX) levels in the plasma of AAA patients. Using western blotting, we detected CA IX protein in 12 out of 15 AAA tissue samples. Immunohistochemistry staining proved CA IX expression in the media of the aneurysmal wall. Evaluation of phosphorylated (p-AKT) and total AKT showed elevated levels of both forms in AAA compared to normal aorta. Using ELISA, we determined the concentration of s-CA IX >20 pg/mL in 13 out of 15 AAA patients. Results obtained from in silico analysis of CA9 and aneurysm-associated genes suggest a role for CA IX in aneurysmal wall remodeling. Our results prove the presence of hypoxia-related CA IX in AAA tissues and indicate a possible role of CA IX in hypoxia-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Katarina Grossmannova
- Department of Cancer Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská Cesta 9, 84505 Bratislava, Slovakia; (K.G.); (P.B.); (L.C.)
| | - Monika Barathova
- Department of Cancer Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská Cesta 9, 84505 Bratislava, Slovakia; (K.G.); (P.B.); (L.C.)
- Correspondence: ; Tel.: +421-2-59302439
| | - Petra Belvoncikova
- Department of Cancer Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská Cesta 9, 84505 Bratislava, Slovakia; (K.G.); (P.B.); (L.C.)
| | - Viliam Lauko
- Department of Laboratory Medicine, National Institute of Cardiovascular Disease, Pod Krásnou Hôrkou 1, 83101 Bratislava, Slovakia;
| | - Lucia Csaderova
- Department of Cancer Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská Cesta 9, 84505 Bratislava, Slovakia; (K.G.); (P.B.); (L.C.)
| | - Jan Tomka
- Department of Vascular Surgery, National Institute of Cardiovascular Disease, Pod Krásnou Hôrkou 1, 83101 Bratislava, Slovakia; (J.T.); (T.D.)
| | - Tomas Dulka
- Department of Vascular Surgery, National Institute of Cardiovascular Disease, Pod Krásnou Hôrkou 1, 83101 Bratislava, Slovakia; (J.T.); (T.D.)
| | | | - Juraj Madaric
- Department of Angiology, National Institute of Cardiovascular Disease, Pod Krásnou Hôrkou 1, 83101 Bratislava, Slovakia;
| |
Collapse
|
10
|
Manning EP, Ramachandra AB, Schupp JC, Cavinato C, Raredon MSB, Bärnthaler T, Cosme C, Singh I, Tellides G, Kaminski N, Humphrey JD. Mechanisms of Hypoxia-Induced Pulmonary Arterial Stiffening in Mice Revealed by a Functional Genetics Assay of Structural, Functional, and Transcriptomic Data. Front Physiol 2021; 12:726253. [PMID: 34594238 PMCID: PMC8478173 DOI: 10.3389/fphys.2021.726253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023] Open
Abstract
Hypoxia adversely affects the pulmonary circulation of mammals, including vasoconstriction leading to elevated pulmonary arterial pressures. The clinical importance of changes in the structure and function of the large, elastic pulmonary arteries is gaining increased attention, particularly regarding impact in multiple chronic cardiopulmonary conditions. We establish a multi-disciplinary workflow to understand better transcriptional, microstructural, and functional changes of the pulmonary artery in response to sustained hypoxia and how these changes inter-relate. We exposed adult male C57BL/6J mice to normoxic or hypoxic (FiO2 10%) conditions. Excised pulmonary arteries were profiled transcriptionally using single cell RNA sequencing, imaged with multiphoton microscopy to determine microstructural features under in vivo relevant multiaxial loading, and phenotyped biomechanically to quantify associated changes in material stiffness and vasoactive capacity. Pulmonary arteries of hypoxic mice exhibited an increased material stiffness that was likely due to collagen remodeling rather than excessive deposition (fibrosis), a change in smooth muscle cell phenotype reflected by decreased contractility and altered orientation aligning these cells in the same direction as the remodeled collagen fibers, endothelial proliferation likely representing endothelial-to-mesenchymal transitioning, and a network of cell-type specific transcriptomic changes that drove these changes. These many changes resulted in a system-level increase in pulmonary arterial pulse wave velocity, which may drive a positive feedback loop exacerbating all changes. These findings demonstrate the power of a multi-scale genetic-functional assay. They also highlight the need for systems-level analyses to determine which of the many changes are clinically significant and may be potential therapeutic targets.
Collapse
Affiliation(s)
- Edward P Manning
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Abhay B Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Jonas C Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States.,Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Micha Sam Brickman Raredon
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Thomas Bärnthaler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States.,Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Carlos Cosme
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Inderjit Singh
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - George Tellides
- VA Connecticut Healthcare System, West Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States
| |
Collapse
|
11
|
Pedrero-Prieto CM, Frontiñán-Rubio J, Alcaín FJ, Durán-Prado M, Peinado JR, Rabanal-Ruiz Y. Biological Significance of the Protein Changes Occurring in the Cerebrospinal Fluid of Alzheimer's Disease Patients: Getting Clues from Proteomic Studies. Diagnostics (Basel) 2021; 11:1655. [PMID: 34573996 PMCID: PMC8467255 DOI: 10.3390/diagnostics11091655] [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: 06/30/2021] [Revised: 08/18/2021] [Accepted: 08/26/2021] [Indexed: 11/16/2022] Open
Abstract
The fact that cerebrospinal fluid (CSF) deeply irrigates the brain together with the relative simplicity of sample extraction from patients make this biological fluid the best target for biomarker discovery in neurodegenerative diseases. During the last decade, biomarker discovery has been especially fruitful for the identification new proteins that appear in the CSF of Alzheimer's disease (AD) patients together with amyloid-β (Aβ42), total tau (T-tau), and phosphorylated tau (P-tau). Thus, several proteins have been already stablished as important biomarkers, due to an increase (i.e., CHI3L1) or a decrease (i.e., VGF) in AD patients' CSF. Notwithstanding this, only a deep analysis of a database generated with all the changes observed in CSF across multiple proteomic studies, and especially those using state-of-the-art methodologies, may expose those components or metabolic pathways disrupted at different levels in AD. Deep comparative analysis of all the up- and down-regulated proteins across these studies revealed that 66% of the most consistent protein changes in CSF correspond to intracellular proteins. Interestingly, processes such as those associated to glucose metabolism or RXR signaling appeared inversely represented in CSF from AD patients in a significant manner. Herein, we discuss whether certain cellular processes constitute accurate indicators of AD progression by examining CSF. Furthermore, we uncover new CSF AD markers, such as ITAM, PTPRZ or CXL16, identified by this study.
Collapse
Affiliation(s)
- Cristina M. Pedrero-Prieto
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha (UCLM), 13005 Ciudad Real, Spain
| | - Javier Frontiñán-Rubio
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Francisco J. Alcaín
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Mario Durán-Prado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Juan R. Peinado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Yoana Rabanal-Ruiz
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| |
Collapse
|
12
|
Osteopontin in Cardiovascular Diseases. Biomolecules 2021; 11:biom11071047. [PMID: 34356671 PMCID: PMC8301767 DOI: 10.3390/biom11071047] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Unprecedented advances in secondary prevention have greatly improved the prognosis of cardiovascular diseases (CVDs); however, CVDs remain a leading cause of death globally. These findings suggest the need to reconsider cardiovascular risk and optimal medical therapy. Numerous studies have shown that inflammation, pro-thrombotic factors, and gene mutations are focused not only on cardiovascular residual risk but also as the next therapeutic target for CVDs. Furthermore, recent clinical trials, such as the Canakinumab Anti-inflammatory Thrombosis Outcomes Study trial, showed the possibility of anti-inflammatory therapy for patients with CVDs. Osteopontin (OPN) is a matricellular protein that mediates diverse biological functions and is involved in a number of pathological states in CVDs. OPN has a two-faced phenotype that is dependent on the pathological state. Acute increases in OPN have protective roles, including wound healing, neovascularization, and amelioration of vascular calcification. By contrast, chronic increases in OPN predict poor prognosis of a major adverse cardiovascular event independent of conventional cardiovascular risk factors. Thus, OPN can be a therapeutic target for CVDs but is not clinically available. In this review, we discuss the role of OPN in the development of CVDs and its potential as a therapeutic target.
Collapse
|
13
|
Wendt TS, Li YJ, Gonzales RJ. Ozanimod, an S1PR 1 ligand, attenuates hypoxia plus glucose deprivation-induced autophagic flux and phenotypic switching in human brain VSM cells. Am J Physiol Cell Physiol 2021; 320:C1055-C1073. [PMID: 33788630 DOI: 10.1152/ajpcell.00044.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Vascular smooth muscle (VSM) cell phenotypic expression and autophagic state are dynamic responses to stress. Vascular pathologies, such as hypoxemia and ischemic injury, induce a synthetic VSM phenotype and autophagic flux resulting in a loss of vascular integrity and VSM cell death respectfully. Both clinical pilot and experimental stroke studies demonstrate that sphingosine-1-phosphate receptor (S1PR) modulation improves stroke outcome; however, specific mechanisms associated with a beneficial outcome at the level of the cerebrovasculature have not been clearly elucidated. We hypothesized that ozanimod, a selective S1PR type 1 ligand, will attenuate VSM synthetic phenotypic expression and autophagic flux in primary human brain VSM cells following acute hypoxia plus glucose deprivation (HGD; in vitro ischemic-like injury) exposure. Cells were treated with ozanimod and exposed to normoxia or HGD. Crystal violet staining, standard immunoblotting, and immunocytochemical labeling techniques assessed cellular morphology, vacuolization, phenotype, and autophagic state. We observed that HGD temporally decreased VSM cell viability and concomitantly increased vacuolization, both of which ozanimod reversed. HGD induced a simultaneous elevation and reduction in levels of pro- and antiautophagic proteins respectfully, and ozanimod attenuated this response. Protein levels of VSM phenotypic biomarkers, smoothelin and SM22, were decreased following HGD. Furthermore, we observed an HGD-induced epithelioid and synthetic morphological appearance accompanied by disorganized cytoskeletal filaments, which was rescued by ozanimod. Thus, we conclude that ozanimod, a selective S1PR1 ligand, protects against acute HGD-induced phenotypic switching and promotes cell survival, in part, by attenuating HGD-induced autophagic flux thus improving vascular patency in response to acute ischemia-like injury.
Collapse
Affiliation(s)
- Trevor S Wendt
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Yu Jing Li
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| | - Rayna J Gonzales
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona
| |
Collapse
|
14
|
Plasma osteopontin as a biomarker of Alzheimer's disease and vascular cognitive impairment. Sci Rep 2021; 11:4010. [PMID: 33597603 PMCID: PMC7889621 DOI: 10.1038/s41598-021-83601-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
Cerebrovascular disease (CeVD) and neurodegenerative dementia such as Alzheimer’s disease (AD) are frequently associated comorbidities in the elderly, sharing common risk factors and pathophysiological mechanisms including neuroinflammation. Osteopontin (OPN) is an inflammatory marker found upregulated in vascular diseases as well as in AD. However, its involvement in vascular dementia (VaD) and pre-dementia stages, namely cognitive impairment no dementia (CIND), both of which fall under the spectrum of vascular cognitive impairment (VCI), has yet to be examined. Its correlations with inflammatory cytokines in cognitive impairment also await investigation. 80 subjects with no cognitive impairment (NCI), 160 with CIND and 144 with dementia were included in a cross-sectional study on a Singapore-based memory clinic cohort. All subjects underwent comprehensive clinical, neuropsychological and brain neuroimaging assessments, together with clinical diagnoses based on established criteria. Blood samples were collected and OPN as well as inflammatory cytokines interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF) were measured using immunoassays. Multivariate regression analyses showed significant associations between increased OPN and VCI groups, namely CIND with CeVD, AD with CeVD and VaD. Interestingly, higher OPN was also significantly associated with AD even in the absence of CeVD. We further showed that increased OPN significantly associated with neuroimaging markers of CeVD and neurodegeneration, including cortical infarcts, lacunes, white matter hyperintensities and brain atrophy. OPN also correlated with elevated levels of IL-6, IL-8 and TNF. Our findings suggest that OPN may play a role in both VCI and neurodegenerative dementias. Further longitudinal analyses are needed to assess the prognostic utility of OPN in disease prediction and monitoring.
Collapse
|
15
|
Bai G, Matsuba T, Niki T, Hattori T. Stimulation of THP-1 Macrophages with LPS Increased the Production of Osteopontin-Encapsulating Exosome. Int J Mol Sci 2020; 21:ijms21228490. [PMID: 33187327 PMCID: PMC7696453 DOI: 10.3390/ijms21228490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Osteopontin (OPN) mediates bone remodeling and tissue debridement. The OPN protein is cleaved, but it is unclear how full-length (FL)-OPN or its cleaved form perform their biological activities in target cells. We, therefore, performed the molecular characterization of OPN in exosomes (Exo). The Exo were isolated from lipopolysaccharide (LPS)-stimulated phorbol 12-myristate 13-acetate (PMA)-differentiated THP-1 macrophages. The Exo were also isolated from PMA-differentiated THP-1 macrophages. The Exo were identified using the qNano multiple analyzer (diameter 59–315 nm) and western blotting with a CD9 antibody. LPS-stimulated cells produced more particles than non-stimulated cells. The presence of the FL or the cleaved form of OPN was confirmed using western blot analysis. A mixture of FL and cleaved OPN was also measured using an ELISA system (Ud-OPN) and their presence in the Exo was confirmed. Ud/FL ratios became low after LPS stimulation, indicating the enhanced encapsulation of FL-OPN in the Exo by LPS. These findings suggest that LPS stimulation of human macrophages facilitates the synthesis of FL-OPN, which is cleaved in cells or the Exo after release. These findings indicate that Exo is a suitable vehicle to transfer OPN to the target cells.
Collapse
Affiliation(s)
- Gaowa Bai
- Department of Health Science and Social Welfare, Kibi International University, Takahashi 716-8508, Japan;
| | - Takashi Matsuba
- Division of Bacteriology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan;
| | - Toshiro Niki
- Department of Immunology, Kagawa University, Kita-gun, Kagawa 7610793, Japan;
| | - Toshio Hattori
- Department of Health Science and Social Welfare, Kibi International University, Takahashi 716-8508, Japan;
- Correspondence: ; Tel.: +81-866-22-9469
| |
Collapse
|
16
|
Cheng CI, Chang HR, Tai MH, Chou MH, Chen GT, Chen PH, Kao YH. Hepatoma-derived growth factor enhances osteoblastic transformation of rat aortic vascular smooth muscle cells in vitro. Life Sci 2020; 256:117964. [DOI: 10.1016/j.lfs.2020.117964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023]
|
17
|
Wilson KS, Buist H, Suveizdyte K, Liles JT, Budas GR, Hughes C, MacLean MR, Johnson M, Church AC, Peacock AJ, Welsh DJ. Apoptosis signal-regulating kinase 1 inhibition in in vivo and in vitro models of pulmonary hypertension. Pulm Circ 2020; 10:2045894020922810. [PMID: 32523684 PMCID: PMC7235684 DOI: 10.1177/2045894020922810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 04/06/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension, group 1 of the pulmonary hypertension disease family, involves pulmonary vascular remodelling, right ventricular dysfunction and cardiac failure. Oxidative stress, through activation of mitogen-activated protein kinases is implicated in these changes. Inhibition of apoptosis signal-regulating kinase 1, an apical mitogen-activated protein kinase, prevented pulmonary arterial hypertension developing in rodent models. Here, we investigate apoptosis signal-regulating kinase 1 in pulmonary arterial hypertension by examining the impact that its inhibition has on the molecular and cellular signalling in established disease. Apoptosis signal-regulating kinase 1 inhibition was investigated in in vivo pulmonary arterial hypertension and in vitro pulmonary hypertension models. In the in vivo model, male Sprague Dawley rats received a single subcutaneous injection of Sugen SU5416 (20 mg/kg) prior to two weeks of hypobaric hypoxia (380 mmHg) followed by three weeks normoxia (Sugen/hypoxic), then animals were either maintained for three weeks on control chow or one containing apoptosis signal-regulating kinase 1 inhibitor (100 mg/kg/day). Cardiovascular measurements were carried out. In the in vitro model, primary cultures of rat pulmonary artery fibroblasts and rat pulmonary artery smooth muscle cells were maintained in hypoxia (5% O2) and investigated for proliferation, migration and molecular signalling in the presence or absence of apoptosis signal-regulating kinase 1 inhibitor. Sugen/hypoxic animals displayed significant pulmonary arterial hypertension compared to normoxic controls at eight weeks. Apoptosis signal-regulating kinase 1 inhibitor decreased right ventricular systolic pressure to control levels and reduced muscularised vessels in lung tissue. Apoptosis signal-regulating kinase 1 inhibition was found to prevent hypoxia-induced proliferation, migration and cytokine release in rat pulmonary artery fibroblasts and also prevented rat pulmonary artery fibroblast-induced rat pulmonary artery smooth muscle cell migration and proliferation. Apoptosis signal-regulating kinase 1 inhibition reversed pulmonary arterial hypertension in the Sugen/hypoxic rat model. These effects may be a result of intrinsic changes in the signalling of adventitial fibroblast.
Collapse
Affiliation(s)
- Kathryn S Wilson
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Hanna Buist
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Kornelija Suveizdyte
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Colin Hughes
- Central Research Facility, University of Glasgow, Glasgow, UK
| | - Margaret R MacLean
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Martin Johnson
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Clydebank, UK
| | - Alistair C Church
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Clydebank, UK
| | - Andrew J Peacock
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Clydebank, UK
| | - David J Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.,Department of Biological and Biomedical Science, Glasgow Caledonian University, Glasgow, UK
| |
Collapse
|
18
|
Abstract
Inflammatory cytokines are necessary for an acute response to injury and the progressive healing process. However, when this acute response does not resolve and becomes chronic, the same proteins that once promoted healing then contribute to chronic inflammatory pathologies, such as atherosclerosis. OPN (Osteopontin) is a secreted matricellular cytokine that signals through integrin and CD44 receptors, is highly upregulated in acute and chronic inflammatory settings, and has been implicated in physiological and pathophysiologic processes. Evidence from the literature suggests that OPN may fit within the Goldilocks paradigm with respect to cardiovascular disease, where acute increases are protective, attenuate vascular calcification, and promote postischemic neovascularization. In contrast, chronic increases in OPN are clinically associated with an increased risk for a major adverse cardiovascular event, and OPN expression is a strong predictor of cardiovascular disease independent of traditional risk factors. With the recent finding that humans express multiple OPN isoforms as the result of alternative splicing and that these isoforms have distinct biologic functions, future studies are required to determine what OPN isoform(s) are expressed in the setting of vascular disease and what role each of these isoforms plays in vascular disease progression. This review aims to discuss our current understanding of the role(s) of OPN in vascular disease pathologies using evidence from in vitro, animal, and clinical studies. Where possible, we discuss what is known about OPN isoform expression and our understanding of OPN isoform contributions to cardiovascular disease pathologies.
Collapse
Affiliation(s)
- Zoe Shin Yee Lok
- Department of Surgery, School of Clinical Sciences, Monash Health, Clayton, Australia (Z.S.Y.L.)
| | - Alicia N Lyle
- From the Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (A.N.L.)
| |
Collapse
|
19
|
Shi J, Fan J, Su Q, Yang Z. Cytokines and Abnormal Glucose and Lipid Metabolism. Front Endocrinol (Lausanne) 2019; 10:703. [PMID: 31736870 PMCID: PMC6833922 DOI: 10.3389/fendo.2019.00703] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/30/2019] [Indexed: 12/20/2022] Open
Abstract
Clear evidence indicates that cytokines, for instance, adipokines, hepatokines, inflammatory cytokines, myokines, and osteokines, contribute substantially to the development of abnormal glucose and lipid metabolism. Some cytokines play a positive role in metabolism action, while others have a negative metabolic role linking to the induction of metabolic dysfunction. The mechanisms involved are not fully understood, but are associated with lipid accumulation in organs and tissues, especially in the adipose and liver tissue, changes in energy metabolism, and inflammatory signals derived from various cell types, including immune cells. In this review, we describe the roles of certain cytokines in the regulation of metabolism and inter-organ signaling in regard to the pathophysiological aspects. Given the disease-related changes in circulating levels of relevant cytokines, these factors may serve as biomarkers for the early detection of metabolic disorders. Moreover, based on preclinical studies, certain cytokines that can induce improvements in glucose and lipid metabolism and immune response may emerge as novel targets of broader and more efficacious treatments and prevention of metabolic disease.
Collapse
Affiliation(s)
- Jie Shi
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiangao Fan
- Shanghai Key Laboratory of Children's Digestion and Nutrition, Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhen Yang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Irion CI, Parrish K, John-Williams K, Gultekin SH, Shehadeh LA. Osteopontin Expression in Cardiomyocytes Is Increased in Pediatric Patients With Sepsis or Pneumonia. Front Physiol 2018; 9:1779. [PMID: 30618794 PMCID: PMC6295581 DOI: 10.3389/fphys.2018.01779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/23/2018] [Indexed: 11/15/2022] Open
Abstract
Sepsis and pneumonia are major causes of death in the United States, and their pathophysiology includes infection with inflammation and immune dysfunction. Both sepsis and pneumonia cause cardiovascular dysfunction. The expression of Osteopontin (OPN) in cardiomyocytes of patients with sepsis or pneumonia, and its role the induced cardiac dysfunction have not been thoroughly investigated. OPN is a matricellular protein synthesized by multiple diseased tissues and cells including cardiomyocytes. Here, we studied the expression of OPN protein using immunofluorescence in human myocardial autopsy tissues from pediatric and mid age or elderly patients with sepsis and/or pneumonia. Fourteen human myocardial tissues from six pediatric patients and eight mid-age or elderly patients were studied. Immunofluorescence was used to investigate the expression of OPN in paraffin-embedded heart sections co-stained with the myocyte markers Actin Alpha 1 (ACTA1) and Myosin Light Chain 2 (MLC2). A quantitative analysis was performed to determine the number of ACTA1 and MLC2 positive cardiomyocytes that express OPN. The results showed that OPN expression was significantly increased in cardiomyocytes in the hearts from pediatric patients with sepsis and/or pneumonia (N = 3) relative to pediatric patients without sepsis/pneumonia (N = 3), or adult to elderly patients with sepsis/pneumonia (N = 5). Among the older septic hearts, higher levels of cardiomyocyte OPN expression was seen only in conjunction with severe coronary arterial occlusion. This is the first study to document increased OPN expression in cardiomyocytes of pediatric subjects with sepsis or pneumonia. Our findings highlight a potentially important role for OPN in sepsis- or pneumonia-mediated cardiac dysfunction in pediatric patients.
Collapse
Affiliation(s)
- Camila Iansen Irion
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Kiera Parrish
- Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Krista John-Williams
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Sakir H Gultekin
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
21
|
Bendix EF, Johansen E, Ringgaard T, Wolder M, Starup-Linde J. Diabetes and Abdominal Aortic Calcification-a Systematic Review. Curr Osteoporos Rep 2018; 16:42-57. [PMID: 29380116 DOI: 10.1007/s11914-018-0418-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW A systematic literature review was performed to evaluate diabetes mellitus (DM) as a risk factor of abdominal aortic calcification (AAC), and address factors that might contribute to the development of AAC in DM patients. RECENT FINDINGS DM is an independent risk factor of AAC development. Bone metabolism along with lifestyle factors among DM patients makes them more prone to AAC. Hip and vertebral fractures, high phosphate, smoking, hypertension, and low osteocalcin could make DM patients prone to AAC. Low levels of high-density lipoprotein (HDL), high low-density lipoprotein (LDL), high total cholesterol/HDL ratio, low bone mineral density (BMD) may be risk factors, but the literature is more ambiguous. Body mass index (BMI) does not appear to increase risk of AAC. High phosphate levels and low osteocalcin levels seem to be biomarkers of AAC in patients with diabetes. However, the association between DM and AAC is complicated.
Collapse
Affiliation(s)
- Emilie Frey Bendix
- Department of Endocrinology, Aalborg University Hospital, Mølleparkvej 4, 9220, Aalborg Øst, Denmark
- Faculty of Health, Aalborg University, Niels Jernes Vej 10, 9220, Aalborg Øst, Denmark
| | - Eskild Johansen
- Department of Endocrinology, Aalborg University Hospital, Mølleparkvej 4, 9220, Aalborg Øst, Denmark
- Faculty of Health, Aalborg University, Niels Jernes Vej 10, 9220, Aalborg Øst, Denmark
| | - Thomas Ringgaard
- Department of Endocrinology, Aalborg University Hospital, Mølleparkvej 4, 9220, Aalborg Øst, Denmark
- Faculty of Health, Aalborg University, Niels Jernes Vej 10, 9220, Aalborg Øst, Denmark
| | - Martin Wolder
- Department of Endocrinology, Aalborg University Hospital, Mølleparkvej 4, 9220, Aalborg Øst, Denmark
- Faculty of Health, Aalborg University, Niels Jernes Vej 10, 9220, Aalborg Øst, Denmark
| | - Jakob Starup-Linde
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Tage Hansens Gade 2, 8000, Aarhus C, Denmark.
| |
Collapse
|
22
|
ATP-mediated Events in Peritubular Cells Contribute to Sterile Testicular Inflammation. Sci Rep 2018; 8:1431. [PMID: 29362497 PMCID: PMC5780482 DOI: 10.1038/s41598-018-19624-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/03/2018] [Indexed: 12/19/2022] Open
Abstract
Peritubular myoid cells, which form the walls of seminiferous tubules in the testis, are functionally unexplored. While they transport sperm and contribute to the spermatogonial stem cell niche, specifically their emerging role in the immune surveillance of the testis and in male infertility remains to be studied. Recently, cytokine production and activation of Toll-like receptors (TLRs) were uncovered in cultured peritubular cells. We now show that human peritubular cells express purinergic receptors P2RX4 and P2RX7, which are functionally linked to TLRs, with P2RX4 being the prevalent ATP-gated ion channel. Subsequent ATP treatment of cultured peritubular cells resulted in up-regulated (pro-)inflammatory cytokine expression and secretion, while characteristic peritubular proteins, that is smooth muscle cell markers and extracellular matrix molecules, decreased. These findings indicate that extracellular ATP may act as danger molecule on peritubular cells, able to promote inflammatory responses in the testicular environment.
Collapse
|
23
|
Tang XG, Wen J, Zhang XS, Jiang DC. Association between decreased osteopontin and acute mountain sickness upon rapid ascent to 3500 m among young Chinese men. J Travel Med 2018; 25:5079665. [PMID: 30165389 DOI: 10.1093/jtm/tay075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/23/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Hypoxia causes oxidative stress and a decrease in osteopontin (OPN) in rats; however, little is known about the change in OPN in lowlander humans during hypobaric hypoxia. We explore the role of the predicted decrease in plasma OPN levels in humans upon high-altitude exposure and its relationship with acute mountain sickness (AMS), as well as superoxide dismutase (SOD) and malondialdehyde (MDA). METHODS Before and during acute altitude exposure, 261 men's plasma OPN, SOD, MDA, heart rate and pulse oximeter saturation (SpO2) were measured. AMS as assessed using the Lake Louise score (LLS) was defined as headache with a total LLS ≥3. Subjects were divided into AMS-0 (non-AMS subjects), mild AMS (headache with total LLS = 3 or 4) and severe AMS groups (headache with total LLS ≥5). RESULTS At 600 m, no difference in plasma OPN, SOD and MDA was observed between groups. At 3500 m, plasma OPN in severe AMS group was significantly decreased as compared with 600 m. Plasma SOD showed a tendency to decrease during altitude exposure. The opposite trend was observed for plasma MDA. Correlation analysis showed that total LLS was significantly correlated with OPN (ρ = -0.247, P < 0.001) and SOD (ρ = -0.224, P < 0.001). OPN showed significant correlation with SOD (r = 0.235, P < 0.001). Multivariate logistic regression analysis showed that higher plasma OPN was a protective factor for AMS [adjusted odds ratio (OR) 0.924, 95% confidence interval (CI) 0.884-0.966, P < 0.01]. CONCLUSION Our results suggest that decreased plasma OPN is correlated with AMS, and oxidative stress may be implicated in this phenomenon. Decreased plasma SOD is also correlated with AMS.
Collapse
Affiliation(s)
- Xu-Gang Tang
- Department of Cardiology, No. 37 Hospital of People's Liberation Army, Ya'an, Sichuan Province, People's Republic of China
| | - Jing Wen
- Department of Pediatrics, People's Hospital of Hechuan district, Chongqing, People's Republic of China
| | - Xue-Sen Zhang
- Department of Cardiology, No. 37 Hospital of People's Liberation Army, Ya'an, Sichuan Province, People's Republic of China
| | - Da-Chun Jiang
- Department of Cardiology, No. 37 Hospital of People's Liberation Army, Ya'an, Sichuan Province, People's Republic of China
| |
Collapse
|
24
|
Yu SH, Yoo HJ, Kang DH, Moon SJ, Yu JM. Intermittent High Glucose Enhances the Proliferation of Rat Aortic Vascular Smooth Muscle Cells More Than Constant High Glucose via the Mitogen-Activated Protein Kinase Pathway. Ann Geriatr Med Res 2017. [DOI: 10.4235/agmr.2017.21.3.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
25
|
Rani K, Aung NY. Docosahexaenoic Acid Inhibits Vascular Smooth Muscle Cell Proliferation Induced by Glucose Variability. Open Biochem J 2017; 11:56-65. [PMID: 28839472 PMCID: PMC5543665 DOI: 10.2174/1874091x01711010056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 02/23/2017] [Accepted: 03/17/2017] [Indexed: 12/02/2022] Open
Abstract
Background: Vascular Smooth Muscle cells (VSMC) enact crucial roles in early vasculogenesis and sustenance of vascular integrity. However, aberrant proliferation of VSMC followed by migration into the blood vessel wall leads to the formation of vascular lesions accounting for the degeneration and remodelling of vascular basement membrane. In diabetes, hyperglycaemia accelerates VSMC proliferation and contributes to the initiation and progression of atherosclerotic lesions. Recently, acute glucose fluctuations have been implicated in the abnormal VSMC proliferation and complications of diabetic atherosclerosis. Docosahexaenoic acid (DHA), a ω-3 polyunsaturated fatty acid (PUFA) has been shown to inhibit proliferation of several cell types implicating several different mechanisms. In the present study, we have investigated the effects of DHA on VSMC proliferation induced by stable and intermittent high glucose levels. Method: Confluent cultures of rat aortic VSMCs were treated with DHA for 24 hrs and then exposed to stable high glucose (25 mmol/L, SHG) or intermittent high glucose (5 mmol/L and 25 mmol/L alternating every 12 hrs, IHG) for 72 hrs. Cell proliferation was examined by the MTT viability assay, while apoptosis process was evaluated by the Hoechst staining, flow cytometry and caspase-3 activity assays. Results: Our data demonstrated that the hyper proliferation induced by stable and intermittent high glucose levels was significantly inhibited by the DHA pre-treatment. DHA significantly increased caspase-3 activity, resulting in enhanced DNA fragmentation and apoptosis. Conclusion: Our results suggest that DHA reduced the high glucose-induced proliferation of VSMC and induced cell apoptosis.
Collapse
Affiliation(s)
| | - Nway Y Aung
- Nanyang Technological University Singapore, Singapore
| |
Collapse
|
26
|
Jiang P, Zhang D, Qiu H, Yi X, Zhang Y, Cao Y, Zhao B, Xia Z, Wang C. Tiron ameliorates high glucose-induced cardiac myocyte apoptosis by PKCδ-dependent inhibition of osteopontin. Clin Exp Pharmacol Physiol 2017; 44:760-770. [PMID: 28394420 DOI: 10.1111/1440-1681.12762] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/17/2017] [Accepted: 03/31/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Ping Jiang
- Department of Cardiovascular Medicine; The People's Hospital of Gongan County; Gongan China
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Deling Zhang
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Hong Qiu
- Department of Laboratory; Dongfeng General Hospital of Hubei Medical University; Shiyan China
| | - Xianqi Yi
- Department of Cardiovascular Medicine; The People's Hospital of Gongan County; Gongan China
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Yemin Zhang
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Yingkang Cao
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Bo Zhao
- Department of Anesthesiology; Wuhan University Renmin Hospital; Wuhan China
| | - Zhongyuan Xia
- Department of Anesthesiology; Wuhan University Renmin Hospital; Wuhan China
| | - Changhua Wang
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| |
Collapse
|
27
|
Immunomodulatory effects of adipose tissue-derived stem cells on elastin scaffold remodeling in diabetes. Tissue Eng Regen Med 2016; 13:701-712. [PMID: 30603451 DOI: 10.1007/s13770-016-0018-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 01/11/2023] Open
Abstract
Diabetes is a major risk factor for the progression of vascular disease, contributing to elevated levels of glycoxidation, chronic inflammation and calcification. Tissue engineering emerges as a potential solution for the treatment of vascular diseases however there is a considerable gap in the understanding of how scaffolds and stem cells will perform in patients with diabetes. We hypothesized that adipose tissue-derived stem cells (ASCs) by virtue of their immunosuppressive potential would moderate the diabetes-intensified inflammatory reactions and induce positive construct remodeling. To test this hypothesis, we prepared arterial elastin scaffolds seeded with autologous ASCs and implanted them subdermally in diabetic rats and compared inflammatory markers, macrophage polarization, matrix remodeling, calcification and bone protein expression to control scaffolds implanted with and without cells in nondiabetic rats. ASC-seeded scaffolds exhibited lower levels of CD8+ T-cells and CD68+ pan-macrophages and higher numbers of M2 macrophages, smooth muscle cell-like and fibroblast-like cells. Calcification and osteogenic markers were reduced in ASCseeded scaffolds implanted in non-diabetic rats but remained unchanged in diabetes, unless the scaffolds were first pre-treated with penta-galloyl glucose (PGG), a known anti-oxidative elastin-binding polyphenol. In conclusion, autologous ASC seeding in elastin scaffolds is effective in combating diabetes-related complications. To prevent calcification, the oxidative milieu needs to be reduced by elastin-binding antioxidants such as PGG.
Collapse
|
28
|
Wogonin suppresses osteopontin expression in adipocytes by activating PPARα. Acta Pharmacol Sin 2015; 36:987-97. [PMID: 26073326 PMCID: PMC4564880 DOI: 10.1038/aps.2015.37] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/30/2015] [Indexed: 12/14/2022] Open
Abstract
AIM Wogonin (5,7-dihydroxy-8-methoxyflavone), a major bioactive compound of the flavonoid family, is commonly extracted from the traditional Chinese medicine Scutellaria baicalensis and possesses antioxidant and anti-inflammatory activities and is assumed to have anti-diabetes function. Indeed, a current study has shown that it can possibly treat metabolic disorders such as those found in db/db mice. However, the underlying molecular mechanism remains largely unclear. The aim of this study was to investigate the impact of wogonin on osteopontin (OPN) expression in adipose tissue from type 1 diabetic mice and in 3T3-L1 adipocytes. METHODS Type 1 diabetes was induced by streptozotocin (STZ) injection. 3T3-L1 preadipocytes were converted to 3T3-L1 adipocytes through treatment with insulin, dexamethasone, and 3-isobutyl-1-methylxanthine (IBMX). Western blot analysis and RT-PCR were performed to detect protein expression and mRNA levels, respectively. RESULTS Wogonin treatment suppressed the increase in serum OPN levels and reduced OPN expression in adipose tissue from STZ-induced type 1 diabetic mice. Administration of wogonin enhanced PPARα expression and activity. Silencing of PPARα diminished the inhibitory effects of wogonin on OPN expression in 3T3-L1 adipocytes. Furthermore, the levels of c-Fos and phosphorylated c-Jun were reduced in wogonin-treated adipose tissue and 3T3-L1 adipocytes. In addition, wogonin treatment dramatically mitigated p38 MAPK phosphorylation. Pharmacological inhibition of p38 MAPK by its specific inhibitor SB203580 increased PPARα activity and decreased OPN expression. CONCLUSION Our results suggest that wogonin downregulated OPN expression in adipocytes through the inhibition of p38 MAPK and the sequential activation of the PPARα pathway. Given the adverse effects of high OPN levels on metabolism, our results provide evidence for the potential administration of wogonin as a treatment for diabetes.
Collapse
|
29
|
Chen J, Green J, Yurdagul A, Albert P, McInnis MC, Orr AW. αvβ3 Integrins Mediate Flow-Induced NF-κB Activation, Proinflammatory Gene Expression, and Early Atherogenic Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [PMID: 26212910 DOI: 10.1016/j.ajpath.2015.05.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endothelial cell interactions with transitional matrix proteins, such as fibronectin, occur early during atherogenesis and regulate shear stress-induced endothelial cell activation. Multiple endothelial cell integrins bind transitional matrix proteins, including α5β1, αvβ3, and αvβ5. However, the role these integrins play in mediating shear stress-induced endothelial cell activation remains unclear. Therefore, we sought to elucidate which integrin heterodimers mediate shear stress-induced endothelial cell activation and early atherogenesis. We now show that inhibiting αvβ3 integrins (S247, siRNA), but not α5β1 or αvβ5, blunts shear stress-induced proinflammatory signaling (NF-κB, p21-activated kinase) and gene expression (ICAM1, VCAM1). Importantly, inhibiting αvβ3 did not affect cytokine-induced proinflammatory responses or inhibit all shear stress-induced signaling, because Akt, endothelial nitric oxide synthase, and extracellular regulated kinase activation remained intact. Furthermore, inhibiting αv integrins (S247), but not α5 (ATN-161), in atherosclerosis-prone apolipoprotein E knockout mice significantly reduced vascular remodeling after acute induction of disturbed flow. S247 treatment similarly reduced early diet-induced atherosclerotic plaque formation associated with both diminished inflammation (expression of vascular cell adhesion molecule 1, plaque macrophage content) and reduced smooth muscle incorporation. Inducible, endothelial cell-specific αv integrin deletion similarly blunted inflammation in models of disturbed flow and diet-induced atherogenesis but did not affect smooth muscle incorporation. Our studies identify αvβ3 as the primary integrin heterodimer mediating shear stress-induced proinflammatory responses and as a key contributor to early atherogenic inflammation.
Collapse
Affiliation(s)
- Jie Chen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Jonette Green
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Arif Yurdagul
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Patrick Albert
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Marshall C McInnis
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - A Wayne Orr
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana.
| |
Collapse
|
30
|
Jovic S, Shikhagaie M, Mörgelin M, Erjefält JS, Kjellström S, Egesten A. Osteopontin is increased in cystic fibrosis and can skew the functional balance between ELR-positive and ELR-negative CXC-chemokines. J Cyst Fibros 2015; 14:453-63. [DOI: 10.1016/j.jcf.2014.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 12/13/2022]
|
31
|
Zhang M, Sara JD, Wang FL, Liu LP, Su LX, Zhe J, Wu X, Liu JH. Increased plasma BMP-2 levels are associated with atherosclerosis burden and coronary calcification in type 2 diabetic patients. Cardiovasc Diabetol 2015; 14:64. [PMID: 26003174 PMCID: PMC4450848 DOI: 10.1186/s12933-015-0214-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/08/2015] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Although Bone morphogenetic protein-2 (BMP-2) is a known mediator of bone regeneration and vascular calcification, to date no study has investigated the relationship between BMP-2 and type 2 diabetes mellitus (T2DM) and its possible role in coronary artery disease (CAD). The purpose of this study is to evaluate the relationship of BMP-2 with atherosclerosis and calcification in patients with T2DM. METHODS 124 subjects were enrolled in this study: 29 patients with T2DM and CAD; 26 patients with T2DM and without CAD; 36 patients with CAD and without T2DMand 34 without T2DM or CAD (control group). Severity of coronary lesions was assessed using coronary angiography and intravascular ultrasound (IVUS). Plasma BMP-2 levels were quantified using a commercially available ELISA kit. RESULTS Compared to the control group, the mean plasma BMP-2 level was significantly higher in T2DM patients with or without CAD (20.1 ± 1.7 or 19.3 ± 1.5 pg/ml, vs 17.2 ± 3.3 pg/ml, P < 0.001). In a multivariable linear regression analysis, both T2DM and CAD were significantly and positively associated with BMP-2 (Estimate, 0.249; standard error (SE), 0.063; p <0.0001; Estimate, 0.400; SE, 0.06; p < 0.0001). Plasma BMP-2 was also strongly correlated with glycosylated hemoglobin A1c (HbA1c) (Spearman ρ = -0.31; p = 0.0005). SYNTAX score was also significantly associated with BMP-2 (Spearman ρ = 0.46; p = 0.0002). Using the results from IVUS, plasma BMP-2 levels were shown to positively correlate with plaque burden (Spearman ρ = 0.38, P = 0.002) and plaque calcification (Spearman ρ =0.44, P = 0.0003) and to negatively correlate with lumen volume (Spearman ρ =0.31, P = 0.01). CONCLUSIONS Our study demonstrates that patients with T2DM had higher circulating levels of BMP-2 than normal controls. Plasma BMP-2 levels correlated positively with plaque burden and calcification in patients with T2DM.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, PR China.
- Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| | - Jaskanwal Deep Sara
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA.
| | - Fei-long Wang
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA.
| | - Li-Ping Liu
- Department of Nephrology, First Hospital of Tsinghua University, Beijing, China.
| | - Li-Xiao Su
- Department of Biostatistics, Rutgers School of Public Health, The State University of New Jersey, Piscataway, NJ, USA.
| | - Jing Zhe
- Department of Biostatistics, University at Buffalo, the State University of New York, Buffalo, NY, 14214, USA.
| | - Xi Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, PR China.
- Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| | - Jing-hua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, PR China.
| |
Collapse
|
32
|
Mohamed IA, Gadeau AP, Fliegel L, Lopaschuk G, Mlih M, Abdulrahman N, Fillmore N, Mraiche F. Na+/H+ exchanger isoform 1-induced osteopontin expression facilitates cardiomyocyte hypertrophy. PLoS One 2015; 10:e0123318. [PMID: 25884410 PMCID: PMC4401699 DOI: 10.1371/journal.pone.0123318] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 03/02/2015] [Indexed: 01/02/2023] Open
Abstract
Enhanced expression and activity of the Na+/H+ exchanger isoform 1 (NHE1) has been implicated in cardiomyocyte hypertrophy in various experimental models. The upregulation of NHE1 was correlated with an increase in osteopontin (OPN) expression in models of cardiac hypertrophy (CH), and the mechanism for this remains to be delineated. To determine whether the expression of active NHE1-induces OPN and contributes to the hypertrophic response in vitro, cardiomyocytes were infected with the active form of the NHE1 adenovirus or transfected with OPN silencing RNA (siRNA-OPN) and characterized for cardiomyocyte hypertrophy. Expression of NHE1 in cardiomyocytes resulted in a significant increase in cardiomyocyte hypertrophy markers: cell surface area, protein content, ANP mRNA and expression of phosphorylated-GATA4. NHE1 activity was also significantly increased in cardiomyocytes expressing active NHE1. Interestingly, transfection of cardiomyocytes with siRNA-OPN significantly abolished the NHE1-induced cardiomyocyte hypertrophy. siRNA-OPN also significantly reduced the activity of NHE1 in cardiomyocytes expressing NHE1 (68.5±0.24%; P<0.05), confirming the role of OPN in the NHE1-induced hypertrophic response. The hypertrophic response facilitated by NHE1-induced OPN occurred independent of the extracellular-signal-regulated kinases and Akt, but required p90-ribosomal S6 kinase (RSK). The ability of OPN to facilitate the NHE1-induced hypertrophic response identifies OPN as a potential therapeutic target to reverse the hypertrophic effect induced by the expression of active NHE1.
Collapse
Affiliation(s)
| | - Alain-Pierre Gadeau
- University of Bordeaux, Adaptation Cardiovasculaire à L'ischémie, UMR1034, Pessac, France
| | - Larry Fliegel
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gary Lopaschuk
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Mohamed Mlih
- College of Pharmacy, Qatar University, Doha, Qatar
| | | | - Natasha Fillmore
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Fatima Mraiche
- College of Pharmacy, Qatar University, Doha, Qatar
- * E-mail:
| |
Collapse
|
33
|
Patouraux S, Rousseau D, Rubio A, Bonnafous S, Lavallard VJ, Lauron J, Saint-Paul MC, Bailly-Maitre B, Tran A, Crenesse D, Gual P. Osteopontin deficiency aggravates hepatic injury induced by ischemia-reperfusion in mice. Cell Death Dis 2014; 5:e1208. [PMID: 24810044 PMCID: PMC4047890 DOI: 10.1038/cddis.2014.174] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 02/26/2014] [Accepted: 03/18/2014] [Indexed: 12/17/2022]
Abstract
Osteopontin (OPN) is a multifunctional protein involved in hepatic steatosis, inflammation, fibrosis and cancer progression. However, its role in hepatic injury induced by ischemia–reperfusion (I–R) has not yet been investigated. We show here that hepatic warm ischemia for 45 min followed by reperfusion for 4 h induced the upregulation of the hepatic and systemic level of OPN in mice. Plasma aspartate aminotransferase and alanine aminotransferase levels were strongly increased in Opn−/− mice compared with wild-type (Wt) mice after I–R, and histological analysis of the liver revealed a significantly higher incidence of necrosis of hepatocytes. In addition, the expression levels of inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNFα), interleukin 6 (IL6) and interferon-γ were strongly upregulated in Opn−/− mice versus Wt mice after I–R. One explanation for these responses could be the vulnerability of the OPN-deficient hepatocyte. Indeed, the downregulation of OPN in primary and AML12 hepatocytes decreased cell viability in the basal state and sensitized AML12 hepatocytes to cell death induced by oxygen–glucose deprivation and TNFα. Further, the downregulation of OPN in AML12 hepatocytes caused a strong decrease in the expression of anti-apoptotic Bcl2 and in the ATP level. The hepatic expression of Bcl2 also decreased in Opn−/− mice versus Wt mice livers after I–R. Another explanation could be the regulation of the macrophage activity by OPN. In RAW macrophages, the downregulation of OPN enhanced iNOS expression in the basal state and sensitized macrophages to inflammatory signals, as evaluated by the upregulation of iNOS, TNFα and IL6 in response to lipopolysaccharide. In conclusion, OPN partially protects from hepatic injury and inflammation induced in this experimental model of liver I–R. This could be due to its ability to partially prevent death of hepatocytes and to limit the production of toxic iNOS-derived NO by macrophages.
Collapse
Affiliation(s)
- S Patouraux
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France [3] Centre Hospitalier Universitaire de Nice, Pôle Biologique, Hôpital Pasteur, Nice, France
| | - D Rousseau
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France
| | - A Rubio
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France
| | - S Bonnafous
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France [3] Centre Hospitalier Universitaire de Nice, Pôle Digestif, Hôpital L'Archet, Nice, France
| | - V J Lavallard
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France
| | - J Lauron
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France
| | - M-C Saint-Paul
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France [3] Centre Hospitalier Universitaire de Nice, Pôle Biologique, Hôpital Pasteur, Nice, France
| | - B Bailly-Maitre
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France
| | - A Tran
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France [3] Centre Hospitalier Universitaire de Nice, Pôle Digestif, Hôpital L'Archet, Nice, France
| | - D Crenesse
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France [3] Centre Hospitalier Universitaire de Nice, Hôpitaux Pédiatriques CHU Lenval, Nice, France
| | - P Gual
- 1] INSERM, U1065, Centre Méditerranéen de médecine Moléculaire (C3M), Équipe 8 « Complications hépatiques de l'obésité», Nice, France [2] Université de Nice-Sophia-Antipolis, Faculté de Médecine, Nice, France
| |
Collapse
|
34
|
Osteopontin: A novel regulator at the cross roads of inflammation, obesity and diabetes. Mol Metab 2014; 3:384-93. [PMID: 24944898 PMCID: PMC4060362 DOI: 10.1016/j.molmet.2014.03.004] [Citation(s) in RCA: 269] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/10/2014] [Accepted: 03/13/2014] [Indexed: 02/06/2023] Open
Abstract
Since its first description more than 20 years ago osteopontin has emerged as an active player in many physiological and pathological processes, including biomineralization, tissue remodeling and inflammation. As an extracellular matrix protein and proinflammatory cytokine osteopontin is thought to facilitate the recruitment of monocytes/macrophages and to mediate cytokine secretion in leukocytes. Modulation of immune cell response by osteopontin has been associated with various inflammatory diseases and may play a pivotal role in the development of adipose tissue inflammation and insulin resistance. Here we summarize recent findings on the role of osteopontin in metabolic disorders, particularly focusing on diabetes and obesity.
Collapse
|
35
|
Peroxisome proliferator-activated receptor γ ligands retard cultured vascular smooth muscle cells calcification induced by high glucose. Cell Biochem Biophys 2014; 66:421-9. [PMID: 23274912 DOI: 10.1007/s12013-012-9490-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) and its ligands have profound effects on glucose homeostasis, cardiovascular diseases, and bone metabolism. To explore the pathophysiological roles of PPARγ in diabetes with concomitant vascular calcification, we investigated changes in PPARγ expression and the effect of the PPARγ ligands troglitazone and rosiglitazone on vascular smooth muscle cell (VSMC) calcification induced by high glucose (HG, 25 mmol/L). Compared with low glucose, HG-induced VSMC calcification, and PPARγ mRNA, protein level was decreased. Troglitazone and rosiglitazone treatment markedly attenuated the VSMC calcification, whereas PPARγ antagonist GW9662 abolished the effect of rosiglitazone on calcification. Pretreatment of VSMCs with rosiglitazone, but not troglitazone, restored the loss of lineage marker expression: the protein levels of α-actin and SM-22α were increased 52 % (P < 0.05) and 53.1% (P < 0.01), respectively, as compared with HG alone. Troglitazone and rosiglitazone reversed the change in bone-related protein expression induced by HG: decreased the mRNA levels of osteocalcin, bone morphogenetic protein 2 (BMP2), and core binding factor α 1 (Cbfα-1) by 26.9% (P > 0.05), 50.0 % (P < 0.01), and 24.4% (P < 0.05), and 48.4% (P < 0.05), 41.4% (P < 0.01) and 56.2% (P < 0.05), respectively, and increased that of matrix Gla protein (MGP) 84.2% (P < 0.01) and 70.0%, respectively (P < 0.05), as compared with HG alone. GW9662 abolished the effect of rosiglitazone on Cbfα-1 and MGP expression. PPARγ ligands can inhibit VSMCs calcification induced by high glucose.
Collapse
|
36
|
Atorvastatin inhibits hyperglycemia-induced expression of osteopontin in the diabetic rat kidney via the p38 MAPK pathway. Mol Biol Rep 2014; 41:2551-8. [PMID: 24452713 DOI: 10.1007/s11033-014-3113-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 01/10/2014] [Indexed: 01/13/2023]
Abstract
Osteopontin (OPN), a large phosphoglycoprotein adhesion molecule, which is up-regulated in the kidneys of humans and mice with diabetes, has emerged as a potentially key pathophysiological contributor in diabetic nephropathy. Here, we investigated the role of OPN in kidney injury caused by diabetic nephropathy and the effect of atorvastatin on the expression of OPN and on diabetic nephropathy. Diabetes was induced with streptozotocin in rats, and atorvastatin (5 mg/kg) was orally administered once a day for 8 weeks. We analyzed the expression and regulation of OPN in the kidneys of streptozotocin-induced diabetic Sprague-Dawley albino rats by immunohistochemistry and western blot analysis. The expression of OPN was increased in diabetic rat kidney, and atorvastatin inhibited this process. Atorvastatin also decreased the expression and phosphorylation of p38. In vitro, atorvastatin inhibited the high glucose-induced OPN expression in Madin-Darby canine kidney epithelial cells through the p38 MAPK signaling pathway. These results suggested that atorvastatin reduced the expression of OPN through inhibition of the p38 MAPK pathway. The expression of OPN was associated with kidney injury. These molecules may represent therapeutic targets for the prevention of acute kidney injury induced by diabetes.
Collapse
|
37
|
Yu M, Gong D, Lim M, Arutyunyan A, Groffen J, Heisterkamp N. Lack of bcr and abr promotes hypoxia-induced pulmonary hypertension in mice. PLoS One 2012; 7:e49756. [PMID: 23152932 PMCID: PMC3495860 DOI: 10.1371/journal.pone.0049756] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 10/16/2012] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Bcr and Abr are GTPase activating proteins that specifically downregulate activity of the small GTPase Rac in restricted cell types in vivo. Rac1 is expressed in smooth muscle cells, a critical cell type involved in the pathogenesis of pulmonary hypertension. The molecular mechanisms that underlie hypoxia-associated pulmonary hypertension are not well-defined. METHODOLOGY/PRINCIPAL FINDINGS Bcr and abr null mutant mice were compared to wild type controls for the development of pulmonary hypertension after exposure to hypoxia. Also, pulmonary arterial smooth muscle cells from those mice were cultured in hypoxia and examined for proliferation, p38 activation and IL-6 production. Mice lacking Bcr or Abr exposed to hypoxia developed increased right ventricular pressure, hypertrophy and pulmonary vascular remodeling. Perivascular leukocyte infiltration in the lungs was increased, and under hypoxia bcr-/- and abr-/- macrophages generated more reactive oxygen species. Consistent with a contribution of inflammation and oxidative stress in pulmonary hypertension-associated vascular damage, Bcr and Abr-deficient animals showed elevated endothelial leakage after hypoxia exposure. Hypoxia-treated pulmonary arterial smooth muscle cells from Bcr- or Abr-deficient mice also proliferated faster than those of wild type mice. Moreover, activated Rac1, phosphorylated p38 and interleukin 6 were increased in these cells in the absence of Bcr or Abr. Inhibition of Rac1 activation with Z62954982, a novel Rac inhibitor, decreased proliferation, p38 phosphorylation and IL-6 levels in pulmonary arterial smooth muscle cells exposed to hypoxia. CONCLUSIONS Bcr and Abr play a critical role in down-regulating hypoxia-induced pulmonary hypertension by deactivating Rac1 and, through this, reducing both oxidative stress generated by leukocytes as well as p38 phosphorylation, IL-6 production and proliferation of pulmonary arterial smooth muscle cells.
Collapse
Affiliation(s)
- Min Yu
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
| | - Dapeng Gong
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
| | - Min Lim
- Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Anna Arutyunyan
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
| | - John Groffen
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
- Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Nora Heisterkamp
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
- Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Wang L, Zhang J, Fu W, Guo D, Jiang J, Wang Y. Association of smooth muscle cell phenotypes with extracellular matrix disorders in thoracic aortic dissection. J Vasc Surg 2012; 56:1698-709, 1709.e1. [PMID: 22960022 DOI: 10.1016/j.jvs.2012.05.084] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 05/16/2012] [Accepted: 05/19/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Extracellular matrix dysregulation in the aortic media has been considered as the intrinsic factor for the formation of thoracic aortic dissection. However, the mechanisms of extracellular matrix disorders in the dissected aortic media remain unclear. This study was designed to investigate the relevance between smooth muscle cell phenotypes and extracellular matrix disorders in the dissected media. Their interaction may account for the pathogenesis of thoracic aortic dissection. METHODS AND RESULTS Thoracic aortic samples were collected from 10 patients with thoracic aortic dissection and 10 controls. Primary cultures of aortic medial smooth muscle cells were obtained with optimized explant technique. In this study, α-smooth muscle actin, smooth muscle myosin heavy chain 2, and smoothelin were applied as the contractile phenotypic markers and osteopontin was applied as the synthetic marker. Compared with controls, immunostaining and immunoblotting demonstrated that in vivo expression of α-smooth muscle actin, smooth muscle myosin heavy chain 2, and smoothelin were significantly decreased in the dissected media, whereas that of osteopontin was elevated (P<.01 for all). In vitro expression of the phenotypic markers showed the similar patterns. Furthermore, smooth muscle cells derived from the dissected media exhibited enhanced proliferation (P<.01), increased collagens I and III synthesis (2.6- and 4.4-fold, respectively; P<.01 for both), and elevated matrix metalloproteinase-2 production (4.2-fold; P<.01). Consistently, the protein levels of type I and III collagens and matrix metalloproteinase-2 in the dissected media were raised by 4.6-, 4.0-, and 3.7-fold, respectively (P<.01 for all). Collagen deposition was correspondingly increased and elastic fibers were decreased and disrupted. CONCLUSIONS Smooth muscle cells in the dissected media exhibit phenotypic switching from the contractile to the synthetic type. The synthetic smooth muscle cells increase collagen synthesis and matrix metalloproteinase-2 production, both of which can promote collagen deposition and elastin degradation in thoracic aortic dissection.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
39
|
Zhou YB, Zhang J, Cai Y, Teng X, Duan XH, Song JQ, Du J, Tang CS, Qi YF. Insulin resistance induces medial artery calcification in fructose-fed rats. Exp Biol Med (Maywood) 2012; 237:50-7. [PMID: 22238287 DOI: 10.1258/ebm.2011.011252] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Osteogenic differentiation of vascular smooth muscle cells (VSMCs) results in medial artery calcification, which is common in diabetes, but the pathogenesis is poorly understood. We aimed to explore the pathophysiological roles of insulin resistance (IR) on medial artery calcification in rats with 10% fructose in drinking water. After 12 weeks of fructose feeding, rats showed severe IR, with increased levels of fasting blood glucose, serum insulin and oral glucose tolerance test (OGTT). Fructose-fed rats showed aortic calcification, increased aortic calcium deposition and irregular elastic fibers in the medial layer of the vessel wall. Moreover, plasma phosphorus concentration, calcium × phosphorus product and alkaline phosphatase (ALP) activity, and aortic calcium content and ALP activity were significantly increased. Fructose feeding increased mRNA levels of osteopontin, type III sodium-dependent phosphate co-transporter, bone morphogenetic protein-2 and the key transcription factor core binding factor alpha 1 in aortic tissue and downregulated mRNA levels of osteoprotegerin and matrix γ-carboxyglutamic acid protein. Fructose feeding decreased protein levels of smooth-muscle lineage markers and induced severe lipid peroxidation injury. IR induced by high fructose feeding could evoke osteogenic transdifferentiation of VSMCs and promote vascular calcification.
Collapse
MESH Headings
- Alkaline Phosphatase/biosynthesis
- Alkaline Phosphatase/metabolism
- Animals
- Aorta, Thoracic/pathology
- Blood Glucose/metabolism
- Bone Morphogenetic Proteins/biosynthesis
- Bone Morphogenetic Proteins/genetics
- Calcium/analysis
- Calcium-Binding Proteins/biosynthesis
- Cell Differentiation
- Core Binding Factor Alpha 1 Subunit/biosynthesis
- Core Binding Factor Alpha 1 Subunit/genetics
- Dietary Carbohydrates/administration & dosage
- Extracellular Matrix Proteins/biosynthesis
- Fructose/administration & dosage
- Glucose Tolerance Test
- Insulin/blood
- Insulin Resistance
- Lipid Peroxidation
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Osteopontin/biosynthesis
- Osteopontin/genetics
- Osteoprotegerin/biosynthesis
- Phosphorus/blood
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Sodium-Phosphate Cotransporter Proteins, Type III/biosynthesis
- Sodium-Phosphate Cotransporter Proteins, Type III/genetics
- Tunica Media/pathology
- Vascular Calcification/pathology
- Vascular Calcification/physiopathology
- Matrix Gla Protein
Collapse
Affiliation(s)
- Ye-bo Zhou
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, Beijing 100029, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang Y, Han DD, Wang HM, Liu M, Zhang XH, Wang HL. Downregulation of osteopontin is associated with fluoxetine amelioration of monocrotaline-induced pulmonary inflammation and vascular remodelling. Clin Exp Pharmacol Physiol 2011; 38:365-72. [PMID: 21418086 DOI: 10.1111/j.1440-1681.2011.05516.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Osteopontin (OPN) has emerged as a key factor in inflammatory activation and cardiovascular remodelling. The aim of the present study was to investigate the involvement of OPN in fluoxetine amelioration of monocrotaline (MCT)-induced pulmonary inflammation and vascular remodelling in rats. 2. Wistar rats were divided into control, MCT and two fluoxetine-treated groups. Pulmonary arterial hypertension (PAH) was induced by a single injection of MCT (60 mg/kg, i.p.). Fluoxetine (2 and 10 mg/kg) was administered via the intragastric route once a day for 21 days. On Day 22, pulmonary haemodynamic measurements were undertaken, followed by ELISA, western blotting and immunohistochemistry. 3. Monocrotaline caused pulmonary inflammation and vascular remodelling and significantly enhanced OPN expression in the plasma, lungs and pulmonary arteries. Fluoxetine decreased pulmonary arterial pressure and ameliorated pulmonary inflammation and pulmonary vascular remodelling. At 10 mg/kg, fluoxetine significantly inhibited MCT-induced increases in the expression of serotonin transporter (SERT) and phosphorylated extracellular signal-regulated kinase 1/2 and downregulated the expression of OPN, macrophage inflammatory protein-1β and matrix metalloproteinase 2/tissue inhibitor of metalloproteinase 2. Although 2 mg/kg fluoxetine tended to ameliorate some MCT-induced changes in the lung, the differences did not always reach statistical significance. Linear regression analysis showed that there was a positive correlation between plasma OPN concentrations and mean pulmonary arterial pressure, as well as percentage medial wall thickness and percentage wall area in the pulmonary artery. 4. In conclusion, the amelioration by fluoxetine of MCT-induced pulmonary inflammation and vascular remodelling is associated with downregulation of OPN expression in rats.
Collapse
Affiliation(s)
- Yun Wang
- Department of Clinical Pharmacology, China Medical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
41
|
Cascella T, Radhakrishnan Y, Maile LA, Busby WH, Gollahon K, Colao A, Clemmons DR. Aldosterone enhances IGF-I-mediated signaling and biological function in vascular smooth muscle cells. Endocrinology 2010; 151:5851-64. [PMID: 20881255 PMCID: PMC2999491 DOI: 10.1210/en.2010-0350] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The IGF-I pathway and renin-angiotensin-aldosterone axis are both involved in the pathogenesis of hypertension and atherosclerosis, but no information is available about IGF-I and aldosterone interaction or their potential synergistic effects in vascular smooth muscle cells (VSMCs). The aims of this study were to investigate whether aldosterone influences IGF-I signaling and to determine the mechanism(s) by which aldosterone affects IGF-I function. Aldosterone resulted in significant increases in the Akt (1.87 ± 0.24, P < 0.001), MAPK (1.78 ± 0.13, P < 0.001), p70S6kinase (1.92 ± 0.15, P < 0.001), IGF-I receptor (1.69 ± 0.05, P < 0.01), and insulin receptor substrate-1 (1.7 ± 0.04, P < 0.01) (fold increase, mean ± SEM, n = 3) phosphorylation responses to IGF-I compared with IGF-I treatment alone. There were also significant increases in VSMC proliferation, migration, and protein synthesis (1.63 ± 0.03-, 1.56 ± 0.08-, and 1.51 ± 0.04-fold increases compared with IGF-I alone, respectively, n = 3, P < 0.001). Aldosterone induced osteopontin (OPN) mRNA expression and activation of αVβ3-integrin as well as an increase in the synthesis of IGF-I receptor. The enhancing effects of aldosterone were inhibited by eplerenone (10 μmol/liter), actinomycin-D (20 nmol/liter), and an anti-αVβ3-integrin antibody that blocks OPN binding. The antioxidant N-acetylcysteine (2 mmol/liter) completely inhibited the ability of aldosterone to induce any of these changes. In conclusion, our results show that aldosterone enhances IGF-I signaling and biological actions in VSMCs through induction of OPN followed by its subsequent activation of the αVβ3-integrin and by increasing IGF-I receptor. These changes are mediated in part through increased oxidative stress. The findings suggest a new mechanism by which aldosterone could accelerate the development of atherosclerosis.
Collapse
Affiliation(s)
- Teresa Cascella
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Maile LA, Busby WH, Nichols TC, Bellinger DA, Merricks EP, Rowland M, Veluvolu U, Clemmons DR. A monoclonal antibody against alphaVbeta3 integrin inhibits development of atherosclerotic lesions in diabetic pigs. Sci Transl Med 2010; 2:18ra11. [PMID: 20371482 DOI: 10.1126/scitranslmed.3000476] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atherosclerotic lesions develop and progress more rapidly in diabetic patients than in nondiabetic individuals. This may be caused by accelerated lesion formation in the high-glucose environment of diabetes. Smooth muscle cells (SMCs) cultured in high glucose are more responsive to growth factors such as insulin-like growth factor-1 (IGF-1). This enhanced response to IGF-1 is due in part to increased activation of the alpha(V)beta(3) integrin. We tested whether alpha(V)beta(3) integrin activation was increased in diabetic animals and whether an antibody to beta(3) would inhibit IGF-1 action and development of atherosclerosis. Eight male pigs were made diabetic with streptozotocin and fed a high-fat diet. A F(ab)(2) antibody fragment directed at beta(3) was infused into one femoral artery, whereas the other artery received control F(ab)(2) for 3.5 months. There was a 65 +/- 8% reduction in atherosclerotic lesion area in the arteries treated with F(ab)(2) antibody to beta(3). Phosphorylation of beta(3) was reduced by 75 +/- 18% in vessels treated with the antibody. Shc and mitogen-activated protein kinase phosphorylation, which are required for IGF-1-stimulated SMC proliferation, were also significantly reduced. We conclude that activation of IGF-1 receptor and alpha(V)beta(3)-linked signaling pathways accelerates atherosclerosis in diabetes and that administration of an antibody to beta(3) to diabetic pigs inhibits alpha(V)beta(3) activation, IGF-1-stimulated signaling, and atherosclerotic lesion development. This approach offers a potential therapeutic approach to the treatment of this disorder.
Collapse
Affiliation(s)
- Laura A Maile
- Division of Endocrinology, University of North Carolina, Chapel Hill, NC 27599-7170, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kurata M, Okura T, Irita J, Enomoto D, Nagao T, Jotoku M, Miyoshi K, Desilva VR, Higaki J. Angiotensin II receptor blockade with valsartan decreases plasma osteopontin levels in patients with essential hypertension. J Hum Hypertens 2010; 25:334-9. [PMID: 20664555 DOI: 10.1038/jhh.2010.73] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Osteopontin (OPN) has recently emerged as a key factor in both vascular remodelling and development of atherosclerosis. It has been reported that OPN is regulated by the renin-angiotensin-aldosterone system (RAAS). The aim of this study was to clarify the effect of angiotensin II receptor blockade with valsartan on plasma OPN levels in patients with essential hypertension (EHT). Forty-six patients (mean age, 64±11 years) with EHT were randomly assigned to treatment with amlodipine or valsartan. There were no significant differences in baseline clinical characteristics between the two groups. Blood sampling and blood pressure evaluation were performed before and after 24 weeks of treatment. After 24 weeks, both systolic blood pressure (SBP) and diastolic blood pressure (DBP) were decreased significantly and by the same degree in each treatment group. However, valsartan but not amlodipine decreased plasma OPN levels (baseline and 24-week data-valsartan: 614±224 ng ml(-1), 472±268 ng ml(-1), P=0.006; amlodipine: 680±151 ng ml(-1), 687±234 ng ml(-1), P>0.999). A positive correlation between the reduction in OPN and the log natural (ln) C-reactive protein (CRP) was seen in the valsartan-treated group. Stepwise regression analysis showed that treatment with valsartan and the reduction of ln CRP were associated with the reduction in OPN levels, and this association was independent of the reduction in SBP or aldosterone levels (valsartan: β=0.332, P=0.026; ln CRP reduction: β=0.366, P=0.015). These results suggest that suppression of the RAAS and inflammation may decrease plasma OPN levels.
Collapse
Affiliation(s)
- M Kurata
- Department of Integrated Medicine and Informatics, Ehime University Graduate School of Medicine, Shitsukawa, Toon City, Ehime, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Volkmer E, Kallukalam BC, Maertz J, Otto S, Drosse I, Polzer H, Bocker W, Stengele M, Docheva D, Mutschler W, Schieker M. Hypoxic preconditioning of human mesenchymal stem cells overcomes hypoxia-induced inhibition of osteogenic differentiation. Tissue Eng Part A 2010; 16:153-64. [PMID: 19642854 DOI: 10.1089/ten.tea.2009.0021] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Osteogenic differentiation of human mesenchymal stem cells (hMSCs) into osteoblasts is a prerequisite for subsequent bone formation. Numerous studies have explored osteogenic differentiation under standard tissue culture conditions, which usually employ 21% of oxygen. However, bone precursor cells such as hMSCs reside in stem cell niches of low-oxygen atmospheres. Furthermore, they are subjected to low oxygen concentrations when cultured on three-dimensional scaffolds in vitro, and even more so after transplantation when vascularization has yet to be established. Similarly, hMSCs are exposed to low oxygen in the fracture microenvironment following bony injury. Recent studies revealed that hypoxic preconditioning improves cellular engraftment and survival in low-oxygen atmospheres. In our study we investigated the osteogenic differentiation potential of hMSCs under 2% O(2) (hypoxia) in comparison to a standard tissue culture oxygen atmosphere of 21% (normoxia). We assessed the osteogenic differentiation of hMSCs following hypoxic preconditioning to address whether this pretreatment is beneficial for subsequent differentiation processes as well. To validate our findings we carefully characterized the extent of hypoxia exerted and its effect on cell survival and proliferation. We found that hMSCs proliferate better if cultured under 2% of oxygen. We confirmed that osteogenic differentiation of hMSCs is indeed inhibited if osteogenic induction is carried out under constant hypoxia. Finally, we showed for the first time that hypoxic preconditioning of hMSCs prior to osteogenic induction restores osteogenic differentiation of hMSCs under hypoxic conditions. Collectively, our results indicate that maintaining constant levels of oxygen improves the osteogenic potential of hMSCs and suggest that low oxygen concentrations may preserve the stemness of hMSCs. In addition, our data support the hypothesis that if low-oxygen atmospheres are expected at the site of implantation, hypoxic pretreatment may be beneficial for the cells' subsequent in vivo performance.
Collapse
Affiliation(s)
- Elias Volkmer
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yuan XX, Ma XY, Wang FR, Wang Z, Wang X, Wang SH. Significance of osteopontin and hypoxia-inducible factor-α expression in colorectal carcinoma and adenoma. Shijie Huaren Xiaohua Zazhi 2009; 17:3174-3179. [DOI: 10.11569/wcjd.v17.i30.3174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of osteopontin (OPN) and hypoxia-inducible factor-α (HIF-1α) and analyze their correlation with clinical and pathophysiological parameters in colorectal carcinoma and adenoma.
METHODS: The expression of OPN and HIF-1α in 50 colorectal carcinoma specimens, 24 colorectal adenoma specimens and 12 normal colorectal tissues were examined by immunohistochemistry using the streptavidin-peroxidase method.
RESULTS: OPN and HIF-1α expression was not detected in normal colorectal tissues. In contrast, OPN and HIF-1α were highly expressed in colorectal carcinoma and adenoma. Although no significant difference was noted in OPN expression level between colorectal adenoma and carcinoma, significant difference was found in HIF-1α expression level between colorectal adenoma and carcinoma (33.33% vs 62.00%, P < 0.05). Both OPN and HIF-1α expression were positively correlated with lymph node metastasis, depth of tumor invasion and Dukes' stage, but not with patients' gender and age. A significant correlation was noted between the expression of OPN and HIF-1α in colorectal carcinoma (r = 0.700, P < 0.01).
CONCLUSION: OPN and HIF-1α may play an important role in the invasion and metastasis of colorectal carcinoma. Tumors expressing high level of HIF-1α have greater invasive and metastatic capacity.
Collapse
|
46
|
Kunii Y, Niwa SI, Hagiwara Y, Maeda M, Seitoh T, Suzuki T. The immunohistochemical expression profile of osteopontin in normal human tissues using two site-specific antibodies reveals a wide distribution of positive cells and extensive expression in the central and peripheral nervous systems. Med Mol Morphol 2009; 42:155-61. [PMID: 19784742 DOI: 10.1007/s00795-009-0459-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 06/03/2009] [Indexed: 12/22/2022]
Abstract
To elucidate the cellular distribution of osteopontin (OPN) in normal human tissues, we undertook immunohistochemistry using two site-specific OPN antibodies. The 10A16 monoclonal antibody was raised against the amino acid sequence just downstream of the thrombin cleavage site, while the O-17 polyclonal antibody was raised against the N-terminal peptide. Each antibody has been confirmed previously to react with both whole OPN and its relevant fragments. The expression pattern for these two antibodies was similar in distribution. In addition, we also identified expression in Ebner's gland, type II pneumocytes, Kupffer cells, cells of the endocrine organs, anterior lens capsule and ciliary body, synovial type A cells, mesothelia, adipocytes, and mast cells. Neurons and glia in the central nervous system and spinal cord, cranial and peripheral nerve sheaths, ganglion cells in the sympathetic ganglion, intestinal plexuses, retina, and choroid plexus also regularly exhibited OPN positivity. Testicular germ cells, pancreatic exocrine cells, and follicular dendritic cells reacted with 10A16 only, whereas lutein cells and taste bud cells exhibited O-17 reactivity alone. These minor differences were hypothesized to reflect the state of OPN in the cells; that is, whether OPN was in its whole molecule or fragmented form. In conclusion, we demonstrate that OPN is widely distributed in normal human cells, particularly those comprising the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Yasuto Kunii
- Department of Pathology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Xiao Y, Huang Z, Yin H, Lin Y, Wang S. In vitro differences between smooth muscle cells derived from varicose veins and normal veins. J Vasc Surg 2009; 50:1149-54. [PMID: 19703751 DOI: 10.1016/j.jvs.2009.06.048] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 06/08/2009] [Accepted: 06/21/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The theory of primary venous dilatation leading to secondary valvular incompetence and varicose vein formation has received more attention nowadays. Although many studies have investigated the role of the main components of the venous wall in the development of varicose veins, the leading cause remains unknown. The present study was designed to establish the role of smooth muscle cells (SMCs) of the tunica media on the pathogenesis of varicose veins by analyzing the phenotypic and functional differences between SMCs derived from varicose veins and normal veins. METHODS SMCs were isolated and cultured from saphenous veins of patients with varicose veins and normal veins. Cell proliferation and migration rates were compared. Expression of phenotype-dependent markers and matrix metalloproteinase-2 (MMP) production were analyzed by immunoblotting. Total collagen synthesis was evaluated by measuring the radioactivity of L-[3, 4-(3)H]proline in the media and the cell layer. RESULTS SMCs derived from varicose veins demonstrated increased proliferation (2-fold, P < .01), migration (3-fold, P < .001), MMP-2 production (3-fold, P < .01), and collagen synthesis (>2-fold, P < .001), with decreased expression of phenotype-dependent markers compared with SMCs derived from normal veins (P < .05). CONCLUSION SMCs derived from varicose veins are more dedifferentiated and demonstrate increased proliferative and synthetic capacity than SMCs derived from normal veins. These properties may contribute to the remodeling of the venous wall and the weakening of its antipressure capacity.
Collapse
Affiliation(s)
- Ying Xiao
- Research Center of Vascular Surgery, Department of Vascular Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Peoples Republic of China
| | | | | | | | | |
Collapse
|
48
|
Zhou H, Lu N, Chen ZQ, Song QL, Yu HM, Li XJ. Osteopontin mediates dense culture-induced proliferation and adhesion of prostate tumour cells: role of protein kinase C, p38 mitogen-activated protein kinase and calcium. Basic Clin Pharmacol Toxicol 2008; 104:164-70. [PMID: 19143755 DOI: 10.1111/j.1742-7843.2008.00348.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cells growing in high density were observed to undergo a variety of responses due to cell-cell contact, pericellular hypoxia, etc. In order to investigate the influence of cell density on cell proliferation and adhesion and to elucidate possible mechanisms, we tested the growth ability of human prostate tumour (PC-3M) cells in dense culture and the influences of density on cell adhesion. Our results demonstrate that increasing cell density exerted stress on PC-3M cells, which decreased cell proliferation in dense cultures, but tended to facilitate tumour metastasis since cell adhesion ability was elevated and the cells showed an increased growth rate after being moved to a favourable growth environment. We conclude that higher cell density-mediated pericellular hypoxia was an important factor inducing expression of the intrinsic hypoxia marker osteopontin, another mechanism contributing to cell adhesion enhancement in PC-3M cells. In addition, cell density enhanced adhesion ability due to the activation of p38 mitogen-activated protein kinase (p38 MAPK) and protein kinase C. Intracellular calcium also played positive roles at least partially through activating p38 MAPK.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Pharmacology, School of Basic Medical Sciences and State Key Laboratory of Natural & Biomimetic Drugs, Peking University, Beijing, China
| | | | | | | | | | | |
Collapse
|
49
|
Fong GH. Mechanisms of adaptive angiogenesis to tissue hypoxia. Angiogenesis 2008; 11:121-40. [PMID: 18327686 DOI: 10.1007/s10456-008-9107-3] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2007] [Accepted: 02/25/2008] [Indexed: 12/18/2022]
Abstract
Angiogenesis is mostly an adaptive response to tissue hypoxia, which occurs under a wide variety of situations ranging from embryonic development to tumor growth. In general, angiogenesis is dependent on the accumulation of hypoxia inducible factors (HIFs), which are heterodimeric transcription factors of alpha and beta subunits. Under normoxia, HIF heterodimers are not abundantly present due to oxygen dependent hydroxylation, polyubiquitination, and proteasomal degradation of alpha subunits. Under hypoxia, however, alpha subunits are stabilized and form heterodimers with HIF-1beta which is not subject to oxygen dependent regulation. The accumulation of HIFs under hypoxia allows them to activate the expression of many angiogenic genes and therefore initiates the angiogenic process. In recent years, however, it has become clear that various other mechanisms also participate in fine tuning angiogenesis. In this review, I discuss the relationship between hypoxia and angiogenesis under five topics: (1) regulation of HIF-alpha abundance and activity by oxygen tension and other conditions including oxygen independent mechanisms; (2) hypoxia-regulated expression of angiogenic molecules by HIFs and other transcription factors; (3) responses of vascular cells to hypoxia; (4) angiogenic phenotypes due to altered HIF signaling in mice; and (5) role of the HIF pathway in pathological angiogenesis. Studies discussed under these topics clearly indicate that while mechanisms of oxygen-regulated HIF-alpha stability provide exciting opportunities for the development of angiogenesis or anti-angiogenesis therapies, it is also highly important to consider various other mechanisms for the optimization of these procedures.
Collapse
Affiliation(s)
- Guo-Hua Fong
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3501, USA.
| |
Collapse
|
50
|
Raheja LF, Genetos DC, Yellowley CE. Hypoxic osteocytes recruit human MSCs through an OPN/CD44-mediated pathway. Biochem Biophys Res Commun 2007; 366:1061-6. [PMID: 18155656 DOI: 10.1016/j.bbrc.2007.12.076] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2007] [Accepted: 12/13/2007] [Indexed: 02/07/2023]
Abstract
Little is known about the role or identity of signaling molecules released by osteocytes to recruit MSCs to areas of matrix damage. Vascular disruption at fracture sites results in hypoxia which is known to up-regulate genes involved in cell migration including osteopontin (OPN). We examined the effect of conditioned media from hypoxic osteocytes on MSC migration. Hypoxic osteocyte media significantly increased MSC migration and expression of OPN was significantly increased in hypoxic osteocytes. OPN and CD44 neutralizing antibodies significantly reduced MSC migration. Further, recombinant OPN significantly increased MSC migration in a dose-dependent manner. Our data support the hypothesis that hypoxia at a fracture site stimulates the release of chemotactic factors, such as OPN, from osteocytes, that induce MSC migration to aid in fracture repair. To our knowledge, these are the first data to suggest a role for osteocytes and OPN in the recruitment of MSCs to aid in fracture repair.
Collapse
Affiliation(s)
- Leah Forquer Raheja
- Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, University of California, 4206 VM3A, 1285 Veterinary Medicine Drive, Davis, CA 95616, USA
| | | | | |
Collapse
|