1
|
Kajuluri LP, Guo YY, Lee S, Christof M, Malhotra R. Epigenetic Regulation of Human Vascular Calcification. Genes (Basel) 2025; 16:506. [PMID: 40428328 PMCID: PMC12111397 DOI: 10.3390/genes16050506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Vascular diseases present a significant threat to human health worldwide. Atherosclerosis is the most prevalent vascular disease, accounting for the majority of morbidity and mortality globally. Vascular calcification is a dynamic pathological process underlying the development of atherosclerotic plaques and involves the phenotypic transformation of vascular smooth muscle cells (VSMCs) into osteogenic cells. Specifically, the phenotypic switch in VSMCs often involves modifications in gene expression due to epigenetic changes, including DNA methylation, histone modification, and non-coding RNAs. Understanding the role of these epigenetic changes in regulating the pathophysiology of vascular calcification, along with the proteins and pathways that mediate these changes, will aid in identifying new therapeutic candidates to enhance vascular health. This review discusses a comprehensive range of epigenetic modifications and their implications for vascular health and the development of vascular calcification.
Collapse
Affiliation(s)
- Lova Prasadareddy Kajuluri
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Yugene Young Guo
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Sujin Lee
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Michael Christof
- School of Arts and Sciences, University of Rochester, Rochester, NY 14627, USA;
| | - Rajeev Malhotra
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| |
Collapse
|
2
|
Resuela-González JL, González-Gómez MJ, Rodríguez-Cano MM, López-López S, Monsalve EM, Díaz-Guerra MJM, Laborda J, Nueda ML, Baladrón V. NOTCH1, 2, and 3 receptors enhance osteoblastogenesis of mesenchymal C3H10T1/2 cells and inhibit this process in preosteoblastic MC3T3-E1 cells. Differentiation 2025; 142:100837. [PMID: 39879823 DOI: 10.1016/j.diff.2025.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
Osteoblastogenesis is governed by complex interplays among signaling pathways, which modulate the expression of specific markers at each differentiation stage. This process enables osteoblast precursor cells to adopt the morphological and biochemical characteristics of mature bone cells. Our study investigates the role of NOTCH signaling in osteogenesis in MC3T3-E1 and C3H10T1/2 cell lines. MC3T3-E1 cells are preosteoblast precursors widely recognized as a model for bone biology research, offering a convenient and physiologically relevant system to study osteoblast transcriptional regulation. Conversely, the mesenchymal C3H10T1/2 cells are multipotent, capable of differentiating into osteoblasts, adipocytes, and chondrocytes under specific extracellular cues. The core of this in vitro study is the comparative analysis of the impact of overexpressing each mammalian NOTCH receptor on osteoblastogenesis in two cell lines reflecting different cell differentiation stages. We generated stable transfectant pools of both cell lines for each of the four NOTCH receptors and characterized their effect on osteoblastogenesis. We successfully obtained transfectant pools that overexpress Notch1, Notch2 and Notch3 at both mRNA and protein levels. However, we were unable to obtain cells overexpressing Notch4 at protein level. Our findings reveal that the overexpression of NOTCH1, NOTCH2, and NOTCH3 receptors promotes osteoblast differentiation in mesenchymal C3H10T1/2 cells, while inhibiting it in preosteoblastic MC3T3-E1 cells. These results provide novel insights into the distinct roles of NOTCH receptors in osteoblastogenesis across two different precursor cell types, potentially guiding the development of new therapeutic approaches for bone diseases.
Collapse
Affiliation(s)
- Jose-Luis Resuela-González
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)/Instituto Nacional de Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de MonteGancedo UPM, Pozuelo de Alarcón, 28223, Spain; Departamento de Biotecnología-Biotecnología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040, Madrid, Spain
| | - María-Julia González-Gómez
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, ETSIAMB/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-Milagros Rodríguez-Cano
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Farmacia/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain
| | - Susana López-López
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, ETSIAMB/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain
| | - Eva-María Monsalve
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-José M Díaz-Guerra
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain
| | - Jorge Laborda
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Farmacia/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-Luisa Nueda
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Farmacia/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain.
| | - Victoriano Baladrón
- Área de Bioquímica y Biología Molecular, Departamento de Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina/IB-UCLM/Unidad de Biomedicina, Universidad de Castilla-La Mancha/CSIC, Albacete, Spain.
| |
Collapse
|
3
|
Llewellyn J, Baratam R, Culig L, Beerman I. Cellular stress and epigenetic regulation in adult stem cells. Life Sci Alliance 2024; 7:e202302083. [PMID: 39348938 PMCID: PMC11443024 DOI: 10.26508/lsa.202302083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024] Open
Abstract
Stem cells are a unique class of cells that possess the ability to differentiate and self-renew, enabling them to repair and replenish tissues. To protect and maintain the potential of stem cells, the cells and the environment surrounding these cells (stem cell niche) are highly responsive and tightly regulated. However, various stresses can affect the stem cells and their niches. These stresses are both systemic and cellular and can arise from intrinsic or extrinsic factors which would have strong implications on overall aging and certain disease states. Therefore, understanding the breadth of drivers, namely epigenetic alterations, involved in cellular stress is important for the development of interventions aimed at maintaining healthy stem cells and tissue homeostasis. In this review, we summarize published findings of epigenetic responses to replicative, oxidative, mechanical, and inflammatory stress on various types of adult stem cells.
Collapse
Affiliation(s)
- Joey Llewellyn
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Rithvik Baratam
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Luka Culig
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
4
|
Rodríguez-Cano MM, González-Gómez MJ, Monsalve EM, Díaz-Guerra MJM, Kassem M, Laborda J, Nueda ML, Baladrón V. DLK1 and DLK2, two non-canonical ligands of NOTCH receptors, differentially modulate the osteogenic differentiation of mesenchymal C3H10T1/2 cells. Biol Res 2024; 57:77. [PMID: 39473022 PMCID: PMC11523663 DOI: 10.1186/s40659-024-00561-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND C3H10T1/2 is a mesenchymal cell line capable of differentiating into osteoblasts, adipocytes and chondrocytes. The differentiation of these cells into osteoblasts is modulated by various transcription factors, such as RUNX2. Additionally, several interconnected signaling pathways, including the NOTCH pathway, play a crucial role in modulating their differentiation into mature bone cells. We have investigated the roles of DLK1 and DLK2, two non-canonical inhibitory ligands of NOTCH receptors, in the osteogenic differentiation of C3H10T1/2 cells. RESULTS Our results corroborate existing evidence that DLK1 acts as an inhibitor of osteogenesis. In contrast, we demonstrate for the first time that DLK2 enhances this differentiation process. Additionally, our data suggest that NOTCH2, 3 and 4 receptors may promote osteogenesis, as indicated by their increased expression during this process, whereas NOTCH1 expression, which decreases during cell differentiation, might inhibit osteogenesis. Moreover, treatment with DAPT, a NOTCH signaling inhibitor, impeded osteogenic differentiation. We have confirmed the increase in ERK1/2 MAPK and p38 MAPK phosphorylation in C3H10T1/2 cells induced to differentiate to osteoblasts. Our new findings reveal increased ERK1/2 MAPK phosphorylation in differentiated C3H10T1/2 cells with a decrease in DLK1 expression or an overexpression of DLK2, which is coincident with the behavior of those transfectants where we have detected an increase in osteogenic differentiation. Additionally, p38 MAPK phosphorylation increases in differentiated C3H10T1/2 cells with reduced DLK1 levels. CONCLUSIONS Our results suggest that DLK1 may inhibit osteogenesis, while DLK2 may promote it, by modulating NOTCH signaling and the phosphorylation of ERK1/2 and p38 MAPK pathways. Given the established inhibitory effect of DLK proteins on NOTCH signaling, these new insights could pave the way for developing future therapeutic strategies aimed at treating bone diseases.
Collapse
Affiliation(s)
- María-Milagros Rodríguez-Cano
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Pharmacy School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-Julia González-Gómez
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, ETSIAMB/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - Eva-María Monsalve
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Medical School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-José M Díaz-Guerra
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Medical School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - Moustapha Kassem
- Molecular Endocrinology Unit (KMEB), Department of Endocrinology and Metabolism, University Hospital of Odense and Danish Institute for Advanced Study, University of Southern Denmark, Odense, Denmark
| | - Jorge Laborda
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Pharmacy School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain
| | - María-Luisa Nueda
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Pharmacy School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain.
| | - Victoriano Baladrón
- Biochemistry and Molecular Biology Branch, Department of Inorganic, Organic Chemistry and Biochemistry, Medical School/IB-UCLM/Biomedicine Unit, University of Castilla-La Mancha/CSIC, Albacete, Spain.
| |
Collapse
|
5
|
Mitsis A, Khattab E, Christodoulou E, Myrianthopoulos K, Myrianthefs M, Tzikas S, Ziakas A, Fragakis N, Kassimis G. From Cells to Plaques: The Molecular Pathways of Coronary Artery Calcification and Disease. J Clin Med 2024; 13:6352. [PMID: 39518492 PMCID: PMC11545949 DOI: 10.3390/jcm13216352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Coronary artery calcification (CAC) is a hallmark of atherosclerosis and a critical factor in the development and progression of coronary artery disease (CAD). This review aims to address the complex pathophysiological mechanisms underlying CAC and its relationship with CAD. We examine the cellular and molecular processes that drive the formation of calcified plaques, highlighting the roles of inflammation, lipid accumulation, and smooth muscle cell proliferation. Additionally, we explore the genetic and environmental factors that contribute to the heterogeneity in CAC and CAD presentation among individuals. Understanding these intricate mechanisms is essential for developing targeted therapeutic strategies and improving diagnostic accuracy. By integrating current research findings, this review provides a comprehensive overview of the pathways linking CAC to CAD, offering insights into potential interventions to mitigate the burden of these interrelated conditions.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Elina Khattab
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Evi Christodoulou
- Cardiology Department, Limassol General Hospital, State Health Services Organization, Limassol 3304, Cyprus;
| | - Kimon Myrianthopoulos
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Michael Myrianthefs
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Stergios Tzikas
- Third Department of Cardiology, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| |
Collapse
|
6
|
Tóth A, Balogh E, Jeney V. In Vitro Models of Cardiovascular Calcification. Biomedicines 2024; 12:2155. [PMID: 39335668 PMCID: PMC11429067 DOI: 10.3390/biomedicines12092155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular calcification, characterized by hydroxyapatite deposition in the arterial wall and heart valves, is associated with high cardiovascular morbidity and mortality. Cardiovascular calcification is a hallmark of aging but is frequently seen in association with chronic diseases, such as chronic kidney disease (CKD), diabetes, dyslipidemia, and hypertension in the younger population as well. Currently, there is no therapeutic approach to prevent or cure cardiovascular calcification. The pathophysiology of cardiovascular calcification is highly complex and involves osteogenic differentiation of various cell types of the cardiovascular system, such as vascular smooth muscle cells and valve interstitial cells. In vitro cellular and ex vivo tissue culture models are simple and useful tools in cardiovascular calcification research. These models contributed largely to the discoveries of the numerous calcification inducers, inhibitors, and molecular mechanisms. In this review, we provide an overview of the in vitro cell culture and the ex vivo tissue culture models applied in the research of cardiovascular calcification.
Collapse
Affiliation(s)
- Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
7
|
Huang A, Xu T, Lu X, Ma L, Ma H, Yu Y, Yao L. Shh-Gli2-Runx2 inhibits vascular calcification. Nephrol Dial Transplant 2024; 39:305-316. [PMID: 37451818 DOI: 10.1093/ndt/gfad165] [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: 05/12/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND In patients with chronic kidney disease (CKD), vascular calcification (VC) is common and is associated with a higher risk of all-cause mortality. Shh, one ligand for Hedgehog (Hh) signaling, participates in osteogenesis and several cardiovascular diseases. However, it remains unclear whether Shh is implicated in the development of VC. METHODS Inorganic phosphorus 2.6 mM was used to induce vascular smooth muscle cells (VSMCs) calcification. Mice were fed with adenine diet supplement with 1.2% phosphorus to induce VC. RESULTS Shh was decreased in VSMCs exposed to inorganic phosphorus, calcified arteries in mice fed with an adenine diet, as well as radial arteries from patients with CKD presenting VC. Overexpression of Shh inhibited VSMCs ostosteoblastic differentiation and calcification, whereas its silencing accelerated these processes. Likewise, mice treated with smoothened agonist (SAG; Hh signaling agonist) showed alleviated VC, and mice treated with cyclopamine (CPN; Hh signaling antagonist) exhibited severe VC. Additionally, overexpression of Gli2 significantly reversed the pro-calcification effect of Shh silencing on VSMCs, suggesting that Shh inhibited VC via Gli2. Mechanistically, Gli2 interacted with Runx2 and promoted its ubiquitin proteasomal degradation, therefore protecting against VC. Of interest, the pro-degradation effect of Gli2 on Runx2 was independent of Smurf1 and Cullin4B. CONCLUSIONS Our study provided deeper insight to the pathogenesis of VC, and Shh might be a novel potential target for VC treatment.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ling Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Haiying Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yanqiu Yu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Shenyang Engineering Technology R&D Center of Cell Therapy Co. Ltd, Shenyang, China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Kang JH, Kawano T, Murata M, Toita R. Vascular calcification and cellular signaling pathways as potential therapeutic targets. Life Sci 2024; 336:122309. [PMID: 38042282 DOI: 10.1016/j.lfs.2023.122309] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Increased vascular calcification (VC) is observed in patients with cardiovascular diseases such as atherosclerosis, diabetes, and chronic kidney disease. VC is divided into three types according to its location: intimal, medial, and valvular. Various cellular signaling pathways are associated with VC, including the Wnt, mitogen-activated protein kinase, phosphatidylinositol-3 kinase/Akt, cyclic nucleotide-dependent protein kinase, protein kinase C, calcium/calmodulin-dependent kinase II, adenosine monophosphate-activated protein kinase/mammalian target of rapamycin, Ras homologous GTPase, apoptosis, Notch, and cytokine signaling pathways. In this review, we discuss the literature concerning the key cellular signaling pathways associated with VC and their role as potential therapeutic targets. Inhibitors to these pathways represent good candidates for use as potential therapeutic agents for the prevention and treatment of VC.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
9
|
Vazirian F, Sadeghi M, Kelesidis T, Budoff MJ, Zandi Z, Samadi S, Mohammadpour AH. Predictive value of lipoprotein(a) in coronary artery calcification among asymptomatic cardiovascular disease subjects: A systematic review and meta-analysis. Nutr Metab Cardiovasc Dis 2023; 33:2055-2066. [PMID: 37567791 PMCID: PMC11073574 DOI: 10.1016/j.numecd.2023.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/20/2023] [Accepted: 07/11/2023] [Indexed: 08/13/2023]
Abstract
AIMS Studies have indicated inconsistent results regarding the association between plasma levels of Lipoprotein(a) [Lp(a)] and coronary artery calcification (CAC). We performed a systematic review and meta-analysis to investigate the association between elevated levels of Lp(a) and risk of CAC in populations free of cardiovascular disease (CVD) symptoms. DATA SYNTHESIS PubMed, Web of Science, Embase, and Scopus were searched up to July 2022 and the methodological quality was assessed using Newcastle-Ottawa Scale (NOS) scale. Random-effects meta-analysis was used to estimate pooled odds ratio (OR) and 95% confidence interval. Out of 298 studies, data from 8 cross-sectional (n = 18,668) and 4 cohort (n = 15,355) studies were used in meta-analysis. Cohort studies demonstrated a positive significant association between Lp(a) and CAC, so that individuals with Lp(a)≥30-50 exposed to about 60% risk of CAC incidence compared to those with lower Lp(a) concentrations in asymptomatic CVD subjects (OR, 1.58; 95% CI, 1.38-1.80; l2, 0.0%; P, 0.483); Subgroup analysis showed that a cut-off level for Lp(a) measurement could not statistically affect the association, but race significantly affected the relationship between Lp(a) and CAC (OR,1.60; 95% CI, 1.41-1.81). Analyses also revealed that both men and women with higher Lp(a) concentrations are at the same risk for increased CAC. CONCLUSIONS Blood Lp(a) level was significantly associated with CAC incidence in asymptomatic populations with CVD, indicating that measuring Lp(a) may be a useful biomarker for diagnosing subclinical atherosclerosis in individuals at higher risk of CAC score. PROSPERO REGISTRATION NUMBER CRD42022350297.
Collapse
Affiliation(s)
- Fatemeh Vazirian
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Sadeghi
- Department of Epidemiology, Faculty of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew J Budoff
- Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Zahra Zandi
- Department of Cardiovascular Disease, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Samadi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amir Hooshang Mohammadpour
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Wen T, Duan Y, Gao D, Zhang X, Zhang X, Liang L, Yang Z, Zhang P, Zhang J, Sun J, Feng Y, Zheng Q, Han H, Yan X. miR-342-5p promotes vascular smooth muscle cell phenotypic transition through a negative-feedback regulation of Notch signaling via targeting FOXO3. Life Sci 2023:121828. [PMID: 37270171 DOI: 10.1016/j.lfs.2023.121828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/05/2023]
Abstract
AIM Under various pathological conditions such as cancer, vascular smooth muscle cells (vSMCs) transit their contractile phenotype into phenotype(s) characterized by proliferation and secretion, a process called vSMC phenotypic transition (vSMC-PT). Notch signaling regulates vSMC development and vSMC-PT. This study aims to elucidate how the Notch signal is regulated. MAIN METHODS Gene-modified mice with a SM22α-CreERT2 transgene were generated to activate/block Notch signaling in vSMCs. Primary vSMCs and MOVAS cells were cultured in vitro. RNA-seq, qRT-PCR and Western blotting were used to evaluated gene expression level. EdU incorporation, Transwell and collagen gel contraction assays were conducted to determine the proliferation, migration and contraction, respectively. KEY FINDINGS Notch activation upregulated, while Notch blockade downregulated, miR-342-5p and its host gene Evl in vSMCs. However, miR-342-5p overexpression promoted vSMC-PT as shown by altered gene expression profile, increased migration and proliferation, and decreased contraction, while miR-342-5p blockade exhibited the opposite effects. Moreover, miR-342-5p overexpression significantly suppressed Notch signaling, and Notch activation partially abolished miR-342-5p-induced vSMC-PT. Mechanically, miR-342-5p directly targeted FOXO3, and FOXO3 overexpression rescued miR-342-5p-induced Notch repression and vSMC-PT. In a simulated tumor microenvironment, miR-342-5p was upregulated by tumor cell-derived conditional medium (TCM), and miR-342-5p blockade abrogated TCM-induced vSMC-PT. Meanwhile, conditional medium from miR-342-5p-overexpressing vSMCs significantly enhanced tumor cell proliferation, while miR-342-5p blockade had the opposite effects. Consistently, in a co-inoculation tumor model, miR-342-5p blockade in vSMCs significantly delayed tumor growth. SIGNIFICANCE miR-342-5p promotes vSMC-PT through a negative-feedback regulation of Notch signaling via downregulating FOXO3, which could be a potential target for cancer therapy.
Collapse
Affiliation(s)
- Ting Wen
- Faculty of Life Sciences, Northwest University, Xi'an 710069, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Yanyan Duan
- Faculty of Life Sciences, Northwest University, Xi'an 710069, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Dan Gao
- Faculty of Life Sciences, Northwest University, Xi'an 710069, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Xinxin Zhang
- College of Pulmonary and Critical Care Medicine, The 8th Medical Centre of Chinese PLA General Hospital, Beijing 100091, China
| | - Xiaoyan Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Liang Liang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Ziyan Yang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Peiran Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Jiayulin Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Jiaxing Sun
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yixuan Feng
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, Shenzhen 518020, China.
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| | - Xianchun Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
11
|
Huang A, Li L, Liu X, Lian Q, Guo G, Xu T, Lu X, Ma L, Ma H, Yu Y, Yao L. Hedgehog signaling is a potential therapeutic target for vascular calcification. Gene 2023; 872:147457. [PMID: 37141952 DOI: 10.1016/j.gene.2023.147457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/07/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) suffered from vascular calcification (VC), one major contributor for their increased mortality rate. Hedgehog (Hh) signaling plays a crucial role in physiological bone mineralization and is associated with several cardiovascular diseases. However, the molecular changes underlying VC is ill defined and it remains unclear whether Hh signaling intervention affects VC. METHODS We constructed human primary vascular smooth muscle cell (VSMC) calcification model and performed RNA sequencing. Alizarin red staining and calcium content assay were conducted to identify the occurrence of VC. Three different R packages were applied to determine differentially expressed genes (DEGs). Enrichment analysis and protein-protein interaction (PPI) network analysis were carried out to explore the biological roles of DEGs. qRT-PCR assay was then applied to validate the expression of key genes. By using Connectivity Map (CMAP) analysis, several small molecular drugs targeting these key genes were obtained, including SAG (Hedgehog signaling activator) and cyclopamine (CPN) (Hedgehog signaling inhibitor), which were subsequently used to treat VSMC. RESULTS Obvious Alizarin red staining and increased calcium content identified the occurrence of VC. By integrating results from three R packages, we totally obtained 166 DEGs (86 up-regulated and 80 down-regulated), which were significantly enriched in ossification, osteoblast differentiation, and Hh signaling. PPI network analysis identified 10 key genes and CMAP analysis predicted several small molecular drugs targeting these key genes including chlorphenamine, isoeugenol, CPN and phenazopyridine. Notably, our in vitro experiment showed that SAG markedly alleviated VSMC calcification, whereas CPN significantly exacerbated VC. CONCLUSIONS Our research provided deeper insight to the pathogenesis of VC and indicated that targeting Hh signaling pathway may represent a potential and effective therapy for VC.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Lu Li
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Xiaoxu Liu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Qiuting Lian
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Guangying Guo
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Xiaomei Lu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Ling Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Haiying Ma
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Yanqiu Yu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China; Shenyang Engineering Technology R&D Center of Cell Therapy Co. LTD., Shenyang 110169, China.
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
12
|
Osawa T, Koizumi T, Ito Y. Sustained Lumen Area by Paclitaxel-Coated Balloon Following Rotational Atherectomy for Napkin-Ring Left Main Trunk Ostial Lesion. Tex Heart Inst J 2023; 50:e227883. [PMID: 36947442 PMCID: PMC10178657 DOI: 10.14503/thij-22-7883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Late lumen enlargement after percutaneous coronary intervention (PCI) with drug-coated balloon has contributed to good clinical results. However, late lumen enlargement with drug-coated balloon following rotational atherectomy has not been well reported. This report describes a case of calcified napkin-ring ostial lesion at the left main trunk that showed a sustained lumen area after PCI with drug-coated balloon following rotational atherectomy. An 85-year-old female patient was admitted to the hospital with dyspnea. Echocardiography showed hypokinesis in the anteroseptal and inferior walls. Electrocardiograph-gated cardiac computed tomography showed a calcified ostial lesion in the left main trunk. Invasive angiography of the coronary artery showed severe stenosis in the left main trunk ostium. Percutaneous coronary intervention was performed with a drug-coated balloon after rotational atherectomy. The minimal lumen area measured by intravascular ultrasound grew mildly from 4.09 to 4.17 mm2 immediately after PCI. Follow-up angiography and intravascular ultrasound performed after 6 months showed that the minimal lumen area in the left main trunk ostium was further enlarged from 4.17 to 4.69 mm2. The presence of sustained lumen area after PCI with drug-coated balloon following rotational atherectomy for a napkin-ring left main trunk ostial lesion was confirmed. This case demonstrates sustained lumen area after drug-coated balloon following rotational atherectomy in the left main trunk ostium, improving the patient's chest symptom. Hence, drug-coated balloon after rotational atherectomy may be an option for complex stent sites, such as the left main trunk ostium in geriatric patients and sites with highly calcified lesions.
Collapse
Affiliation(s)
- Takumi Osawa
- Department of Cardiovascular Medicine, National Hospital Organization Mito Medical Center, Ibaraki, Japan
| | - Tomomi Koizumi
- Department of Cardiovascular Medicine, National Hospital Organization Mito Medical Center, Ibaraki, Japan
| | - Yuta Ito
- Department of Cardiovascular Medicine, National Hospital Organization Mito Medical Center, Ibaraki, Japan
| |
Collapse
|
13
|
Sibuea S, Ho JK, Pouton CW, Haynes JM. TGFβ3, dibutyryl cAMP and a notch inhibitor modulate phenotype late in stem cell-derived dopaminergic neuron maturation. Front Cell Dev Biol 2023; 11:1111705. [PMID: 36819101 PMCID: PMC9928866 DOI: 10.3389/fcell.2023.1111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/19/2023] [Indexed: 02/04/2023] Open
Abstract
The generation of midbrain dopaminergic neurons (mDAs) from pluripotent stem cells (hPSC) holds much promise for both disease modelling studies and as a cell therapy for Parkinson's disease (PD). Generally, dopaminergic neuron differentiation paradigms rely on inhibition of smad signalling for neural induction followed by hedgehog signalling and an elevation of β-catenin to drive dopaminergic differentiation. Post-patterning, differentiating dopaminergic neuron cultures are permitted time for maturation after which the success of these differentiation paradigms is usually defined by expression of tyrosine hydroxylase (TH), the rate limiting enzyme in the synthesis of dopamine. However, during maturation, culture media is often supplemented with additives to promote neuron survival and or promote cell differentiation. These additives include dibutyryl cyclic adenosine monophosphate (dbcAMP), transforming growth factor β3 (TGFβ3) and or the γ-secretase inhibitor (DAPT). While these factors are routinely added to cultures, their impact upon pluripotent stem cell-derived mDA phenotype is largely unclear. In this study, we differentiate pluripotent stem cells toward a dopaminergic phenotype and investigate how the omission of dbcAMP, TGFβ3 or DAPT, late in maturation, affects the regulation of multiple dopaminergic neuron phenotype markers. We now show that the removal of dbcAMP or TGFβ3 significantly and distinctly impacts multiple markers of the mDA phenotype (FOXA2, EN1, EN2, FOXA2, SOX6), while commonly increasing both MSX2 and NEUROD1 and reducing expression of both tyrosine hydroxylase and WNT5A. Removing DAPT significantly impacted MSX2, OTX2, EN1, and KCNJ6. In the absence of any stressful stimuli, we suggest that these culture additives should be viewed as mDA phenotype-modifying, rather than neuroprotective. We also suggest that their addition to cultures is likely to confound the interpretation of both transplantation and disease modelling studies.
Collapse
Affiliation(s)
- Shanti Sibuea
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia,National Agency of Drug and Food Control, Jakarta, Indonesia
| | - Joan K. Ho
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia
| | - Colin W. Pouton
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia
| | - John M. Haynes
- Stem Cell Biology Group, Monash Institute of Pharmaceutical Sciences Monash University, Parkville, VIC, Australia,*Correspondence: John M. Haynes,
| |
Collapse
|
14
|
Hashimoto D, Fujimoto K, Kim SW, Lee YS, Nakata M, Suzuki K, Wada Y, Asamura S, Yamada G. Emerging structural and pathological analyses on the erectile organ, corpus cavernous containing sinusoids. Reprod Med Biol 2023; 22:e12539. [PMID: 37663955 PMCID: PMC10472535 DOI: 10.1002/rmb2.12539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/08/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023] Open
Abstract
Background The corpus cavernosum (CC) containing sinusoids plays fundamental roles for erection. Analysis of pathological changes in the erectile system is studied by recent experimental systems. Various in vitro models utilizing genital mesenchymal-derived cells and explant culture systems are summarized. Methods 3D reconstruction of section images of murine CC was created. Ectopic chondrogenesis in aged mouse CC was shown by a gene expression study revealing the prominent expression of Sox9. Various experimental strategies utilizing mesenchyme-derived primary cells and tissue explants are introduced. Main Findings Possible roles of Sox9 in chondrogenesis and its regulation by several signals are suggested. The unique character of genital mesenchyme is shown by various analyses of external genitalia (ExG) derived cells and explant cultures. Such strategies are also applied to the analysis of erectile contraction/relaxation responses to many signals and aging process. Conclusion Erectile dysfunction (ED) is one of the essential topics for the modern aged society. More comprehensive studies are necessary to reveal the nature of the erectile system by combining multiple cell culture strategies.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Developmental Genetics, Institute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
- Department of Physiology and Regenerative Medicine, Faculty of MedicineKindai UniversityOsakaJapan
| | - Kota Fujimoto
- Department of Developmental Genetics, Institute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Sang Woon Kim
- Department of Urology, Urological Science InstituteYonsei University College of MedicineSeoulSouth Korea
| | - Yong Seung Lee
- Department of Urology, Urological Science InstituteYonsei University College of MedicineSeoulSouth Korea
| | - Masanori Nakata
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kentaro Suzuki
- Faculty of Life and Environmental SciencesUniversity of YamanashiYamanashiJapan
| | - Yoshitaka Wada
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced MedicineWakayama Medical UniversityWakayamaJapan
- Department of Plastic and Reconstructive Surgery, Graduate School of MedicineWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
15
|
Peng J, Liu MM, Liu HH, Xu RX, Zhu CG, Guo YL, Wu NQ, Dong Q, Cui CJ, Li JJ. Lipoprotein (a)-mediated vascular calcification: population-based and in vitro studies. Metabolism 2022; 127:154960. [PMID: 34954251 DOI: 10.1016/j.metabol.2021.154960] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/16/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lipoprotein (a) [Lp(a)] is a causal risk factor for cardiovascular diseases, while its role in vascular calcification has not been well-established. Here, we investigated an association of Lp(a) with vascular calcification using population-based and in vitro study designs. METHODS A total of 2806 patients who received coronary computed tomography were enrolled to assess the correlation of Lp(a) with the severity of coronary artery calcification (CAC). Human aortic smooth muscle cells (HASMCs) were used to explore mechanisms of Lp(a)-induced vascular calcification. RESULTS In the population study, Lp(a) was independently correlated with the presence and severity of CAC (all p < 0.05). In vitro study showed that cell calcific depositions and alkaline phosphatase (ALP) activity were increased and the expression of pro-calcific proteins, including bone morphogenetic protein-2 (BMP2) and osteopontin (OPN), were up-regulated by Lp(a) stimulation. Interestingly, Lp(a) activated Notch1 signaling, resulting in cell calcification, which was inhibited by the Notch1 signaling inhibitor, DAPT. Lp(a)-induced Notch1 activation up-regulated BMP2-Smad1/5/9 pathway. In contrast, Noggin, an inhibitor of BMP2-Smad1/5/9 pathway, significantly blocked Lp(a)-induced HASMC calcification. Notch1 activation also induced translocation of nuclear factor-κB (NF-κB) accompanied by OPN overexpression and elevated inflammatory cytokines production, while NF-κB silencing alleviated Lp(a)-induced vascular calcification. CONCLUSIONS Elevated Lp(a) concentrations are independently associated with the presence and severity of CAC and the impact of Lp(a) on vascular calcification is involved in the activation of Notch1-NF-κB and Notch1-BMP2-Smad1/5/9 pathways, thus implicating Lp(a) as a potential novel therapeutic target for vascular calcification.
Collapse
Affiliation(s)
- Jia Peng
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Ming-Ming Liu
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Hui-Hui Liu
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Rui-Xia Xu
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Cheng-Gang Zhu
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Yuan-Lin Guo
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Na-Qiong Wu
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Qian Dong
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China
| | - Chuan-Jue Cui
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China.
| | - Jian-Jun Li
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No 167 BeiLiShi Road, XiCheng District, Beijing 100037, China.
| |
Collapse
|
16
|
Deng Y, Li R, Wang H, Yang B, Shi P, Zhang Y, Yang Q, Li G, Bian L. Biomaterial-Mediated Presentation of Jagged-1 Mimetic Ligand Enhances Cellular Activation of Notch Signaling and Bone Regeneration. ACS NANO 2022; 16:1051-1062. [PMID: 34967609 DOI: 10.1021/acsnano.1c08728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The development from stem cells to adult tissues requires the delicate presentation of numerous crucial inductive cues and the activation of associated signaling pathways. The Notch signaling pathways triggered by ligands such as Jagged-1 have been demonstrated to be essential in various development processes especially in osteogenesis and ossification. However, few studies have capitalized on the osteoinductivity of the Jagged-1 mimetic ligands to enhance the osteogenesis and skeleton regeneration. In this study, we conjugate the porous hyaluronic acid hydrogels with a Jagged-1 mimetic peptide ligand (Jagged-1) and investigate the efficacy of such biomimetic functionalization to promote the mechanotransduction and osteogenesis of human mesenchymal stem cells by activating the Notch signaling pathway. Our findings indicate that the immobilized Jagged-1 mimetic ligand activates Notch signaling via the upregulation of NICD and downstream MSX2, leading to the enhanced mechanotransduction and osteogenesis of stem cells. We further demonstrate that the functionalization of the Jagged-1 ligand in the porous scaffold promotes angiogenesis, regulates macrophage recruitment and polarization, and enhances in situ regeneration of rat calvarial defects. Our findings provide valuable guidance to the design of development-inspired bioactive biomaterials for diverse biomedical applications.
Collapse
Affiliation(s)
- Yingrui Deng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P.R. China
| | - Rui Li
- Department of Mechanical and Aerospace Engineering, Tandon School of Engineering, New York University, Brooklyn, New York 11201, United States
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, P.R China
| | - Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong, P.R. China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin 300211, P.R. China
| | - Gang Li
- Department of Orthopaedics and Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, P.R China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P.R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P.R. China
| |
Collapse
|
17
|
Yang L, Dai R, Wu H, Cai Z, Xie N, Zhang X, Shen Y, Gong Z, Jia Y, Yu F, Zhao Y, Lin P, Ye C, Hu Y, Fu Y, Xu Q, Li Z, Kong W. Unspliced XBP1 Counteracts β-catenin to Inhibit Vascular Calcification. Circ Res 2021; 130:213-229. [PMID: 34870453 DOI: 10.1161/circresaha.121.319745] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Vascular calcification is a prevalent complication in chronic kidney disease and contributes to increased cardiovascular morbidity and mortality. XBP1 (X-box binding protein 1), existing as the unspliced (XBP1u) and spliced (XBP1s) forms, is a key component of the endoplasmic reticulum stress involved in vascular diseases. However, whether XBP1u participates in the development of vascular calcification remains unclear. Methods: We aim to investigate the role of XBP1u in vascular calcification.XBP1u protein levels were reduced in high phosphate (Pi)-induced calcified vascular smooth muscle cells (VSMCs), calcified aortas from mice with adenine diet-induced chronic renal failure (CRF) and calcified radial arteries from CRF patients. Results: Inhibition of XBP1u rather than XBP1s upregulated in the expression of the osteogenic markers runt-related transcription factor 2 (Runx2) and msh homeobox2 (Msx2), and exacerbated high Pi-induced VSMC calcification, as verified by calcium deposition and Alizarin red S staining. In contrast, XBP1u overexpression in high Pi-induced VSMCs significantly inhibited osteogenic differentiation and calcification. Consistently, SMC-specific XBP1 deficiency in mice markedly aggravated the adenine diet- and 5/6 nephrectomy-induced vascular calcification compared with that in the control littermates. Further interactome analysis revealed that XBP1u bound directly to β-catenin, a key regulator of vascular calcification, via aa 205-230 in its C-terminal degradation domain. XBP1u interacted with β-catenin to promote its ubiquitin-proteasomal degradation and thus inhibited β-catenin/T-cell factor (TCF)-mediated Runx2 and Msx2 transcription. Knockdown of β-catenin abolished the effect of XBP1u deficiency on VSMC calcification, suggesting a β-catenin-mediated mechanism. Moreover, the degradation of β-catenin promoted by XBP1u was independent of glycogen synthase kinase 3β (GSK-3β)-involved destruction complex. Conclusions: Our study identified XBP1u as a novel endogenous inhibitor of vascular calcification by counteracting β-catenin and promoting its ubiquitin-proteasomal degradation, which represents a new regulatory pathway of β-catenin and a promising target for vascular calcification treatment.
Collapse
Affiliation(s)
- Liu Yang
- Physiology and Pathophysiology, Peking University, CHINA
| | - Rongbo Dai
- Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, CHINA
| | - Hao Wu
- Physiology and Pathophysiology, Peking University, CHINA
| | - Zeyu Cai
- Physiology and Pathophysiology, Peking University, CHINA
| | - Nan Xie
- Physiology and Pathophysiology, Peking University, CHINA
| | - Xu Zhang
- Physiology and Pathophysiology, Peking University, CHINA
| | - Yicong Shen
- Physiology and Pathophysiology, Peking University, CHINA
| | - Ze Gong
- Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, CHINA
| | - Yiting Jia
- Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, CHINA
| | - Fang Yu
- School of Basic Medical Sciences, Peking University
| | - Ying Zhao
- Biochemistry and Molecular Biology, Peking University, CHINA
| | - Pinglan Lin
- Nephrology, Shanghai University of Traditional Chinese Medicine, CHINA
| | - Chaoyang Ye
- Nephrology, Shanghai University of Traditional Chinese Medicine, CHINA
| | - Yanhua Hu
- Cardiology, Zhejiang University, CHINA
| | - Yi Fu
- Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, CHINA
| | - Qingbo Xu
- Cardiology, Zhejing University, CHINA
| | - Zhiqing Li
- Physiology and Pathophysiology, Peking University, CHINA
| | - Wei Kong
- Physiology and Pathophysiology, Peking University, CHINA
| |
Collapse
|
18
|
Regulation of MDM2 E3 ligase-dependent vascular calcification by MSX1/2. Exp Mol Med 2021; 53:1781-1791. [PMID: 34845330 PMCID: PMC8639964 DOI: 10.1038/s12276-021-00708-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/24/2021] [Accepted: 10/06/2021] [Indexed: 11/27/2022] Open
Abstract
Vascular calcification increases morbidity and mortality in patients with cardiovascular and renal diseases. Previously, we reported that histone deacetylase 1 prevents vascular calcification, whereas its E3 ligase, mouse double minute 2 homolog (MDM2), induces vascular calcification. In the present study, we identified the upstream regulator of MDM2. By utilizing cellular models and transgenic mice, we confirmed that E3 ligase activity is required for vascular calcification. By promoter analysis, we found that both msh homeobox 1 (Msx1) and msh homeobox 2 (Msx2) bound to the MDM2 promoter region, which resulted in transcriptional activation of MDM2. The expression levels of both Msx1 and Msx2 were increased in mouse models of vascular calcification and in calcified human coronary arteries. Msx1 and Msx2 potentiated vascular calcification in cellular and mouse models in an MDM2-dependent manner. Our results establish a novel role for MSX1/MSX2 in the transcriptional activation of MDM2 and the resultant increase in MDM2 E3 ligase activity during vascular calcification. The identification of a signaling pathway involved in triggering vascular calcification, the deposition of calcium phosphate crystals in blood vessels, could inform new therapeutic interventions for related cardiovascular complications. Vascular calcification causes significant complications in patients with metabolic syndrome, renal failure, or cardiovascular disease. In their previous work, Hyun Kook and Duk-Hwa Kwon at Chonnam National University Medical School, Jeollanamdo, Republic of Korea, and coworkers demonstrated that the E3 ligase activity of a protein called MDM2 induces calcification. Now, following further mouse trials, the team have identified an upstream signaling pathway involving several development proteins such as MSX1 and MSX2 which activate MDM2. The activation of this signaling axis leads to the degradation of a key protein that would otherwise prevent calcification. The results may provide a platform for novel therapies targeting the condition.
Collapse
|
19
|
Wang L, Chennupati R, Jin YJ, Li R, Wang S, Günther S, Offermanns S. YAP/TAZ Are Required to Suppress Osteogenic Differentiation of Vascular Smooth Muscle Cells. iScience 2020; 23:101860. [PMID: 33319178 PMCID: PMC7726335 DOI: 10.1016/j.isci.2020.101860] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/10/2020] [Accepted: 11/20/2020] [Indexed: 12/22/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) represent the prevailing cell type of arterial vessels and are essential for blood vessel structure and homeostasis. They have substantial potential for phenotypic plasticity when exposed to various stimuli in their local microenvironment. How VSMCs maintain their differentiated contractile phenotype is still poorly understood. Here we demonstrate that the Hippo pathway effectors YAP and TAZ play a critical role in maintaining the differentiated contractile phenotype of VSMCs. In the absence of YAP/TAZ, VSMCs lose their differentiated phenotype and undergo osteogenic differentiation, which results in vascular calcification. Osteogenic transdifferentiation was accompanied by the upregulation of Wnt target genes. The absence of YAP/TAZ in VSMCs led to Disheveled 3 (DVL3) nuclear translocation and upregulation of osteogenesis-associated genes independent of canonical Wnt/β-catenin signaling activation. Our data indicate that cytoplasmic YAP/TAZ interact with DVL3 to avoid its nuclear translocation and osteogenic differentiation, thereby maintaining the differentiated phenotype of VSMCs. YAP/TAZ play an important role in maintaining vascular SMCs contractile phenotype Loss of YAP/TAZ in vSMCs leads to reduced expression of smooth muscle marker genes Loss of YAP/TAZ in vSMCs results in reduced artery contractility Deficiency of YAP/TAZ in vSMCs leads to osteogenic transdifferentiation
Collapse
Affiliation(s)
- Lei Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Ramesh Chennupati
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Young-June Jin
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Rui Li
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - ShengPeng Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an 710061, China
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Center for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhine-Main, 13347 Berlin, Germany
| |
Collapse
|
20
|
Pitrez PR, Estronca L, Monteiro LM, Colell G, Vazão H, Santinha D, Harhouri K, Thornton D, Navarro C, Egesipe AL, Carvalho T, Dos Santos RL, Lévy N, Smith JC, de Magalhães JP, Ori A, Bernardo A, De Sandre-Giovannoli A, Nissan X, Rosell A, Ferreira L. Vulnerability of progeroid smooth muscle cells to biomechanical forces is mediated by MMP13. Nat Commun 2020; 11:4110. [PMID: 32807790 PMCID: PMC7431909 DOI: 10.1038/s41467-020-17901-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 07/14/2020] [Indexed: 12/26/2022] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disease in children that leads to early death. Smooth muscle cells (SMCs) are the most affected cells in HGPS individuals, although the reason for such vulnerability remains poorly understood. In this work, we develop a microfluidic chip formed by HGPS-SMCs generated from induced pluripotent stem cells (iPSCs), to study their vulnerability to flow shear stress. HGPS-iPSC SMCs cultured under arterial flow conditions detach from the chip after a few days of culture; this process is mediated by the upregulation of metalloprotease 13 (MMP13). Importantly, double-mutant LmnaG609G/G609GMmp13-/- mice or LmnaG609G/G609GMmp13+/+ mice treated with a MMP inhibitor show lower SMC loss in the aortic arch than controls. MMP13 upregulation appears to be mediated, at least in part, by the upregulation of glycocalyx. Our HGPS-SMCs chip represents a platform for developing treatments for HGPS individuals that may complement previous pre-clinical and clinical treatments.
Collapse
Affiliation(s)
- Patricia R Pitrez
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Luís Estronca
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Luís Miguel Monteiro
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Guillem Colell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Helena Vazão
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Deolinda Santinha
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Claire Navarro
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- Progelife, Marseille, France
| | - Anne-Laure Egesipe
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| | - Tânia Carvalho
- IMM, Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | | | - Nicolas Lévy
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- Molecular Genetics Laboratory, Department of Medical Genetics, La Timone Children's Hospital, Marseille, France
| | - James C Smith
- Developmental Biology Laboratory, Francis Crick Institute, London, NW1 1AT, UK
| | - João Pedro de Magalhães
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute, 07745, Jena, Germany
| | - Andreia Bernardo
- Developmental Biology Laboratory, Francis Crick Institute, London, NW1 1AT, UK
| | - Annachiara De Sandre-Giovannoli
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- Molecular Genetics Laboratory, Department of Medical Genetics, La Timone Children's Hospital, Marseille, France
- CRB Assistance Publique des Hôpitaux de Marseille (CRB AP-HM, TAC), Marseille, France
| | - Xavier Nissan
- CECS, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, Evry Cedex, France
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
21
|
Manokawinchoke J, Sumrejkanchanakij P, Boonprakong L, Pavasant P, Egusa H, Osathanon T. NOTCH2 participates in Jagged1-induced osteogenic differentiation in human periodontal ligament cells. Sci Rep 2020; 10:13329. [PMID: 32770090 PMCID: PMC7414879 DOI: 10.1038/s41598-020-70277-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Jagged1 activates Notch signaling and subsequently promotes osteogenic differentiation in human periodontal ligament cells (hPDLs). The present study investigated the participation of the Notch receptor, NOTCH2, in the Jagged1-induced osteogenic differentiation in hPDLs. NOTCH2 and NOTCH4 mRNA expression levels increased during hPDL osteogenic differentiation. However, the endogenous NOTCH2 expression levels were markedly higher compared with NOTCH4. NOTCH2 expression knockdown using shRNA in hPDLs did not dramatically alter their proliferation or osteogenic differentiation compared with the shRNA control. After seeding on Jagged1-immobilized surfaces and maintaining the hPDLs in osteogenic medium, HES1 and HEY1 mRNA levels were markedly reduced in the shNOTCH2-transduced cells compared with the shControl group. Further, shNOTCH2-transduced cells exhibited less alkaline phosphatase enzymatic activity and in vitro mineralization than the shControl cells when exposed to Jagged1. MSX2 and COL1A1 mRNA expression after Jagged1 activation were reduced in shNOTCH2-transduced cells. Endogenous Notch signaling inhibition using a γ-secretase inhibitor (DAPT) attenuated mineralization in hPDLs. DAPT treatment significantly promoted TWIST1, but decreased ALP, mRNA expression, compared with the control. In conclusion, Notch signaling is involved in hPDL osteogenic differentiation. Moreover, NOTCH2 participates in the mechanism by which Jagged1 induced osteogenic differentiation in hPDLs.
Collapse
Affiliation(s)
- Jeeranan Manokawinchoke
- Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Piyamas Sumrejkanchanakij
- Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Lawan Boonprakong
- Oral Biology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Prasit Pavasant
- Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan
| | - Thanaphum Osathanon
- Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand. .,Oral Biology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand. .,Genomics and Precision Dentistry Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
22
|
Lin X, Li F, Xu F, Cui RR, Xiong D, Zhong JY, Zhu T, Shan SK, Wu F, Xie XB, Liao XB, Yuan LQ. Aberration methylation of miR-34b was involved in regulating vascular calcification by targeting Notch1. Aging (Albany NY) 2020; 11:3182-3197. [PMID: 31129659 PMCID: PMC6555467 DOI: 10.18632/aging.101973] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/12/2019] [Indexed: 12/21/2022]
Abstract
Vascular calcification is one of the most important factors for cardiovascular and all-cause mortality in patients with end-stage renal diseases (ESRD). The current study was aimed to investigate the function and mechanisms of miR-34b on the calcification of vascular smooth muscle cells (VSMCs) both in vitro and in vivo. We found that the expression of miR-34b was significantly suppressed in VSMCs with high inorganic phosphate (Pi) treatment, as well as mouse arteries derived from 5/6 nephrectomy with a high-phosphate diet (0.9% Pi, 5/6 NTP) and human renal arteries from uraemia patients. Overexpression of miR-34b alleviated calcification of VSMCs, while VSMCs calcification was enhanced by inhibiting the expression of miR-34b. Bisulphite sequencing PCR (BSP) uncovered that CpG sites upstream of miR-34b DNA were hypermethylated in calcified VSMCs and calcified arteries due to 5/6 NTP, as well as calcified renal arterial tissues from uraemia patients. Meantime, increased DNA methyltransferase 3a (DNMT3a) resulted in the hypermethylation of miR-34b in VSMCs, while 5-aza-2′-deoxycytidine (5-aza) reduced the methylation rate of miR-34b and restored the expression of miR-34b in VSMCs. When DNMT3a was knocked down using DNMT3a siRNA, the effect of 3.5 mM of Pi on calcification of VSMCs was abrogated. In addition, Notch1 was validated as the functional target of miR-34b and involved in the process of calcification of VSMCs. Taken together, our data showed a specific role for miR-34b in regulating calcification of VSMCs both in vitro and in vivo, which was regulated by upstream DNA methylation of miR-34b and modulated by the downstream target gene expression, Notch1. These results suggested that modulation of miR-34b may offer new insight into a novel therapeutic approach for vascular calcification.
Collapse
Affiliation(s)
- Xiao Lin
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Fuxingzi Li
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Feng Xu
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Rong-Rong Cui
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Xiong
- Department of Endocrinology, Central Hospital of Yiyang, Yiyang, Hunan, People's Republic of China
| | - Jia-Yu Zhong
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Ting Zhu
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Endocrinology, Central Hospital of Yiyang, Yiyang, Hunan, People's Republic of China
| | - Su-Kang Shan
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Feng Wu
- Department of Pathology, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xu-Biao Xie
- Center of Organ Transplantation, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Ling-Qing Yuan
- Department of Endocrinology and Metabolism, National Clinical Research Center for Metabolic Diseases, The Second Xiang-Ya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
23
|
Liu N, Zhou M, Zhang Q, Yong L, Zhang T, Tian T, Ma Q, Lin S, Zhu B, Cai X. Effect of substrate stiffness on proliferation and differentiation of periodontal ligament stem cells. Cell Prolif 2018; 51:e12478. [PMID: 30039894 DOI: 10.1111/cpr.12478] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/07/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The aim of this study was to understand the effect of substrate stiffness (a mechanical factor of the extracellular matrix) on periodontal ligament stem cells (PDLSCs) and its underlying mechanism. MATERIALS AND METHODS Elastic substrates were fabricated by mixing 2 components, a base and curing agent in proportions of 10:1, 20:1, 30:1 or 40:1. PDLSC morphology was observed using scanning electron microscopy (SEM). Cell proliferation and differentiation were assessed after PDLSCs was cultured on various elastic substrates. Data were analysed using one-way ANOVA. RESULTS SEM revealed variations in the morphology of PDLSCs cultured on elastic substrates. PDLSC proliferation increased with substrate stiffness (P < .05). Osteogenic differentiation of PDLSCs was higher on stiff substrates. Notch pathway markers were up-regulated in PDLSCs cultured on stiff substrates. CONCLUSIONS Results suggested that the osteogenic differentiation of PDLSCs might be promoted by culturing them in a stiffness-dependent manner, which regulates the Notch pathway. This might provide a new method of enhancing osteogenesis in PDLSCs.
Collapse
Affiliation(s)
- Nanxin Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mi Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Li Yong
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Taoran Tian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quanquan Ma
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shiyu Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bofeng Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Department of Forensic Genetics, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Wang J, Zhou JJ, Robertson GR, Lee VW. Vitamin D in Vascular Calcification: A Double-Edged Sword? Nutrients 2018; 10:nu10050652. [PMID: 29786640 PMCID: PMC5986531 DOI: 10.3390/nu10050652] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 01/13/2023] Open
Abstract
Vascular calcification (VC) as a manifestation of perturbed mineral balance, is associated with aging, diabetes and kidney dysfunction, as well as poorer patient outcomes. Due to the current limited understanding of the pathophysiology of vascular calcification, the development of effective preventative and therapeutic strategies remains a significant clinical challenge. Recent evidence suggests that traditional risk factors for cardiovascular disease, such as left ventricular hypertrophy and dyslipidaemia, fail to account for clinical observations of vascular calcification. Therefore, more complex underlying processes involving physiochemical changes to mineral balance, vascular remodelling and perturbed hormonal responses such as parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF-23) are likely to contribute to VC. In particular, VC resulting from modifications to calcium, phosphate and vitamin D homeostasis has been recently elucidated. Notably, deregulation of vitamin D metabolism, dietary calcium intake and renal mineral handling are associated with imbalances in systemic calcium and phosphate levels and endothelial cell dysfunction, which can modulate both bone and soft tissue calcification. This review addresses the current understanding of VC pathophysiology, with a focus on the pathogenic role of vitamin D that has provided new insights into the mechanisms of VC.
Collapse
Affiliation(s)
- Jeffrey Wang
- Centre for Transplantation and Renal Research, Westmead Institute of Medical Research, Westmead, NSW 2145, Australia.
| | - Jimmy J Zhou
- Centre for Transplantation and Renal Research, Westmead Institute of Medical Research, Westmead, NSW 2145, Australia.
- Centre for Kidney Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia.
| | | | - Vincent W Lee
- Centre for Transplantation and Renal Research, Westmead Institute of Medical Research, Westmead, NSW 2145, Australia.
| |
Collapse
|
25
|
Kwon DH, Kim YK, Kook H. New Aspects of Vascular Calcification: Histone Deacetylases and Beyond. J Korean Med Sci 2017; 32:1738-1748. [PMID: 28960024 PMCID: PMC5639052 DOI: 10.3346/jkms.2017.32.11.1738] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/19/2017] [Indexed: 11/20/2022] Open
Abstract
Vascular calcification is a pathologic phenomenon in which calcium phosphate is ectopically deposited in the arteries. Previously, calcification was considered to be a passive process in response to metabolic diseases, vascular or valvular diseases, or even aging. However, now calcification is recognized as a highly-regulated consequence, like bone formation, and many clinical trials have been carried out to elucidate the correlation between vascular calcification and cardiovascular events and mortality. As a result, vascular calcification has been implicated as an independent risk factor in cardiovascular diseases. Many molecules are now known to be actively associated with this process. Recently, our laboratory found that posttranslational modification of histone deacetylase (HDAC) 1 is actively involved in the development of vascular calcification. In addition, we found that modulation of the activity of HDAC as well as its protein stability by MDM2, an HDAC1-E3 ligase, may be a therapeutic target in vascular calcification. In the present review, we overview the pathomechanism of vascular calcification and the involvement of posttranslational modification of epigenetic regulators.
Collapse
Affiliation(s)
- Duk Hwa Kwon
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea
| | - Young Kook Kim
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea
- Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Korea
- Basic Research Laboratory for Cardiac Remodeling, Chonnam National University Medical School, Gwangju, Korea.
| |
Collapse
|
26
|
Cao J, Wei Y, Lian J, Yang L, Zhang X, Xie J, Liu Q, Luo J, He B, Tang M. Notch signaling pathway promotes osteogenic differentiation of mesenchymal stem cells by enhancing BMP9/Smad signaling. Int J Mol Med 2017; 40:378-388. [PMID: 28656211 PMCID: PMC5504972 DOI: 10.3892/ijmm.2017.3037] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 04/26/2017] [Indexed: 12/19/2022] Open
Abstract
Notch is an important pathway in that it regulates cell-to-cell signal transduction, which plays an essential role in skeletal remodeling. Bone morphogenetic protein (BMP)9 has been regarded as one of the most efficient BMPs by which to induce osteogenic differentiation in mesenchymal stem cells (MSCs). Understanding the interaction between Notch and BMP9 signaling is a critical issue for optimizing the application of MSCs and BMPs in bone tissue engineering. In the present study, we investigated the role of Notch signaling in the BMP9‑induced osteogenic differentiation of MSCs. Our data demonstrated that Notch signaling obviously enhanced BMP9‑induced osteogenic differentiation in MSCs in vitro and in vivo. Notch signaling augmented the activity of BMP9‑induced BMP/Smad signaling and increased the gene expression of essential osteogenic factors induced by BMP9 in MSCs, such as runt‑related transcription factor 2 (Runx2), type I collagen (Colla1) and inhibitor of differentiation (Id)1. We also found that Notch signaling promoted the expression of activin‑like kinase 2 (ALK2) induced by BMP9, and the inhibitory effect of dnALK2 on BMP9‑induced osteogenic differentiation was rescued by constitutive overexpression of Delta‑like 1 (DLL1). Notch signaling also exhibited an apparent effect on the proliferation of mouse embryo fibroblasts (MEFs) during BMP9‑induced osteogenic differentiation. These results indicate that Notch plays a significant role in mediating BMP9‑induced osteogenic differentiation in MSCs, which may be partly regulated by upregulation of the expression of ALK2.
Collapse
Affiliation(s)
- Junjie Cao
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yalin Wei
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jing Lian
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lunyun Yang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaoyan Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jiaying Xie
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qiang Liu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Baicheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Min Tang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
27
|
Gong C, Li L, Qin C, Wu W, Liu Q, Li Y, Gan L, Ou S. The Involvement of Notch1-RBP-Jk/Msx2 Signaling Pathway in Aortic Calcification of Diabetic Nephropathy Rats. J Diabetes Res 2017; 2017:8968523. [PMID: 29464183 PMCID: PMC5804331 DOI: 10.1155/2017/8968523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/27/2017] [Accepted: 10/26/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND This study explored the changes in expression of vascular smooth muscle cell (VSMC) markers and osteogenic markers, as well as the involvement of Notch1-RBP-Jk/Msx2 pathway in a rat model of diabetic nephropathy (DN) with vascular calcification. METHODS A rat model of DN with concomitant vascular calcification was created by intraperitoneal injection of streptozotocin followed by administration of vitamin D3 and nicotine. Biochemical analysis and histological examination of aortic tissue were performed. VSMC markers and osteogenic markers as well as target molecules in Notch1-RBP-Jk/Msx2 were determined by quantitative real-time polymerase chain reaction and immunohistochemical analysis. RESULTS Serum calcium and phosphorus levels were significantly increased in model rats as compared to that in normal controls. Diabetic rats with vascular calcification exhibited mineral deposits in aortic intima-media accompanied by decreased expression of VSMC markers and increased expression of osteogenic markers. Notch1, RBP-Jk, Msx2, Jagged1, and N1-ICD were barely expressed in the aortic wall of normal rats. In contrast, these were significantly increased in the model group at all time points (8, 12, and 16 weeks), as compared to that in the normal rats. CONCLUSION Activation of the Notch1-RBP-Jk/Msx2 signaling pathway may be involved in the development and progression of vascular calcification in DN.
Collapse
Affiliation(s)
- Caipan Gong
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Li Li
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunmei Qin
- Department of Nephrology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China
| | - Weihua Wu
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qi Liu
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ying Li
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Linwang Gan
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Santao Ou
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
28
|
Nakano T, Fukuda D, Koga JI, Aikawa M. Delta-Like Ligand 4-Notch Signaling in Macrophage Activation. Arterioscler Thromb Vasc Biol 2016; 36:2038-47. [PMID: 27562914 PMCID: PMC5033717 DOI: 10.1161/atvbaha.116.306926] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022]
Abstract
The Notch signaling pathway regulates the development of various cell types and organs, and also contributes to disease mechanisms in adults. Accumulating evidence suggests its role in cardiovascular and metabolic diseases. Notch signaling components also control the phenotype of immune cells. Delta-like ligand 4 (Dll4) of the Notch pathway promotes proinflammatory activation of macrophages in vitro and in vivo. Dll4 blockade attenuates chronic atherosclerosis, vein graft disease, vascular calcification, insulin resistance, and fatty liver in mice. The Dll4-Notch axis may, thus, participate in the shared mechanisms for cardiometabolic disorders, serving as a potential therapeutic target for ameliorating these global health problems.
Collapse
Affiliation(s)
- Toshiaki Nakano
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Daiju Fukuda
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jun-Ichiro Koga
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
29
|
Nakahara T, Kawai-Kowase K, Matsui H, Sunaga H, Utsugi T, Iso T, Arai M, Tomono S, Kurabayashi M. Fibroblast growth factor 23 inhibits osteoblastic gene expression and induces osteoprotegerin in vascular smooth muscle cells. Atherosclerosis 2016; 253:102-110. [DOI: 10.1016/j.atherosclerosis.2016.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 08/05/2016] [Accepted: 08/18/2016] [Indexed: 12/20/2022]
|
30
|
Zhang H, Nan W, Wang S, Zhang T, Si H, Wang D, Yang F, Li G. Epidermal growth factor promotes proliferation of dermal papilla cells via Notch signaling pathway. Biochimie 2016; 127:10-8. [DOI: 10.1016/j.biochi.2016.04.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/18/2016] [Indexed: 01/06/2023]
|
31
|
Peng S, Gao D, Gao C, Wei P, Niu M, Shuai C. MicroRNAs regulate signaling pathways in osteogenic differentiation of mesenchymal stem cells (Review). Mol Med Rep 2016; 14:623-9. [PMID: 27222009 PMCID: PMC4918597 DOI: 10.3892/mmr.2016.5335] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 04/18/2016] [Indexed: 12/15/2022] Open
Abstract
Osteogenesis is a complex multi-step process involving the differentiation of mesenchymal stem cells (MSCs) into osteoblast progenitor cells, preosteoblasts, osteoblasts and osteocytes, and the crosstalk between multiple cell types for the formation and remodeling of bone. The signaling regulatory networks during osteogenesis include various components, including growth factors, transcription factors, micro (mi)RNAs and effectors, a number of which form feedback loops controlling the balance of osteogenic differentiation by positive or negative regulation. miRNAs have been found to be important regulators of osteogenic signaling pathways in multiple aspects and multiple signaling pathways. The present review focusses on the progress in elucidating the role of miRNA in the osteogenesis signaling networks of MSCs as a substitute for bone implantation the the field of bone tissue engineering. In particular, the review classifies which miRNAs promote or suppress the osteogenic process, and summarizes which signaling pathway these miRNAs are involved in. Improvements in knowledge of the characteristics of miRNAs in osteogenesis provide an important step for their application in translational investigations of bone tissue engineering and bone disease.
Collapse
Affiliation(s)
- Shuping Peng
- Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Dan Gao
- Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Chengde Gao
- State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha, Hunan 410083, P.R. China
| | - Pingpin Wei
- Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Man Niu
- Hunan Provincial Tumor Hospital and The Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha, Hunan 410083, P.R. China
| |
Collapse
|
32
|
Mechanical stimulation orchestrates the osteogenic differentiation of human bone marrow stromal cells by regulating HDAC1. Cell Death Dis 2016; 7:e2221. [PMID: 27171263 PMCID: PMC4917651 DOI: 10.1038/cddis.2016.112] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/21/2016] [Accepted: 03/24/2016] [Indexed: 01/18/2023]
Abstract
Mechanical stimulation and histone deacetylases (HDACs) have essential roles in regulating the osteogenic differentiation of bone marrow stromal cells (BMSCs) and bone formation. However, little is known regarding what regulates HDAC expression and therefore the osteogenic differentiation of BMSCs during osteogenesis. In this study, we investigated whether mechanical loading regulates HDAC expression directly and examined the role of HDACs in mechanical loading-triggered osteogenic differentiation and bone formation. We first studied the microarrays of samples from patients with osteoporosis and found that the NOTCH pathway and skeletal development gene sets were downregulated in the BMSCs of patients with osteoporosis. Then we demonstrated that mechanical stimuli can regulate osteogenesis and bone formation both in vivo and in vitro. NOTCH signaling was upregulated during cyclic mechanical stretch (CMS)-induced osteogenic differentiation, whereas HDAC1 protein expression was downregulated. The perturbation of HDAC1 expression also had a significant effect on matrix mineralization and JAG1-mediated Notch signaling, suggesting that HDAC1 acts as an endogenous attenuator of Notch signaling in the mechanotransduction of BMSCs. Chromatin immunoprecipitation (ChIP) assay results suggest that HDAC1 modulates the CMS-induced histone H3 acetylation level at the JAG1 promoter. More importantly, we found an inhibitory role of Hdac1 in regulating bone formation in response to hindlimb unloading in mice, and pretreatment with an HDAC1 inhibitor partly rescued the osteoporosis caused by mechanical unloading. Our results demonstrate, for the first time, that mechanical stimulation orchestrates genes expression involved in the osteogenic differentiation of BMSCs via the direct regulation of HDAC1, and the therapeutic inhibition of HDAC1 may be an efficient strategy for enhancing bone formation under mechanical stimulation.
Collapse
|
33
|
Ongaro A, Pellati A, Bagheri L, Rizzo P, Caliceti C, Massari L, De Mattei M. Characterization of Notch Signaling During Osteogenic Differentiation in Human Osteosarcoma Cell Line MG63. J Cell Physiol 2016; 231:2652-63. [PMID: 26946465 DOI: 10.1002/jcp.25366] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 03/01/2016] [Indexed: 11/10/2022]
Abstract
Osteogenic differentiation is a multi-step process controlled by a complex molecular framework. Notch is an evolutionarily conserved intercellular signaling pathway playing a prominent role in cell fate and differentiation, although the mechanisms by which this pathway regulates osteogenesis remain controversial. This study aimed to investigate, in vitro, the involvement of Notch pathway during all the developmental stages of osteogenic differentiation in human osteosarcoma cell line MG63. Cells were cultured in basal condition (control) and in osteoinductive medium (OM). Notch inhibitors were also added in OM to block Notch pathway. During osteogenic differentiation, early (alkaline phosphatase activity and collagen type I) and late osteogenic markers (osteocalcin levels and matrix mineralization), as well as the gene expression of the main osteogenic transcription factors (Runx2, Osterix, and Dlx5) increased. Time dependent changes in the expression of specific Notch receptors were identified in OM versus control with a significant reduction in the expression of Notch1 and Notch3 receptors in the early phase of differentiation, and an increase of Notch2 and Notch4 receptors in the late phase. Among Notch nuclear target genes, Hey1 expression was significantly higher in OM than control, while Hes5 expression decreased. Osteogenic markers were reduced and Hey1 was significantly inhibited by Notch inhibitors, suggesting a role for Notch through the canonical pathway. In conclusion, Notch pathway might be involved with a dual role in osteogenesis of MG63, through the activation of Notch2, Notch4, and Hey1, inducing osteoblast differentiation and the depression of Notch1, Notch3, and Hes5, maintaining an undifferentiated status. J. Cell. Physiol. 231: 2652-2663, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alessia Ongaro
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Agnese Pellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Leila Bagheri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Cristiana Caliceti
- Department of Chemistry "Giacomo Ciamician," Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Leo Massari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Monica De Mattei
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
34
|
Leem J, Lee IK. Mechanisms of Vascular Calcification: The Pivotal Role of Pyruvate Dehydrogenase Kinase 4. Endocrinol Metab (Seoul) 2016; 31:52-61. [PMID: 26996423 PMCID: PMC4803561 DOI: 10.3803/enm.2016.31.1.52] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/19/2016] [Accepted: 02/23/2016] [Indexed: 01/08/2023] Open
Abstract
Vascular calcification, abnormal mineralization of the vessel wall, is frequently associated with aging, atherosclerosis, diabetes mellitus, and chronic kidney disease. Vascular calcification is a key risk factor for many adverse clinical outcomes, including ischemic cardiac events and subsequent cardiovascular mortality. Vascular calcification was long considered to be a passive degenerative process, but it is now recognized as an active and highly regulated process similar to bone formation. However, despite numerous studies on the pathogenesis of vascular calcification, the mechanisms driving this process remain poorly understood. Pyruvate dehydrogenase kinases (PDKs) play an important role in the regulation of cellular metabolism and mitochondrial function. Recent studies show that PDK4 is an attractive therapeutic target for the treatment of various metabolic diseases. In this review, we summarize our current knowledge regarding the mechanisms of vascular calcification and describe the role of PDK4 in the osteogenic differentiation of vascular smooth muscle cells and development of vascular calcification. Further studies aimed at understanding the molecular mechanisms of vascular calcification will be critical for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Jaechan Leem
- Department of Immunology, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - In Kyu Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
- BK21 PLUS KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Korea.
| |
Collapse
|
35
|
Beazley KE, Nurminsky D, Lima F, Gandhi C, Nurminskaya MV. Wnt16 attenuates TGFβ-induced chondrogenic transformation in vascular smooth muscle. Arterioscler Thromb Vasc Biol 2015; 35:573-9. [PMID: 25614285 PMCID: PMC4344425 DOI: 10.1161/atvbaha.114.304393] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Phenotypic plasticity of vascular smooth muscle cells (VSMCs) contributes to cardiovascular disease. Chondrocyte-like transformation of VSMCs associates with vascular calcification and underlies the formation of aortic cartilaginous metaplasia induced in mice by genetic loss of matrix Gla protein (MGP). Previous microarray analysis identified a dramatic downregulation of Wnt16 in calcified MGP-null aortae, suggesting an antagonistic role for Wnt16 in the chondrogenic transformation of VSMCs. APPROACH AND RESULTS Wnt16 is significantly downregulated in MGP-null aortae, before the histological appearance of cartilaginous metaplasia, and in primary MGP-null VSMCs. In contrast, intrinsic TGFβ is activated in MGP-null VSMCs and is necessary for spontaneous chondrogenesis of these cells in high-density micromass cultures. TGFβ3-induced chondrogenic transformation in wild-type VSMCs associates with Smad2/3-dependent Wnt16 downregulation, but Wnt16 does not suppress TGFβ3-induced Smad activation. In addition, TGFβ3 inhibits Notch signaling in wild-type VSMCs, and this pathway is downregulated in MGP-null aortae. Exogenous Wnt16 stimulates Notch activity and attenuates TGFβ3-induced downregulation of Notch in wild-type VSMCs, prevents chondrogenesis in MGP-null and TGFβ3-treated wild-type VSMCs, and stabilizes expression of contractile markers of differentiated VSMCs. CONCLUSIONS We describe a novel TGFβ-Wnt16-Notch signaling conduit in the chondrocyte-like transformation of VSMCs and identify endogenous TGFβ activity in MGP-null VSMCs as a critical mediator of chondrogenesis. Our proposed model suggests that the activated TGFβ pathway inhibits expression of Wnt16, which is a positive regulator of Notch signaling and a stabilizer of VSMC phenotype. These data advance the comprehensive mechanistic understanding of VSMC transformation and may identify a novel potential therapeutic target in vascular calcification.
Collapse
Affiliation(s)
- Kelly E Beazley
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Dmitry Nurminsky
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Florence Lima
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Chintan Gandhi
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore
| | - Maria V Nurminskaya
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore.
| |
Collapse
|
36
|
Mu X, Tang Y, Lu A, Takayama K, Usas A, Wang B, Weiss K, Huard J. The role of Notch signaling in muscle progenitor cell depletion and the rapid onset of histopathology in muscular dystrophy. Hum Mol Genet 2015; 24:2923-37. [PMID: 25678553 DOI: 10.1093/hmg/ddv055] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/09/2015] [Indexed: 02/05/2023] Open
Abstract
Although it has been speculated that stem cell depletion plays a role in the rapid progression of the muscle histopathology associated with Duchenne Muscular Dystrophy (DMD), the molecular and cellular mechanisms responsible for stem cell depletion remain poorly understood. The rapid depletion of muscle stem cells has not been observed in the dystrophin-deficient model of DMD (mdx mouse), which may explain the relatively mild dystrophic phenotype observed in this animal model. In contrast, we have observed a rapid occurrence of stem cell depletion in the dystrophin/utrophin double knockout (dKO) mouse model, which exhibits histopathological features that more closely recapitulate the phenotype observed in DMD patients compared with the mdx mouse. Notch signaling has been found to be a key regulator of stem cell self-renewal and myogenesis in normal skeletal muscle; however, little is known about the role that Notch plays in the development of the dystrophic histopathology associated with DMD. Our results revealed an over-activation of Notch in the skeletal muscles of dKO mice, which correlated with sustained inflammation, impaired muscle regeneration and the rapid depletion and senescence of the muscle progenitor cells (MPCs, i.e. Pax7+ cells). Consequently, the repression of Notch in the skeletal muscle of dKO mice delayed/reduced the depletion and senescence of MPCs, and restored the myogenesis capacity while reducing inflammation and fibrosis. We suggest that the down-regulation of Notch could represent a viable approach to reduce the dystrophic histopathologies associated with DMD.
Collapse
Affiliation(s)
- Xiaodong Mu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ying Tang
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Aiping Lu
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Koji Takayama
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Arvydas Usas
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Bing Wang
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Kurt Weiss
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Johnny Huard
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
37
|
Zhu F, Sweetwyne MT, Hankenson KD. PKCδ is required for Jagged-1 induction of human mesenchymal stem cell osteogenic differentiation. Stem Cells 2014; 31:1181-92. [PMID: 23404789 DOI: 10.1002/stem.1353] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 12/29/2012] [Indexed: 12/22/2022]
Abstract
JAG1, the gene for the Jagged-1 ligand (Jag1) in the Notch signaling pathway, is variably mutated in Alagille Syndrome (ALGS). ALGS patients have skeletal defects, and additionally JAG1 has been shown to be associated with low bone mass through genome-wide association studies. Plating human osteoblast precursors (human mesenchymal stem cells-hMSCs) on Jag1 is sufficient to induce osteoblast differentiation; however, exposure of mouse MSC (mMSC) to Jag1 actually inhibits osteoblastogenesis. Overexpression of the notch-2 intracellular domain (NICD2) is sufficient to mimic the effect of Jag1 on hMSC osteoblastogenesis, while blocking Notch signaling with a γ-secretase inhibitor or with dominant-negative mastermind inhibits Jag1-induced hMSC osteoblastogenesis. In pursuit of interacting signaling pathways, we discovered that treatment with a protein kinase C δ (PKCδ) inhibitor abrogates Jag1-induced hMSC osteoblastogenesis. Jag1 results in rapid PKCδ nuclear translocation and kinase activation. Furthermore, Jag1 stimulates the physical interaction of PKCδ with NICD. Collectively, these results suggest that Jag1 induces hMSC osteoblast differentiation through canonical Notch signaling and requires concomitant PKCδ signaling. This research also demonstrates potential deficiencies in using mouse models to study ALGS bone abnormalities.
Collapse
Affiliation(s)
- Fengchang Zhu
- Department of Clinical Studies-New Bolton Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
38
|
Sakamoto A, Ishizaka N, Imai Y, Ando J, Nagai R, Komuro I. Association of serum IgG4 and soluble interleukin-2 receptor levels with epicardial adipose tissue and coronary artery calcification. Clin Chim Acta 2014. [DOI: 10.1016/j.cca.2013.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
39
|
Osathanon T, Nowwarote N, Manokawinchoke J, Pavasant P. bFGF and JAGGED1 regulate alkaline phosphatase expression and mineralization in dental tissue-derived mesenchymal stem cells. J Cell Biochem 2013; 114:2551-61. [DOI: 10.1002/jcb.24602] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 05/17/2013] [Indexed: 01/26/2023]
Affiliation(s)
| | - Nunthawan Nowwarote
- Mineralized Tissue Research Unit; Chulalongkorn University; Bangkok; 10330; Thailand
| | | | | |
Collapse
|
40
|
Zhou S, Fang X, Xin H, Li W, Qiu H, Guan S. Osteoprotegerin inhibits calcification of vascular smooth muscle cell via down regulation of the Notch1-RBP-Jκ/Msx2 signaling pathway. PLoS One 2013; 8:e68987. [PMID: 23874840 PMCID: PMC3711585 DOI: 10.1371/journal.pone.0068987] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/03/2013] [Indexed: 01/06/2023] Open
Abstract
Objective Vascular calcification is a common pathobiological process which occurs among
the elder population and in patients with diabetes and chronic kidney
disease. Osteoprotegerin, a secreted glycoprotein that regulates bone mass,
has recently emerged as an important regulator of the development of
vascular calcification. However, the mechanism is not fully understood. The
purpose of this study is to explore novel signaling mechanisms of
osteoprotegerin in the osteoblastic differentiation in rat aortic vascular
smooth muscle cells (VSMCs). Methods and Results VSMCs were isolated from thoracic aorta of Sprague Dawley rats. Osteoblastic
differentiation of VSMCs was induced by an osteogenic medium. We confirmed
by Von Kossa staining and direct cellular calcium measurement that
mineralization was significantly increased in VSMCs cultured in osteogenic
medium; consistent with an enhanced alkaline phosphatase activity. This
osteoblastic differentiation in VSMCs was significantly reduced by the
addition of osteoprotegerin in a dose responsive manner. Moreover, we
identified, by real-time qPCR and western blotting, that expression of
Notch1 and RBP-Jκ were significantly up-regulated in VSMCs cultured in
osteogenic medium at both the mRNA and protein levels, these effects were
dose-dependently abolished by the treatment of osteoprotegerin. Furthermore,
we identified that Msx2, a downstream target of the Notch1/RBP-Jκ signaling,
was markedly down-regulated by the treatment of osteoprotegerin. Conclusion Osteoprotegerin inhibits vascular calcification through the down regulation
of the Notch1-RBP-Jκ signaling pathway.
Collapse
Affiliation(s)
- Shaoqiong Zhou
- Department of Gerontology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Xing Fang
- Department of Gerontology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Huaping Xin
- Department of Gerontology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Department of Gerontology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
| | - Hongyu Qiu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical
School, University of Medicine and Dentistry of New Jersey (UMDNJ), Newark, New
Jersey, United States of America
- * E-mail:
(SG); (HQ)
| | - Siming Guan
- Department of Gerontology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
(SG); (HQ)
| |
Collapse
|
41
|
ZHOU SHAOQIONG, FANG XIN, XIN HUAPING, GUAN SIMING. Effects of alendronate on the Notch1-RBP-Jκ signaling pathway in the osteogenic differentiation and mineralization of vascular smooth muscle cells. Mol Med Rep 2013; 8:89-94. [DOI: 10.3892/mmr.2013.1489] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 05/09/2013] [Indexed: 11/06/2022] Open
|
42
|
Zeng Q, Song R, Ao L, Weyant MJ, Lee J, Xu D, Fullerton DA, Meng X. Notch1 promotes the pro-osteogenic response of human aortic valve interstitial cells via modulation of ERK1/2 and nuclear factor-κB activation. Arterioscler Thromb Vasc Biol 2013; 33:1580-90. [PMID: 23640488 DOI: 10.1161/atvbaha.112.300912] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Calcific aortic valve disease is a leading cardiovascular disease in the elderly, and progressive calcification results in the failure of valvular function. Aortic valve interstitial cells (AVICs) from stenotic valves express higher levels of bone morphogenetic protein-2 in response to Toll-like receptor 4 stimulation. We recently found that Toll-like receptor 4 interacts with Notch1 in human AVICs. This study tests the hypothesis that Notch1 promotes the pro-osteogenic response of human AVICs. APPROACH AND RESULTS AVICs isolated from diseased human valves expressed higher levels of bone morphogenetic protein-2 and alkaline phosphatase after lipopolysaccharide stimulation. The augmented pro-osteogenic response is associated with elevated cellular levels of Notch1 and enhanced Notch1 cleavage in response to lipopolysaccharide stimulation. Inhibition or silencing of Notch1 suppressed the pro-osteogenic response in diseased cells, and the Notch 1 ligand, Jagged1, enhanced the response in AVICs isolated from normal human valves. Interestingly, extracellular signal-regulated protein kinases 1/2 (ERK1/2) and nuclear factor-κB phosphorylation induced by lipopolysaccharide was markedly reduced by inhibition or silencing of Notch1 and enhanced by Jagged1. Inhibition of ERK1/2 or nuclear factor-κB also reduced bone morphogenetic protein-2 and alkaline phosphatase expression induced by lipopolysaccharide. CONCLUSIONS Notch1 mediates the pro-osteogenic response to Toll-like receptor 4 stimulation in human AVICs. Elevated Notch1 levels and enhanced Notch1 activation play a major role in augmentation of the pro-osteogenic response of AVICs of stenotic valves through modulation of ERK1/2 and nuclear factor-κB activation. These pathways could be potential therapeutic targets for prevention of the progression of calcific aortic valve disease.
Collapse
Affiliation(s)
- Qingchun Zeng
- Department of Surgery, University of Colorado Denver, Aurora, CO, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Sharma B, Albig AR. Matrix Gla protein reinforces angiogenic resolution. Microvasc Res 2013; 85:24-33. [PMID: 23110920 PMCID: PMC3629274 DOI: 10.1016/j.mvr.2012.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 10/01/2012] [Accepted: 10/19/2012] [Indexed: 12/18/2022]
Abstract
Matrix Gla Protein (MGP) is an ECM molecule commonly associated with dysfunctions of large blood vessels such as arteriosclerosis and atherosclerosis. However, the exact role of MGP in the microvasculature is not clear. Utilizing a mouse MGP knockout model we found that MGP suppresses angiogenic sprouting from mouse aorta restricts microvascular density in cardiac and skeletal muscle, and is an endogenous inhibitor of tumor angiogenesis. Similarly, morpholino based knockdown of MGP in zebrafish embryos caused a progressive loss of luminal structures in intersegmental vessels, a phenotype reminiscent of Dll4/Notch inhibition. Accordingly, MGP suppressed Notch-dependent Hes-1 promoter activity and expression of Jagged1 mRNA relative to Dll4 mRNA. However, inhibition of BMP but not Notch or VEGF signaling reversed the excessive angiogenic sprouting phenotype of MGP knockout aortic rings suggesting that MGP may normally suppress angiogenic sprouting by blocking BMP signaling. Collectively, these results suggest that MGP is a multi-functional inhibitor of normal and abnormal angiogenesis that may function by coordinating with both Notch and BMP signaling pathways.
Collapse
Affiliation(s)
- Bikram Sharma
- Department of Biology, Indiana State University, Terre Haute, IN 47809 USA
| | - Allan R. Albig
- Department of Biology, Boise State University, Boise ID. 83725
| |
Collapse
|
44
|
Sallam T, Cheng H, Demer LL, Tintut Y. Regulatory circuits controlling vascular cell calcification. Cell Mol Life Sci 2012; 70:3187-97. [PMID: 23269436 DOI: 10.1007/s00018-012-1231-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/12/2012] [Accepted: 12/03/2012] [Indexed: 12/20/2022]
Abstract
Vascular calcification is a common feature of chronic kidney disease, cardiovascular disease, and aging. Such abnormal calcium deposition occurs in medial and/or intimal layers of blood vessels as well as in cardiac valves. Once considered a passive and inconsequential finding, the presence of calcium deposits in the vasculature is widely accepted as a predictor of increased morbidity and mortality. Recognition of the importance of vascular calcification in health is driving research into mechanisms that govern its development, progression, and regression. Diverse, but highly interconnected factors, have been implicated, including disturbances in lipid metabolism, oxidative stress, inflammatory cytokines, and mineral and hormonal balances, which can lead to formation of osteoblast-like cells in the artery wall. A tight balance of procalcific and anticalcific regulators dictates the extent of disease. In this review, we focus on the main regulatory circuits modulating vascular cell calcification.
Collapse
Affiliation(s)
- Tamer Sallam
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Center for the Health Sciences, A2-237, 10833 Le Conte Ave., Los Angeles, CA, 90095-1679, USA
| | | | | | | |
Collapse
|
45
|
Shimizu T, Tanaka T, Iso T, Kawai-Kowase K, Kurabayashi M. Azelnidipine inhibits Msx2-dependent osteogenic differentiation and matrix mineralization of vascular smooth muscle cells. Int Heart J 2012; 53:331-5. [PMID: 23038096 DOI: 10.1536/ihj.53.331] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Vascular calcification is an active and regulated process that is similar to bone formation. While calcium channel blockers (CCBs) have been shown to improve outcomes in atherosclerotic vascular disease, it remains unknown whether CCBs have an effect on the process of vascular calcification. Here we investigated whether CCBs inhibit osteogenic differentiation and matrix mineralization of vascular smooth muscle cells induced by Msx2, a key factor of vascular calcification. Human aortic smooth muscle cells (HASMCs) were transduced with adenovirus expressing MSX2 and were treated with 3 distinct CCBs. Azelnidipine, a dihydropyridine subclass of CCBs, significantly decreased alkaline phosphatase (ALP) activity of Msx2-overexpressed HASMCs, whereas verapamil and diltiazem had no effect. Furthermore, azelnidipine, but not verapamil and diltiazem, significantly decreased matrix mineralization of Msx2-overexpressing HASMCs. Azelnidipine significantly attenuated the induction of ALP gene expression by Msx2, a key transcription factor in osteogenesis, while it did not reduce enzymatic activity of ALP. Furthermore, azelnidipine inhibited the ability of Msx2 to activate the ALP gene, but had no effect on Notch-induced Msx2 expression. Given that L-type calcium channels are equally blocked by these CCBs, our results suggest that azelnidipine inhibits the Msx2-dependent process of vascular calcification by mechanisms other than inhibition of calcium channel activity.
Collapse
Affiliation(s)
- Takehisa Shimizu
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, Gunma, Japan
| | | | | | | | | |
Collapse
|
46
|
Vo TN, Kasper FK, Mikos AG. Strategies for controlled delivery of growth factors and cells for bone regeneration. Adv Drug Deliv Rev 2012; 64:1292-309. [PMID: 22342771 PMCID: PMC3358582 DOI: 10.1016/j.addr.2012.01.016] [Citation(s) in RCA: 460] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 01/23/2012] [Accepted: 01/30/2012] [Indexed: 12/15/2022]
Abstract
The controlled delivery of growth factors and cells within biomaterial carriers can enhance and accelerate functional bone formation. The carrier system can be designed with pre-programmed release kinetics to deliver bioactive molecules in a localized, spatiotemporal manner most similar to the natural wound healing process. The carrier can also act as an extracellular matrix-mimicking substrate for promoting osteoprogenitor cellular infiltration and proliferation for integrative tissue repair. This review discusses the role of various regenerative factors involved in bone healing and their appropriate combinations with different delivery systems for augmenting bone regeneration. The general requirements of protein, cell and gene therapy are described, with elaboration on how the selection of materials, configurations and processing affects growth factor and cell delivery and regenerative efficacy in both in vitro and in vivo applications for bone tissue engineering.
Collapse
Affiliation(s)
- Tiffany N. Vo
- Department of Bioengineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
| | - F. Kurtis Kasper
- Department of Bioengineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
| | - Antonios G. Mikos
- Department of Bioengineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
- Department of Chemical and Biomolecular Engineering, Rice University, P.O. Box 1892, MS 142, Houston, TX 77251-1892, USA
| |
Collapse
|
47
|
Notch ligand delta-like 4 blockade attenuates atherosclerosis and metabolic disorders. Proc Natl Acad Sci U S A 2012; 109:E1868-77. [PMID: 22699504 DOI: 10.1073/pnas.1116889109] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis and insulin resistance are major components of the cardiometabolic syndrome, a global health threat associated with a systemic inflammatory state. Notch signaling regulates tissue development and participates in innate and adaptive immunity in adults. The role of Notch signaling in cardiometabolic inflammation, however, remains obscure. We noted that a high-fat, high-cholesterol diet increased expression of the Notch ligand Delta-like 4 (Dll4) in atheromata and fat tissue in LDL-receptor-deficient mice. Blockade of Dll4-Notch signaling using neutralizing anti-Dll4 antibody attenuated the development of atherosclerosis, diminished plaque calcification, improved insulin resistance, and decreased fat accumulation. These changes were accompanied by decreased macrophage accumulation, diminished expression of monocyte chemoattractant protein-1 (MCP-1), and lower levels of nuclear factor-κB (NF-κB) activation. In vitro cell culture experiments revealed that Dll4-mediated Notch signaling increases MCP-1 expression via NF-κB, providing a possible mechanism for in vivo effects. Furthermore, Dll4 skewed macrophages toward a proinflammatory phenotype ("M1"). These results suggest that Dll4-Notch signaling plays a central role in the shared mechanism for the pathogenesis of cardiometabolic disorders.
Collapse
|
48
|
Boucher J, Gridley T, Liaw L. Molecular pathways of notch signaling in vascular smooth muscle cells. Front Physiol 2012; 3:81. [PMID: 22509166 PMCID: PMC3321637 DOI: 10.3389/fphys.2012.00081] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Accepted: 03/19/2012] [Indexed: 11/20/2022] Open
Abstract
Notch signaling in the cardiovascular system is important during embryonic development, vascular repair of injury, and vascular pathology in humans. The vascular smooth muscle cell (VSMC) expresses multiple Notch receptors throughout its life cycle, and responds to Notch ligands as a regulatory mechanism of differentiation, recruitment to growing vessels, and maturation. The goal of this review is to provide an overview of the current understanding of the molecular basis for Notch regulation of VSMC phenotype. Further, we will explore Notch interaction with other signaling pathways important in VSMC.
Collapse
Affiliation(s)
- Joshua Boucher
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough, ME, USA
| | | | | |
Collapse
|
49
|
Liu Y, Wang T, Yan J, Jiagbogu N, Heideman DA, Canfield AE, Alexander MY. HGF/c-Met signalling promotes Notch3 activation and human vascular smooth muscle cell osteogenic differentiation in vitro. Atherosclerosis 2011; 219:440-7. [PMID: 21920521 PMCID: PMC3925803 DOI: 10.1016/j.atherosclerosis.2011.08.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 07/21/2011] [Accepted: 08/18/2011] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Vascular calcification is a major clinical problem and elucidating the underlying mechanism is important to improve the prognosis of patients with cardiovascular disease. We aimed to elucidate the role and mechanism of action of Hepatocyte Growth Factor (HGF)/c-Met signalling in vascular calcification and establish whether blocking this pathway could prevent mineralisation of vascular smooth muscle cells (VSMCs) in vitro. METHODS AND RESULTS We demonstrate increased HGF secretion and c-Met up-regulation and phosphorylation during VSMC osteogenic differentiation. Adenoviral-mediated over-expression of HGF (AdHGF) in VSMCs accelerated mineralisation, shown by alizarin red staining, and significantly increased (45)Calcium incorporation (1.96 ± 0.54-fold [P < 0.05]) and alkaline phosphatase (ALP) activity (3.01 ± 0.8-fold [P < 0.05]) compared to controls. AdHGF also significantly elevated mRNA expression of bone-related proteins, Runx2, osteocalcin, BMP2 and osterix in VSMCs. AdHGF-accelerated mineralisation correlated with increased Akt phosphorylation, nuclear translocation of Notch3 intracellular domain (N3IC) and up-regulation of the Notch3 target protein, HES1. In contrast, adenoviral-mediated over-expression of the HGF antagonist, NK4, markedly attenuated VSMC mineralisation, and reduced c-Met phosphorylation, Akt activation and HES1 protein expression compared to AdHGF-treated cells. Furthermore, the Notch inhibitor, DAPT, attenuated N3IC nuclear translocation and AdHGF-induced mineralisation. CONCLUSION We demonstrate HGF induces VSMC osteogenic differentiation via c-Met/Akt/Notch3 signalling, highlighting these pathways as potential targets for intervention of vascular calcification.
Collapse
MESH Headings
- Adenoviridae/genetics
- Alkaline Phosphatase/metabolism
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Bone Morphogenetic Protein 2/genetics
- Calcium/metabolism
- Cell Differentiation
- Cells, Cultured
- Core Binding Factor Alpha 1 Subunit/genetics
- Genetic Vectors
- Hepatocyte Growth Factor/genetics
- Hepatocyte Growth Factor/metabolism
- Homeodomain Proteins/metabolism
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteogenesis
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-met/metabolism
- RNA, Messenger/metabolism
- Receptor, Notch3
- Receptors, Notch/metabolism
- Signal Transduction
- Sp7 Transcription Factor
- Time Factors
- Transcription Factor HES-1
- Transcription Factors/genetics
- Transfection
- Up-Regulation
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Yiwen Liu
- Cardiovascular Research Group, University of Manchester, UK
| | - Tao Wang
- Medical Genetics Research Group, University of Manchester, UK
| | - Jianyun Yan
- Cardiovascular Research Group, University of Manchester, UK
| | - Naomi Jiagbogu
- Cardiovascular Research Group, University of Manchester, UK
| | | | - Ann E. Canfield
- Cardiovascular Research Group, University of Manchester, UK
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, UK
| | | |
Collapse
|
50
|
Boström KI, Rajamannan NM, Towler DA. The regulation of valvular and vascular sclerosis by osteogenic morphogens. Circ Res 2011; 109:564-77. [PMID: 21852555 DOI: 10.1161/circresaha.110.234278] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular calcification increasingly afflicts our aging, dysmetabolic population. Once considered only a passive process of dead and dying cells, vascular calcification has now emerged as a highly regulated form of biomineralization organized by collagenous and elastin extracellular matrices. During skeletal bone formation, paracrine epithelial-mesenchymal and endothelial-mesenchymal interactions control osteochondrocytic differentiation of multipotent mesenchymal progenitor cells. These paracrine osteogenic signals, mediated by potent morphogens of the bone morphogenetic protein and wingless-type MMTV integration site family member (Wnt) superfamilies, are also active in the programming of arterial osteoprogenitor cells during vascular and valve calcification. Inflammatory cytokines, reactive oxygen species, and oxylipids-increased in the clinical settings of atherosclerosis, diabetes, and uremia that promote arteriosclerotic calcification-elicit the ectopic vascular activation of osteogenic morphogens. Specific extracellular and intracellular inhibitors of bone morphogenetic protein-Wnt signaling have been identified as contributing to the regulation of osteogenic mineralization during development and disease. These inhibitory pathways and their regulators afford the development of novel therapeutic strategies to prevent and treat valve and vascular sclerosis.
Collapse
Affiliation(s)
- Kristina I Boström
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLA, 10833 LeConte Ave, Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|