1
|
Choi SH, Kim E, Heo SJ, Seol MY, Chung Y, Yoon HI. Integrative prediction model for radiation pneumonitis incorporating genetic and clinical-pathological factors using machine learning. Clin Transl Radiat Oncol 2024; 48:100819. [PMID: 39161733 PMCID: PMC11332843 DOI: 10.1016/j.ctro.2024.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
Purpose We aimed to develop a machine learning-based prediction model for severe radiation pneumonitis (RP) by integrating relevant clinicopathological and genetic factors, considering the associations of clinical, dosimetric parameters, and single nucleotide polymorphisms (SNPs) of genes in the TGF-β1 pathway with RP. Methods We prospectively enrolled 59 primary lung cancer patients undergoing radiotherapy and analyzed pretreatment blood samples, clinicopathological/dosimetric variables, and 11 functional SNPs in TGFβ pathway genes. Using the Synthetic Minority Over-sampling Technique (SMOTE) and nested cross-validation, we developed a machine learning-based prediction model for severe RP (grade ≥ 2). Feature selection was conducted using four methods (filtered-based, wrapper-based, embedded, and logistic regression), and performance was evaluated using three machine learning models. Results Severe RP occurred in 20.3 % of patients with a median follow-up of 39.7 months. In our final model, age (>66 years), smoking history, PTV volume (>300 cc), and AG/GG genotype in BMP2 rs1979855 were identified as the most significant predictors. Additionally, incorporating genomic variables for prediction alongside clinicopathological variables significantly improved the AUC compared to using clinicopathological variables alone (0.822 vs. 0.741, p = 0.029). The same feature set was selected using both the wrapper-based method and logistic model, demonstrating the best performance across all machine learning models (AUC: XGBoost 0.815, RF 0.805, SVM 0.712, respectively). Conclusion We successfully developed a machine learning-based prediction model for RP, demonstrating age, smoking history, PTV volume, and BMP2 rs1979855 genotype as significant predictors. Notably, incorporating SNP data significantly enhanced predictive performance compared to clinicopathological factors alone.
Collapse
Affiliation(s)
- Seo Hee Choi
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Euidam Kim
- Department of Nuclear Engineering, Hanyang University, Seoul, Republic of Korea
| | - Seok-Jae Heo
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi Youn Seol
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoonsun Chung
- Department of Nuclear Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Heavy Ion Therapy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Cai Z, Liu Z, Zhang Y, Ma H, Li R, Guo S, Wu S, Guo X. Associations Between Life's Essential 8 and Abdominal Aortic Calcification Among Middle-Aged and Elderly Populations. J Am Heart Assoc 2023; 12:e031146. [PMID: 38063150 PMCID: PMC10863763 DOI: 10.1161/jaha.123.031146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Abdominal aortic calcification (AAC) is an independent risk factor for cardiovascular disease. We aim to examine the associations between Life's Essential 8 (LE8), the recently updated measurement of cardiovascular health (CVH), and AAC among participants aged ≥40 years. METHODS AND RESULTS This population-based cross-sectional study used data from the National Health and Nutrition Examination Survey in 2013 to 2014. AAC (AAC score>0) and severe AAC (AAC score>6) were quantified by the Kauppila score system. Multiple linear, multivariable logistic, and restricted cubic spline models were used to assess the associations. A total of 2369 participants were included with a mean AAC score of 1.41 (0.13). Participants in the high-cardiovascular-health group had lower AAC scores, lower prevalence of AAC, and lower prevalence of severe AAC. After the adjustment of potential confounders (age, sex, race and ethnicity, education levels, marital status, poverty income ratio, estimated glomerular filtration rate, serum creatinine, serum uric acid, serum phosphorus, and serum total calcium), higher cardiovascular health was significantly associated with lower risk of AAC. Meanwhile, elevated nicotine exposure score, blood glucose score, and blood pressure score within the LE8 components were significantly associated with lower risk of AAC. Also, nonlinear dose-response relationships were observed. Subgroup analyses (age strata, sex, poverty income ratio, education levels, marital status) indicated the inverse associations of LE8 and AAC were generally similar in different populations. CONCLUSIONS LE8 was negatively and nonlinearly related to the risk of AAC among middle-aged and older populations. Meanwhile, LE8 components should prioritize higher scores for nicotine exposure, blood glucose, and blood pressure evaluations.
Collapse
Affiliation(s)
- Zongao Cai
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
| | - Hongxuan Ma
- Department of Kidney TransportationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Ruihui Li
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuang Guo
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shiyong Wu
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xueli Guo
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
3
|
Carmona A, Guerrero F, Muñoz-Castañeda JR, Jimenez MJ, Rodriguez M, Soriano S, Martin-Malo A. Uremic Toxins Induce THP-1 Monocyte Endothelial Adhesion and Migration through Specific miRNA Expression. Int J Mol Sci 2023; 24:12938. [PMID: 37629118 PMCID: PMC10455080 DOI: 10.3390/ijms241612938] [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: 07/20/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Atherosclerosis is initiated by the activation of endothelial cells that allows monocyte adhesion and transmigration through the vascular wall. The accumulation of uremic toxins such as indoxyl sulphate (IS) and p-cresol (PC) has been associated with atherosclerosis. Currently, miRNAs play a crucial role in the regulation of monocyte activation, adhesion, and trans-endothelial migration. The aim of the present study is to evaluate the effect of IS and PC on monocyte adhesion and migration processes in monocytes co-cultured with endothelial cells as well as to determine the underlying mechanisms. The incubation of HUVECs and THP-1 cells with both IS and PC toxins resulted in an increased migratory capacity of THP-1 cells. Furthermore, the exposure of THP-1 cells to both uremic toxins resulted in the upregulation of BMP-2 and miRNAs-126-3p, -146b-5p, and -223-3p, as well as the activation of nuclear factor kappa B (NF-κB) and a decrease in its inhibitor IĸB. Uremic toxins, such as IS and PC, enhance the migratory and adhesion capacity of THP-1 cells to the vascular endothelium. These toxins, particularly PC, contribute significantly to uremia-associated vascular disease by increasing in THP-1 cells the expression of BMP-2, NF-κB, and key miRNAs associated with the development of atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Andres Carmona
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (A.C.); (M.J.J.); (M.R.); (S.S.); (A.M.-M.)
| | - Fatima Guerrero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (A.C.); (M.J.J.); (M.R.); (S.S.); (A.M.-M.)
- Department of Medicine, University of Cordoba, 14004 Córdoba, Spain
| | - Juan R. Muñoz-Castañeda
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (A.C.); (M.J.J.); (M.R.); (S.S.); (A.M.-M.)
- Nephrology Unit, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria Jose Jimenez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (A.C.); (M.J.J.); (M.R.); (S.S.); (A.M.-M.)
| | - Mariano Rodriguez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (A.C.); (M.J.J.); (M.R.); (S.S.); (A.M.-M.)
- Department of Medicine, University of Cordoba, 14004 Córdoba, Spain
- Nephrology Unit, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sagrario Soriano
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (A.C.); (M.J.J.); (M.R.); (S.S.); (A.M.-M.)
- Nephrology Unit, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alejandro Martin-Malo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (A.C.); (M.J.J.); (M.R.); (S.S.); (A.M.-M.)
- Department of Medicine, University of Cordoba, 14004 Córdoba, Spain
- Nephrology Unit, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Das S, Mondal A, Dey C, Chakraborty S, Bhowmik R, Karmakar S, Sengupta A. ER stress induces upregulation of transcription factor Tbx20 and downstream Bmp2 signaling to promote cardiomyocyte survival. J Biol Chem 2023; 299:103031. [PMID: 36805334 PMCID: PMC10036653 DOI: 10.1016/j.jbc.2023.103031] [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: 11/21/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/17/2023] Open
Abstract
In the mammalian heart, fetal cardiomyocytes proliferate prior to birth; however, they exit the cell cycle shortly after birth. Recent studies show that adult cardiomyocytes re-enters the cell cycle postinjury to promote cardiac regeneration. The endoplasmic reticulum (ER) orchestrates the production and assembly of different types of proteins, and a disruption in this machinery leads to the generation of ER stress, which activates the unfolded protein response. There is a very fine balance between ER stress-mediated protective and proapoptotic responses. T-box transcription factor 20 (Tbx20) promotes embryonic and adult cardiomyocyte proliferation postinjury to restore cardiac homeostasis. However, the function and regulatory interactions of Tbx20 in ER stress-induced cardiomyopathy have not yet been reported. We show here that ER stress upregulates Tbx20, which activates downstream bone morphogenetic protein 2 (Bmp2)-pSmad1/5/8 signaling to induce cardiomyocyte proliferation and limit apoptosis. However, augmenting ER stress reverses this protective response. We also show that increased expression of tbx20 during ER stress is mediated by the activating transcription factor 6 arm of the unfolded protein response. Cardiomyocyte-specific loss of Tbx20 results in decreased cardiomyocyte proliferation and increased apoptosis. Administration of recombinant Bmp2 protein during ER stress upregulates Tbx20 leading to augmented proliferation, indicating a feed-forward loop mechanism. In in vivo ER stress, as well as in diabetic cardiomyopathy, the activity of Tbx20 is increased with concomitant increased cardiomyocyte proliferation and decreased apoptosis. These data support a critical role of Tbx20-Bmp2 signaling in promoting cardiomyocyte survival during ER stress-induced cardiomyopathies.
Collapse
Affiliation(s)
- Shreya Das
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, India
| | - Arunima Mondal
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, India
| | - Chandrani Dey
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, India
| | | | - Rudranil Bhowmik
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Sanmoy Karmakar
- Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Arunima Sengupta
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, India.
| |
Collapse
|
5
|
Sutton NR, Malhotra R, Hilaire C, Aikawa E, Blumenthal RS, Gackenbach G, Goyal P, Johnson A, Nigwekar SU, Shanahan CM, Towler DA, Wolford BN, Chen Y. Molecular Mechanisms of Vascular Health: Insights From Vascular Aging and Calcification. Arterioscler Thromb Vasc Biol 2023; 43:15-29. [PMID: 36412195 PMCID: PMC9793888 DOI: 10.1161/atvbaha.122.317332] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
Cardiovascular disease is the most common cause of death worldwide, especially beyond the age of 65 years, with the vast majority of morbidity and mortality due to myocardial infarction and stroke. Vascular pathology stems from a combination of genetic risk, environmental factors, and the biologic changes associated with aging. The pathogenesis underlying the development of vascular aging, and vascular calcification with aging, in particular, is still not fully understood. Accumulating data suggests that genetic risk, likely compounded by epigenetic modifications, environmental factors, including diabetes and chronic kidney disease, and the plasticity of vascular smooth muscle cells to acquire an osteogenic phenotype are major determinants of age-associated vascular calcification. Understanding the molecular mechanisms underlying genetic and modifiable risk factors in regulating age-associated vascular pathology may inspire strategies to promote healthy vascular aging. This article summarizes current knowledge of concepts and mechanisms of age-associated vascular disease, with an emphasis on vascular calcification.
Collapse
Affiliation(s)
- Nadia R. Sutton
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Rajeev Malhotra
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Cynthia Hilaire
- Division of Cardiology, Departments of Medicine and Bioengineering, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, 1744 BSTWR, 200 Lothrop St, Pittsburgh, PA, 15260 USA
| | - Elena Aikawa
- Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Roger S. Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease; Baltimore, MD
| | - Grace Gackenbach
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Parag Goyal
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Adam Johnson
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Sagar U. Nigwekar
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Catherine M. Shanahan
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | - Dwight A. Towler
- Department of Medicine | Endocrine Division and Pak Center for Mineral Metabolism Research, UT Southwestern Medical Center, Dallas, TX USA
| | - Brooke N. Wolford
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham and Research Department, Veterans Affairs Birmingham Medical Center, Birmingham, AL, USA
| |
Collapse
|
6
|
Hachana S, Larrivée B. TGF-β Superfamily Signaling in the Eye: Implications for Ocular Pathologies. Cells 2022; 11:2336. [PMID: 35954181 PMCID: PMC9367584 DOI: 10.3390/cells11152336] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023] Open
Abstract
The TGF-β signaling pathway plays a crucial role in several key aspects of development and tissue homeostasis. TGF-β ligands and their mediators have been shown to be important regulators of ocular physiology and their dysregulation has been described in several eye pathologies. TGF-β signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis. Inadequate TGF-β signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis. In addition, experimental models of corneal neovascularization, diabetic retinopathy, proliferative vitreoretinopathy, glaucoma, or corneal injury suggest that aberrant TGF-β signaling may contribute to the pathological features of these conditions, showing the potential of modulating TGF-β signaling to treat eye diseases. This review highlights the key roles of TGF-β family members in ocular physiology and in eye diseases, and reviews approaches targeting the TGF-β signaling as potential treatment options.
Collapse
Affiliation(s)
- Soumaya Hachana
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
7
|
Coppola A, Vigorito C, Lombari P, Martínez YG, Borriello M, Trepiccione F, Ingrosso D, Perna AF. Uremic Toxin Lanthionine Induces Endothelial Cell Mineralization In Vitro. Biomedicines 2022; 10:biomedicines10020444. [PMID: 35203651 PMCID: PMC8962276 DOI: 10.3390/biomedicines10020444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Vascular calcification (VC) is a pathological event caused by the unusual deposition of minerals in the vascular system, representing the leading cause of cardiovascular mortality in chronic kidney disease (CKD). In CKD, the deregulation of calcium and phosphate metabolism, along with the effect of several uremic toxins, act as key processes conveying altered mineralization. In this work, we tested the ability of lanthionine, a novel uremic toxin, to promote calcification in human endothelial cell cultures (Ea.hy926). We evaluated the effects of lanthionine, at a concentration similar to that actually detected in CKD patients, alone and under pro-calcifying culture conditions using calcium and phosphate. In pro-calcific culture conditions, lanthionine increased both the intracellular and extracellular calcium content and induced the expression of Bone Morphogenetic Protein 2 (BMP2) and RUNX Family Transcription Factor 2 (RUNX2). Lanthionine treatment, in pro-calcifying conditions, raised levels of tissue-nonspecific alkaline phosphatase (ALPL), whose expression also overlapped with Dickkopf WNT Signaling Pathway Inhibitor 1 (DKK1) gene expression, suggesting a possible role of the latter gene in the activation of ALPL. In addition, treatment with lanthionine alone or in combination with calcium and phosphate reduced Inorganic Pyrophosphate Transport Regulator (ANKH) gene expression, a protective factor toward the mineralizing process. Moreover, lanthionine in a pro-calcifying condition induced the activation of ERK1/2, which is not associated with an increase in DKK1 protein levels. Our data underscored a link between mineral disease and the alterations of sulfur amino acid metabolisms at a cell and molecular level. These results set the basis for the understanding of the link between uremic toxins and mineral-bone disorder during CKD progression.
Collapse
Affiliation(s)
- Annapaola Coppola
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
| | - Carmela Vigorito
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Patrizia Lombari
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Yuselys García Martínez
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Margherita Borriello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
| | - Francesco Trepiccione
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
| | - Diego Ingrosso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (A.C.); (P.L.); (M.B.)
- Correspondence: (D.I.); (A.F.P.)
| | - Alessandra F. Perna
- Department of Translational Medical Science University of Campania “Luigi Vanvitelli”, Via Pansini, Bldg 17, 80131 Naples, Italy; (C.V.); (Y.G.M.); (F.T.)
- Correspondence: (D.I.); (A.F.P.)
| |
Collapse
|
8
|
Gender-Related Differences in BMP Expression and Adult Hippocampal Neurogenesis within Joint-Hippocampal Axis in a Rat Model of Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms222212163. [PMID: 34830044 PMCID: PMC8620092 DOI: 10.3390/ijms222212163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 11/16/2022] Open
Abstract
BMPs regulate synovial quiescence and adult neurogenesis in the hippocampus in non-stress conditions. However, changes in BMP expression that are induced by inflammation during rheumatoid arthritis (RA) have not yet been reported. Here, we show that signalling with synovial BMPs (BMP-4 and -7) mediates the effect of systemic inflammation on adult neurogenesis in the hippocampus during pristane-induced arthritis (PIA) in Dark Agouti (DA) rats, an animal model of RA. Moreover, we show gender differences in BMP expressions and their antagonists (Noggin and Gremlin) during PIA and their correlations with the clinical course and IL-17A and TNF-α levels in serum. Our results indicate gender differences in the clinical course, where male rats showed earlier onset and earlier recovery but a worse clinical course in the first two phases of the disease (onset and peak), which correlates with the initial increase of serum IL-17A level. The clinical course of the female rats worsened in remission. Their prolonged symptoms could be a reflection of an increased TNF-α level in serum during remission. Synovial inflammation was greater in females in PIA-remission with greater synovial BMP and antagonist expressions. More significant correlations between serum cytokines (IL-17A and TNF-α), and synovial BMPs and their antagonists were found in females than in males. On the other hand, males showed an increase in hippocampal BMP-4 expression during the acute phase, but both genders showed a decrease in antagonist expressions during PIA in general. Both genders showed a decrease in the number of Ki-67+ and SOX-2+ and DCX+ cells and in the ratio of DCX+ to Ki67+ cells in the dentate gyrus during PIA. However, in PIA remission, females showed a faster increase in the number of Ki67+, SOX-2+, and DCX+ cells and a faster increase in the DCX/Ki67 ratio than males. Both genders showed an increase of hippocampal BMP-7 expression during remission, although males constantly showed greater BMP-7 expression at all time points. Our data show that gender differences exist in the BMP expressions in the periphery-hippocampus axis and in the IL-17A and TNF-α levels in serum, which could imply differences in the mechanisms for the onset and progression of the disease, the clinical course severity, and adult neurogenesis with subsequent neurological complications between genders.
Collapse
|
9
|
Cai B, Du J. Role of bone morphogenic protein-4 in gestational diabetes mellitus-related hypertension. Exp Ther Med 2021; 22:762. [PMID: 34035859 DOI: 10.3892/etm.2021.10194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Hyperglycaemia stimulates the synthesis and release of bone morphogenetic protein-4 (BMP-4) in vascular endothelial cells, which further induces peroxide production and inflammatory responses, leading to vascular endothelial dysfunction. However, the role of BMP-4 in gestational diabetes mellitus (GDM)-related vascular endothelial dysfunction remains unclear. In the present study, the hypothesis that the overexpression of BMP-4 would induce GDM-related hypertension by impairing vascular endothelial function was evaluated. An animal model of GDM was established in Sprague-Dawley (SD) rats. Based on blood pressure, rats were divided into control, GDM and GDM + hypertension (HT) groups. The expression levels of BMP-4, cyclooxygenase-2 (COX-2), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 (NOX-1) and vascular cell adhesion molecule 1 (VCAM-1) in the endothelium of the abdominal aorta of rats in each group were determined via immunohistochemistry and western blotting. Pregnant SD rats were divided into four groups, separately infused with BMP-4, BMP-4 + noggin, noggin or vehicle by osmotic pumps, and blood pressure and vasorelaxation were examined. Immunohistochemistry indicated that the expression levels of the four proteins were lower in the control group than in the GDM and GDM + HT groups. The positive expression rate of VCAM-1 was significantly lower in the control group than in the GDM and GDM+HT groups, and the differences were statistically significant (χ2=17.325, P<0.05; χ2=10.080, P<0.05). Western blotting revealed that the expression level of the COX-2 protein exhibited a sequential increase in the three groups. The expression level of COX-2 in the control and GDM groups was significantly lower than that in the GDM+HT group (3.358±1.286; P<0.05 and P<0.05, respectively). The expression level of VCAM-1 protein in the three groups also exhibited a significant sequential increase (F=31.732; P≤0.001). The expression level of VCAM-1 in the control and GDM groups was significantly lower than that in the GDM+HT group (2.698±0.223; P≤0.001 and P≤0.001, respectively). Infusion of BMP-4 increased systolic blood pressure (from 82 to 112 mmHg) and impaired vasorelaxation in pregnant SD rats after 2 weeks. Co-treatment with noggin completely blocked BMP-4-induced effects. Thus, the BMP-4/NOX-1/COX-2 signalling pathway may be involved in GDM-related hypertension, but VCAM-1 may be substantially associated with GDM-related hypertension. Furthermore, overexpression of BMP-4 could lead to hypertension by impairing endothelial function in pregnancy.
Collapse
Affiliation(s)
- Benshuo Cai
- Department of Obstetrics and Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Juan Du
- Department of Obstetrics and Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
10
|
Tympanosclerosis and atherosclerosis plaques: a comparative analytical study on some new microbiological and immunohistochemical aspects. Eur Arch Otorhinolaryngol 2020; 278:3743-3752. [PMID: 33140144 DOI: 10.1007/s00405-020-06451-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/20/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE The aim of this study was to compare chemical contents, expression of BMP-8a, and the presence of Mycoplasma and ExoS-ExoU exotoxins producing Pseudomonas aeruginosa in tympanosclerosis (TS) and atherosclerosis (AS) plaques. METHODS Thirty-six cases with TS and AS plaques (18 each) were selected and examined for chemical, immunohistochemical, and microbial analysis. SPSS ver. 21 and t test analysis were used for comparing the findings, and the level of significance was considered as p < 0.05. RESULTS TS plaques showed lower carbon, higher calcium, and phosphorous contents compared to AS plaques (p value < 0.05). Chlorine was detected in AS plaques (1.8 w%) which could probably be due to the presence of myeloperoxidase (MPO) in atherosclerotic artery. Contrary to spherical shape of the surface of TS plaques, AS plaques were needle shaped. BMP-8a expression in TS plaques (59.5%) was significantly higher (p value < 0.0001) than AS plaques (20%). Of the 18 TS specimens, 12, 14, and 3 were positive for ExoS, ExoU Pseudomonas aeruginosa, and Mycoplasma genes, respectively, while of the 18 AS specimens, 2, 2, and 3 were positive for ExoS, ExoU Pseudomonas aeruginosa, and Mycoplasma genes, respectively. CONCLUSION TS plaques are different from AS plaques in terms of elemental components, surface morphology, and BMP-8a expression. Therefore, different calcification process and pathogenesis may be responsible for these two diseases. The results of our study showed that both TS and AS plaques have genetic footprint of Mycoplasma, but the level of calcium concentration-dependent exotoxins genes was found only in TS plaques.
Collapse
|
11
|
Arnold L, Weberbauer M, Herkel M, Fink K, Busch HJ, Diehl P, Grundmann S, Bode C, Elsässer A, Moser M, Helbing T. Endothelial BMP4 Promotes Leukocyte Rolling and Adhesion and Is Elevated in Patients After Survived Out-of-Hospital Cardiac Arrest. Inflammation 2020; 43:2379-2391. [PMID: 32803667 DOI: 10.1007/s10753-020-01307-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Leukocyte recruitment is a fundamental step in the inflammatory response during ischemia/reperfusion injury (IRI). Rolling and adhesion of leukocytes to activated endothelium promote tissue inflammation after IRI and require presentation of adhesion molecules E-selectin and ICAM-1 on the endothelial surface. Bone morphogenetic protein (BMP) 4 is a prominent member of the BMP family expressed and secreted by endothelial cells. BMP4 derived from endothelial cells has important functions in vascular disease but its influence on the leukocyte adhesion cascade during inflammation is incompletely understood. In the present study, we challenged mice with an inducible endothelial-specific BMP4 deletion (referred to as EC-BMP4-/- mice) and their control littermates (EC-BMP4+/+) with thioglycollate i.p. and assessed extravasation of different leukocyte subsets during peritonitis. Peritoneal lavages were performed and peritoneal cells were counted. Total cell count in lavages of EC-BMP4-/- mice was markedly reduced compared with lavages of EC-BMP4+/+ mice. FACS analyses of thioglycollate-elicited peritoneal cells revealed that diverse leukocyte subsets were reduced in EC-BMP4-/- mice. Intravital microscopy of cremaster venules demonstrated that rolling and adhesion of leukocytes were significantly diminished in EC-BMP4-/- mice in comparison with control mice in response to TNFα. These observations indicate that endothelial BMP4 is essential for rolling, adhesion, and extravasation of leukocytes in vivo. To understand the underlying mechanisms, levels of endothelial adhesion molecules E-selectin and ICAM-1 were quantified in EC-BMP4-/- and EC-BMP4+/+ mice by quantitative PCR and Western blotting. Interestingly, ICAM-1 and E-selectin expressions were reduced in the hearts of EC-BMP4-/- mice. Next we confirmed pro-inflammatory properties of BMP4 in a gain of function experiments and found that administration of recombinant BMP4 in male C57BL/6 mice increased leukocyte rolling and adhesion in cremaster venules in vivo. To assess the regulation of BMP4 in inflammatory disease in humans, we collected plasma samples of patients from day 0 to day 7 after survived out-of-hospital cardiac arrest (OHCA, n = 42). Remarkably, plasma of OHCA patients contained significantly higher BMP4 protein levels compared with patients with coronary artery disease (CAD, n = 12) or healthy volunteers (n = 11). Subgroup analysis revealed that elevated plasma BMP4 levels after ROSC are associated with decreased survival and unfavorable neurological outcome. Collectively, endothelial BMP4 is a potent activator of inflammation in vivo that promotes rolling, adhesion, and extravasation of leukocyte subsets by induction of E-selectin and ICAM-1. Elevation of plasma BMP4 levels in the post-resuscitation period suggests that BMP4 contributes to pathophysiology and poor outcome of post-cardiac arrest syndrome.
Collapse
Affiliation(s)
- Linus Arnold
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Miki Weberbauer
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marius Herkel
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katrin Fink
- Department of Emergency Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans-Jörg Busch
- Department of Emergency Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Diehl
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Grundmann
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Albrecht Elsässer
- Department of Cardiology, Heart Center Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
| | - Martin Moser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Helbing
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Department of Cardiology, Heart Center Oldenburg, Carl von Ossietzky University, Oldenburg, Germany.
| |
Collapse
|
12
|
Mitchell CM, Hirst JJ, Mitchell MD, Murray HG, Zakar T. Genes upregulated in the amnion at labour are bivalently marked by activating and repressive histone modifications. Mol Hum Reprod 2020; 25:228-240. [PMID: 30753586 DOI: 10.1093/molehr/gaz007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 01/17/2019] [Accepted: 02/08/2019] [Indexed: 12/19/2022] Open
Abstract
Inflammatory genes are expressed increasingly in the foetal membranes at late gestation triggering birth. Here we have examined whether epigenetic histone modifications contribute to the upregulation of proinflammatory genes in the amnion in late pregnancy and at labour. Amnion samples were collected from early pregnancy, at term in the absence of labour and after spontaneous birth. The expression of the labour-associated proinflammatory genes PTGS2, BMP2 and NAMPT was determined by reverse transcription-coupled quantitative real-time PCR (qRT-PCR). Chromatin immunoprecipitation (ChIP) and sequential double ChIP were performed to determine the levels and co-occurrence of activating histone-3, lysine-4 trimethylation (H3K4me3) and repressive histone-3, lysine-27 trimethylation (H3K27me3) at the gene promoters. H3K4 methyltransferase, H3K27me3 demethylase and H3K27 methyltransferase expression was determined by qRT-PCR and immunofluorescence confocal microscopy. PTGS2, BMP2 and NAMPT expression was upregulated robustly between early pregnancy and term (P < 0.05). The promoters were marked bivalently by both the H3K4me3 and H3K27me3 modifications. Bivalence was reduced at term by the decrease of the H3K27me3-modified fraction of promoter copies marked by H3K4me3 indicating epigenetic activation. Messenger RNAs encoding the H3K4-specific methyl transferases MLL1,-2,-3,-4, SETD1A,-B and the H3K27me3-specific demethylases KDM6A,-B were expressed increasingly while the H3K27 methyl transferase EZH2 was expressed decreasingly at term. Histone modifying enzyme proteins were detected in amnion epithelial and mesenchymal cells. These results with prototypical proinflammatory genes suggest that nucleosomes at labour-promoting genes are marked bivalently in the amnion, which is shifted towards monovalent H3K4me3 modification at term when the genes are upregulated. Bivalent epigenetic regulation by histone modifying enzymes may control the timing of labour.
Collapse
Affiliation(s)
- Carolyn M Mitchell
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia.,Faculty of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Jonathan J Hirst
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia.,Faculty of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Murray D Mitchell
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Henry G Murray
- Department of Obstetrics and Gynaecology, John Hunter Hospital, New Lambton Heights, New South Wales, Australia.,Faculty of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Tamas Zakar
- Mothers and Babies Research Centre, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia.,Department of Obstetrics and Gynaecology, John Hunter Hospital, New Lambton Heights, New South Wales, Australia.,Faculty of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| |
Collapse
|
13
|
Popov NA, Skulachev VP. Neotenic Traits in Heterocephalus glaber and Homo sapiens. BIOCHEMISTRY (MOSCOW) 2019; 84:1484-1489. [DOI: 10.1134/s0006297919120071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
14
|
Carmo LS, Burdmann EA, Fessel MR, Almeida YE, Pescatore LA, Farias-Silva E, Gamarra LF, Lopes GH, Aloia TPA, Liberman M. Expansive Vascular Remodeling and Increased Vascular Calcification Response to Cholecalciferol in a Murine Model of Obesity and Insulin Resistance. Arterioscler Thromb Vasc Biol 2019; 39:200-211. [PMID: 30580565 DOI: 10.1161/atvbaha.118.311880] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective- We hypothesized that ob/ob mice develop expansive vascular remodeling associated with calcification. Approach and Results- We quantified and investigated mechanisms of vascular remodeling and vascular calcification in ob/ob mice after vitamin D3(VD) stimulation or PBS (control), compared with C57BL/6 mice. Both ob/ob (OBVD [VD-treated ob/ob mice]) and C57BL/6 (C57VD [VD-treated C57BL/6 mice]) received 8×103 IU/day of intraperitoneal VD for 14 days. Control ob/ob (OBCT [PBS-treated ob/ob mice]) and C57BL/6 (C57CT [PBS-treated C57BL/6 mice]) received intraperitoneal PBS for 14 days. Hypervitaminosis D increased the external and internal elastic length in aortae from OBVD, resulting in increased total vascular area and lumen vascular area, respectively, which characterizes expansive vascular remodeling. OBVD decreased the aortic wall thickness, resulting in hypotrophic vascular remodeling. We demonstrated increased collagen deposition, elastolysis, and calcification in aortae from OBVD. Our results showed a positive correlation between expansive vascular remodeling and vascular calcification in OBVD. We demonstrated increased serum calcium levels, augmented Bmp (bone morphogenetic protein)-2 and osteochondrogenic proteins expression in OBVD aortae. Furthermore, aortae from OBVD increased oxidative stress, coincidently with augmented in situ MMP (matrix metalloproteinase) activity and exhibited no VDR (VD receptor) inhibition after VD. Conclusions- Our data provide evidence that obese and insulin-resistant mice (ob/ob) developed expansive hypotrophic vascular remodeling correlated directly with increased vascular calcification after chronic VD stimulation. Positive hypotrophic vascular remodeling and vascular calcification in this mouse model is possibly mediated by the convergence of absence VDR downregulation after VD stimulation, increased reactive oxygen species generation, and MMP activation.
Collapse
Affiliation(s)
- Luciana S Carmo
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil.,the Division of Nephrology, LIM 12, University of São Paulo Medical School, Brazil (L.S.C., E.A.B.)
| | - Emmanuel A Burdmann
- the Division of Nephrology, LIM 12, University of São Paulo Medical School, Brazil (L.S.C., E.A.B.)
| | - Melissa R Fessel
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Youri E Almeida
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Luciana A Pescatore
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Elisangela Farias-Silva
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Lionel F Gamarra
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Gabriel H Lopes
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Thiago P A Aloia
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Marcel Liberman
- From the Department of IIEP-Research and Teaching Institute (L.S.C., M.R.F., Y.E.A., L.A.P., E.F.-S., L.F.G., G.H.L., T.P.A.A., M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil.,Department of Critical Care Medicine and Cardiology (M.L.), Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
15
|
Sánchez-de-Diego C, Valer JA, Pimenta-Lopes C, Rosa JL, Ventura F. Interplay between BMPs and Reactive Oxygen Species in Cell Signaling and Pathology. Biomolecules 2019; 9:E534. [PMID: 31561501 PMCID: PMC6843432 DOI: 10.3390/biom9100534] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/12/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
The integration of cell extrinsic and intrinsic signals is required to maintain appropriate cell physiology and homeostasis. Bone morphogenetic proteins (BMPs) are cytokines that belong to the transforming growth factor-β (TGF-β) superfamily, which play a key role in embryogenesis, organogenesis and regulation of whole-body homeostasis. BMPs interact with membrane receptors that transduce information to the nucleus through SMAD-dependent and independent pathways, including PI3K-AKT and MAPKs. Reactive oxygen species (ROS) are intracellular molecules derived from the partial reduction of oxygen. ROS are highly reactive and govern cellular processes by their capacity to regulate signaling pathways (e.g., NF-κB, MAPKs, KEAP1-NRF2 and PI3K-AKT). Emerging evidence indicates that BMPs and ROS interplay in a number of ways. BMPs stimulate ROS production by inducing NOX expression, while ROS regulate the expression of several BMPs. Moreover, BMPs and ROS influence common signaling pathways, including PI3K/AKT and MAPK. Additionally, dysregulation of BMPs and ROS occurs in several pathologies, including vascular and musculoskeletal diseases, obesity, diabetes and kidney injury. Here, we review the current knowledge on the integration between BMP and ROS signals and its potential applications in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Cristina Sánchez-de-Diego
- Departament de Ciències Fisiològiques, Universitat de Barcelona, Carrer Feixa Llarga s/n, 08907 L'Hospitalet Llobregat, Spain.
| | - José Antonio Valer
- Departament de Ciències Fisiològiques, Universitat de Barcelona, Carrer Feixa Llarga s/n, 08907 L'Hospitalet Llobregat, Spain.
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, Carrer Feixa Llarga s/n, 08907 L'Hospitalet Llobregat, Spain.
| | - José Luis Rosa
- Departament de Ciències Fisiològiques, Universitat de Barcelona, Carrer Feixa Llarga s/n, 08907 L'Hospitalet Llobregat, Spain.
- IDIBELL, Avinguda Granvia de l'Hospitalet 199, 08908 L'Hospitalet de Llobregat, Spain.
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, Carrer Feixa Llarga s/n, 08907 L'Hospitalet Llobregat, Spain.
- IDIBELL, Avinguda Granvia de l'Hospitalet 199, 08908 L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
16
|
Pescatore LA, Gamarra LF, Liberman M. Multifaceted Mechanisms of Vascular Calcification in Aging. Arterioscler Thromb Vasc Biol 2019; 39:1307-1316. [DOI: https:/doi.org/10.1161/atvbaha.118.311576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 05/12/2019] [Indexed: 08/30/2023]
Abstract
Approximately 20% of the world’s population will be around or above 65 years of age by the next decade. Out of these, 40% are suspected to have cardiovascular diseases as a cause of mortality. Arteriosclerosis, characterized by increased vascular calcification, impairing Windkessel effect and tissue perfusion, and determining end-organ damage, is a hallmark of vascular pathology in the elderly population. Risk factors accumulated during aging affect the normal physiological and vascular aging process, which contributes to the progression of arteriosclerosis. Traditional risk factors, age-associated diseases, and respective regulating mechanisms influencing vascular calcification and vascular stiffness have been extensively studied for many years. Despite the well-known fact that aging alone can induce vascular damage, specific mechanisms that implicate physiological aging in vascular calcification, contributing to vascular stiffness, are poorly understood. This review focuses on mechanisms activated during normal aging, for example, cellular senescence, autophagy, extracellular vesicles secretion, and oxidative stress, along with the convergence of premature aging models’ pathophysiology, such as Hutchinson-Gilford Progeria (prelamin accumulation) and Klotho deficiency, to understand vascular calcification in aging. Understanding the mechanisms of vascular damage in aging that intersect with age-associated diseases and risk factors is crucial to foster innovative therapeutic targets to mitigate cardiovascular disease.
Visual Overview—
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Luciana A. Pescatore
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
- Laboratório de Biologia Vascular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brazil (L.A.P.)
| | - Lionel F. Gamarra
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| | - Marcel Liberman
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| |
Collapse
|
17
|
Pescatore LA, Gamarra LF, Liberman M. Multifaceted Mechanisms of Vascular Calcification in Aging. Arterioscler Thromb Vasc Biol 2019; 39:1307-1316. [PMID: 31144990 DOI: 10.1161/atvbaha.118.311576] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Approximately 20% of the world's population will be around or above 65 years of age by the next decade. Out of these, 40% are suspected to have cardiovascular diseases as a cause of mortality. Arteriosclerosis, characterized by increased vascular calcification, impairing Windkessel effect and tissue perfusion, and determining end-organ damage, is a hallmark of vascular pathology in the elderly population. Risk factors accumulated during aging affect the normal physiological and vascular aging process, which contributes to the progression of arteriosclerosis. Traditional risk factors, age-associated diseases, and respective regulating mechanisms influencing vascular calcification and vascular stiffness have been extensively studied for many years. Despite the well-known fact that aging alone can induce vascular damage, specific mechanisms that implicate physiological aging in vascular calcification, contributing to vascular stiffness, are poorly understood. This review focuses on mechanisms activated during normal aging, for example, cellular senescence, autophagy, extracellular vesicles secretion, and oxidative stress, along with the convergence of premature aging models' pathophysiology, such as Hutchinson-Gilford Progeria (prelamin accumulation) and Klotho deficiency, to understand vascular calcification in aging. Understanding the mechanisms of vascular damage in aging that intersect with age-associated diseases and risk factors is crucial to foster innovative therapeutic targets to mitigate cardiovascular disease. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Luciana A Pescatore
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.).,Laboratório de Biologia Vascular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brazil (L.A.P.)
| | - Lionel F Gamarra
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| | - Marcel Liberman
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| |
Collapse
|
18
|
Lian D, Yuan H, Yin X, Wu Y, He R, Huang Y, Chen Y. Puerarin inhibits hyperglycemia-induced inter-endothelial junction through suppressing endothelial Nlrp3 inflammasome activation via ROS-dependent oxidative pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 55:310-319. [PMID: 30385134 DOI: 10.1016/j.phymed.2018.10.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 10/04/2018] [Accepted: 10/09/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Recent studies indicate that vascular complications are closely related to diabetes mellitus; in particular, inflammatory-mediated endothelial dysfunction plays a crucial role in diabetes-induced cardiovascular diseases. Therefore, exploring effective methods to suppress endothelial dysfunction via inhibition of inflammatory responses is imperative. Puerarin (Pu), a flavonoid common in Pueraria, has been widely and successfully used to treat cardiovascular diseases in China for many years. However, information on its protective properties in hyperglycemia-induced vascular complications is insufficient. Hypothesis/Purpose: In this study, we investigate the protective effects of puerarin against high glucose-induced endothelial dysfunction and the underlying mechanism of the flavonoid. METHODS we investigated the protective effects of Pu against hyperglycemia-induced inter-endothelial junction by permeability and transendothelial electrical resistance (TEER) assay. In addition, changes in the Nlrp3 inflammasome activation via reactive oxygen species (ROS)-dependent oxidative pathway were investigated using western blot, immunofluorescence microscopy analyses and flow cytometry. ROS scavenger and Nlrp3 gene silencing were used to determine the roles of the ROS-Nlrp3 pathway involved in the molecular mechanism of Pu. RESULTS Our findings demonstrate that puerarin inhibits high glucose-induced Nlrp3 inflammasome formation and activation, as shown by fluorescence confocal microscopy and Western blot. Puerarin decreases Nlrp3 protein, which is a critical factor necessary to form an inflammasome complex. We demonstrate that puerarin exerts anti-oxidation and ROS scavenged effects, similar to apocynin (APO). Interestingly, thioredoxin-interacting protein (TXNIP) protein and TXNIP binding to Nlrp3 markedly decreased with puerarin treatment. Together with these changes, puerarin could decrease high mobility group box 1 (HMGB1) release from mouse vascular endothelial cell (mMVECs). We also demonstrate the decreased expression of the tight junction proteins ZO-1/ZO-2, which are related to endothelial permeability after stimulation by high glucose in endothelial cells. Puerarin could recover the gap junction protein and decrease monolayer cell permeability in endothelial cells. In conclusion, we reveal a new protection mechanism of puerarin that inhibits Nlrp3 inflammasome activation and decreases subsequent caspase-1 activation, triggering the release of HMGB1 by reducing ROS generation. CONCLUSIONS Our findings indicate that puerarin exhibits immense potential and specific therapeutic value in hyperglycemia-related cardiovascular disease and the development of innovative drugs.
Collapse
Affiliation(s)
- Dawei Lian
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, No. 232 Waihuan Dong Rd., Panyu District, Guangzhou Higher Education Mega Center, Guangzhou 510000, China
| | - Huiqi Yuan
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, No. 232 Waihuan Dong Rd., Panyu District, Guangzhou Higher Education Mega Center, Guangzhou 510000, China
| | - Xiongzhang Yin
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yanjiao Wu
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, No. 232 Waihuan Dong Rd., Panyu District, Guangzhou Higher Education Mega Center, Guangzhou 510000, China
| | - Rongrong He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, No. 601W. Huangpu Avenue, Guangzhou 510630, China
| | - Yi Huang
- Department of Stomatology, The First Affiliated Hospital, The school of Dental Medicine, Jinan University, No. 613W. Huangpu Avenue, Guangzhou 510630, China
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, No. 232 Waihuan Dong Rd., Panyu District, Guangzhou Higher Education Mega Center, Guangzhou 510000, China.
| |
Collapse
|
19
|
Peiffer BJ, Qi L, Ahmadi AR, Wang Y, Guo Z, Peng H, Sun Z, Liu JO. Activation of BMP Signaling by FKBP12 Ligands Synergizes with Inhibition of CXCR4 to Accelerate Wound Healing. Cell Chem Biol 2019; 26:652-661.e4. [PMID: 30827938 DOI: 10.1016/j.chembiol.2019.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/04/2018] [Accepted: 01/24/2019] [Indexed: 01/05/2023]
Abstract
The combination of AMD3100 and low-dose FK506 has been shown to accelerate wound healing in vivo. Although AMD3100 is known to work by releasing hematopoietic stem cells into circulation, the mechanism of FK506 in this setting has remained unknown. In this study, we investigated the activities of FK506 in human cells and a diabetic-rat wound model using a non-immunosuppressive FK506 analog named FKVP. While FKVP was incapable of inhibiting calcineurin, wound-healing enhancement with AMD3100 was unaffected. Further study showed that both FK506 and FKVP activate BMP signaling in multiple cell types through FKBP12 antagonism. Furthermore, selective inhibition of BMP signaling abolished stem cell recruitment and wound-healing enhancement by combination treatment. These results shed new light on the mechanism of action of FK506 in acceleration of wound healing, and raise the possibility that less toxic FKBP ligands such as FKVP can replace FK506 for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Brandon J Peiffer
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Le Qi
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ali R Ahmadi
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Yuefan Wang
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Zufeng Guo
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Hanjing Peng
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| | - Jun O Liu
- Department of Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; The SJ Yan and HJ Mao Laboratory of Chemical Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
20
|
Abstract
Hepcidin, the main regulator of iron metabolism, is synthesized and released by hepatocytes in response to increased body iron concentration and inflammation. Deregulation of hepcidin expression is a common feature of genetic and acquired iron disorders: in Hereditary Hemochromatosis (HH) and iron-loading anemias low hepcidin causes iron overload, while in Iron Refractory Iron Deficiency Anemia (IRIDA) and anemia of inflammation (AI), high hepcidin levels induce iron-restricted erythropoiesis. Hepcidin expression in the liver is mainly controlled by the BMP-SMAD pathway, activated in a paracrine manner by BMP2 and BMP6 produced by liver sinusoidal endothelial cells. The BMP type I receptors ALK2 and ALK3 are responsible for iron-dependent hepcidin upregulation and basal hepcidin expression, respectively. Characterization of animal models with genetic inactivation of the key components of the pathway has suggested the existence of two BMP/SMAD pathway branches: the first ALK3 and HH proteins dependent, responsive to BMP2 for basal hepcidin activation, and the second ALK2 dependent, activated by BMP6 in response to increased tissue iron. The erythroid inhibitor of hepcidin Erythroferrone also impacts on the liver BMP-SMAD pathway although its effect is blunted by pathway hyper-activation. The liver BMP-SMAD pathway is required also in inflammation to cooperate with JAK2/STAT3 signaling for full hepcidin activation. Pharmacologic targeting of BMP-SMAD pathway components or regulators may improve the outcome of both genetic and acquired disorders of iron overload and deficiency by increasing or inhibiting hepcidin expression.
Collapse
|
21
|
Xuan Y, Wang W, Zhang H, Tan I, Butlin M, Avolio A, Zuo J. Osteoporosis is inversely associated with arterial stiffness in the elderly: An investigation using the Osteoporosis Self-assessment Tool for Asians index in an elderly Chinese cohort. J Clin Hypertens (Greenwich) 2019; 21:405-411. [PMID: 30734463 DOI: 10.1111/jch.13493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022]
Abstract
Although the association of arterial stiffness and osteoporosis has been reported, the relation of arterial stiffness with risk of osteoporosis and bone fracture is not established. The authors investigated the correlation between arterial stiffness (brachial-ankle pulse wave velocity [baPWV]), including a cutoff value, and risk of osteoporosis as assessed by the Osteoporosis Self-assessment Tool for Asia (OSTA) index in 129 elderly Chinese community-dwelling individuals (age 83.2 ± 12.8 years, 63 females). OSTA was negatively correlated with baPWV (r = -0.326, P = 0.023) after adjusting for confounding factors such as gender, body mass index, low-density lipoprotein, triglycerides, estimated glomerular filtration rate, absence or presence of diabetes, absence or presence of hypertension, and uric acid. baPWV was an independent factor for changes in OSTA (β = -0.001, P = 0.002). ROC curve analysis confirmed association between baPWV and OSTA index (AUC = 0.742 [CI: 0.660, 0.824]; P < 0.001) with a baPWV cutoff value of 1676 cm/s (sensitivity, 80.7%; specificity, 60%) for prediction of high OSTA index. The study showed a significant correlation between OSTA index and baPWV, suggesting a potential predictive value of baPWV in elderly patient at high risk of osteoporosis.
Collapse
Affiliation(s)
- Yan Xuan
- Department of Geriatric Medicine, Shanghai Jiatong School of Medicine, Ruijin Hospital North, Shanghai, China
| | - Weiliang Wang
- Department of Neurology Medicine, Xuhui District Central Hospital, Shanghai, China
| | - Hong Zhang
- Department of Geriatric Medicine, Shanghai Jiatong School of Medicine, Ruijin Hospital North, Shanghai, China
| | - Isabella Tan
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Mark Butlin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Alberto Avolio
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Junli Zuo
- Department of Geriatric Medicine, Shanghai Jiatong School of Medicine, Ruijin Hospital North, Shanghai, China
| |
Collapse
|
22
|
Wang K, Li H, Sun R, Liu C, Luo Y, Fu S, Ying Y. Emerging roles of transforming growth factor β signaling in wet age-related macular degeneration. Acta Biochim Biophys Sin (Shanghai) 2019; 51:1-8. [PMID: 30496406 DOI: 10.1093/abbs/gmy145] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 10/31/2018] [Indexed: 12/25/2022] Open
Abstract
Age-related macular degeneration (AMD) is one of the major causes of irreversible blindness among aging populations in developed countries and can be classified as dry or wet according to its progression. Wet AMD, which is characterized by angiogenesis on the choroidal membrane, is uncommonly seen but more severe. Controlling or completely inhibiting the factors that contribute to the progression of events that lead to angiogenesis may be an effective strategy for treating wet AMD. Emerging evidence has shown that transforming growth factor-β (TGF-β) signaling plays a significant role in the progression of wet AMD. In this review, we described the roles of and changes in TGF-β signaling in the development of AMD and discussed the mechanisms of the TGF-β superfamily in choroidal neovascularization (CNV) and wet AMD, including the modulation of angiogenesis-related factors, inflammation, vascular fibrosis, and immune responses, as well as cross-talk with other signaling pathways. These remarkable findings indicate that TGF-β signaling is a potential target for wet AMD treatment.
Collapse
Affiliation(s)
- Kai Wang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China
- Nanchang Joint Program, Queen Mary University of London, London, UK
| | - Haoran Li
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China
- Nanchang Joint Program, Queen Mary University of London, London, UK
| | - Ruipu Sun
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China
- Nanchang Joint Program, Queen Mary University of London, London, UK
| | - Chaxian Liu
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China
- The Second Clinical Department, School of Medicine, Nanchang University, Nanchang, China
| | - Yunfei Luo
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China
- Department of Pathophysiology, School of Medicine, Nanchang University, Nanchang, China
| | - Shuhua Fu
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathology, Schools of Basic Medical Sciences and Pharmaceutical Sciences, Nanchang University Medical College, Nanchang, China
- Department of Pathophysiology, School of Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
23
|
Cao Y, Drake M, Davis J, Yang B, Ko TC. Opposing Roles of BMP and TGF-β Signaling Pathways in Pancreatitis: Mechanisms and Therapeutic Implication. ADVANCED RESEARCH IN GASTROENTEROLOGY & HEPATOLOGY 2019; 13:555871. [PMID: 32211568 PMCID: PMC7093075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Bone morphogenetic proteins (BMPs) comprise a major subgroup of the transforming growth factor (TGF)-β superfamily. They play pivotal roles in embryonic development and tissue homeostasis in adults. Deregulation of BMP and TGF-β signaling contributes to developmental anomalies and multiple diseases. In this mini-review, we focus on BMP signaling in inflammatory disorders of the pancreas, acute and chronic pancreatitis, in contrast to TGF-β signaling. We then discuss molecular mechanisms that interact with and connect between the BMP and TGF-β signaling pathways. Lastly, we review potential implications of these molecular mechanisms for therapeutic development. In summary, BMP signaling pathway plays different roles during pancreatitis disease development, and the antagonism between BMP and TGF-β signaling can be manipulated for therapeutic development against pancreatitis.
Collapse
Affiliation(s)
- Yanna Cao
- Corresponding author: Yanna Cao, Department of Surgery, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
24
|
Zhao M, Ko SY, Garrett IR, Mundy GR, Gutierrez GE, Edwards JR. The polyphenol resveratrol promotes skeletal growth in mice through a sirtuin 1-bone morphogenic protein 2 longevity axis. Br J Pharmacol 2018; 175:4183-4192. [PMID: 30125963 PMCID: PMC6177622 DOI: 10.1111/bph.14477] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 07/16/2018] [Accepted: 07/19/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The polyphenol resveratrol (RSV) exists in high quantities in certain foods (e.g. grapes and nuts). However, the capacity of RSV to confer physiological health benefits and a biological mechanism through which this might occur remains unclear. EXPERIMENTAL APPROACH Aged, RSV-treated (300 mg·kg-1 ·day-1 ) and genetically modified [endothelial NOS (eNOS-/- )] female mice were assessed using histomorphometric and μCT analysis. Alongside in vivo analysis, molecular siRNA knockdown and pharmacological manipulation of eNOS, BMP2 and sirtuin 1 (SIRT1) and functional cellular assays in an osteoblast cell line panel, explored the mechanism through which RSV might impact overall bone volume. KEY RESULTS RSV promoted osteoblast activity and bone growth in vivo. RSV dose-dependently and simultaneously increased alkaline phosphatase (ALP) and eNOS levels. Similarly, NO-donor treatment increased ALP, runt homology transcription factor 2, BMP2 and stimulated bone formation, whilst eNOS-deficient mice displayed a bone loss phenotype. Moreover, RSV-induced increase in ALP and BMP2 expression was blocked in eNOS-/- osteoblasts and by BMP-inhibitor noggin. The longevity-linked SIRT1 enzyme was positively regulated by RSV and SIRT1 deletion reduced eNOS, BMP2 and ALP. Like eNOS deletion, loss of SIRT1 blocked RSV-induced osteoblast activity; however, SIRT1 levels remained unchanged in eNOS-/- mice, indicating RSV activation of SIRT1 stimulates BMP2 release via eNOS. This signalling axis is supported by decreased SIRT1, eNOS and BMP2 confirmed in old versus young bone. CONCLUSIONS AND IMPLICATIONS These findings suggest a new mechanism of action in bone remodelling and the ageing skeleton, where RSV positively impacts bone homeostasis via SIRT1 activation of BMP2.
Collapse
Affiliation(s)
- Ming Zhao
- School of Medicine, Tulane University, New Orleans, LA, USA
| | - Seon-Yle Ko
- School of Dentistry, Dankook University, Cheonan, Korea
| | - I Ross Garrett
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio and OsteoScreen Inc., San Antonio, TX, USA
| | - Gregory R Mundy
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio and OsteoScreen Inc., San Antonio, TX, USA
| | - Gloria E Gutierrez
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio and OsteoScreen Inc., San Antonio, TX, USA
| | - James R Edwards
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Pardali E, Makowski LM, Leffers M, Borgscheiper A, Waltenberger J. BMP-2 induces human mononuclear cell chemotaxis and adhesion and modulates monocyte-to-macrophage differentiation. J Cell Mol Med 2018; 22:5429-5438. [PMID: 30102472 PMCID: PMC6201342 DOI: 10.1111/jcmm.13814] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/07/2018] [Accepted: 06/29/2018] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a cardiovascular risk factor which leads to atherosclerosis, an inflammatory disease characterized by the infiltration of mononuclear cells in the vessel. Bone morphogenetic protein (BMP)‐2 is a cytokine which has been recently shown to be elevated in atherosclerosis and T2DM and to contribute to vascular inflammation. However, the role of BMP‐2 in the regulation of mononuclear cell function remains to be established. Herein, we demonstrate that BMP‐2 induced human monocyte chemotaxis via phosphoinositide 3 kinase and mitogen‐activated protein kinases. Inhibition of endogenous BMP‐2 signalling, by Noggin or a BMP receptor inhibitor, interfered with monocyte migration. Although BMP‐2 expression was increased in monocytes from T2DM patients, it could still stimulate their migration. Furthermore, BMP‐2 interfered with their differentiation into M2 macrophages. Finally, BMP‐2 both induced the adhesion of monocytes to fibronectin and endothelial cells (ECs), and promoted the adhesive properties of ECs, by increasing expression of adhesion and pro‐inflammatory molecules. Our data demonstrate that BMP‐2 could exert its pro‐inflammatory effects by inducing monocyte migration and adhesiveness to ECs and by interfering with the monocyte differentiation into M2 macrophages. Our findings provide novel insights into the mechanisms by which BMP‐2 may contribute to the development of atherosclerosis.
Collapse
Affiliation(s)
- Evangelia Pardali
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Lena-Maria Makowski
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Merle Leffers
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Andreas Borgscheiper
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| |
Collapse
|
26
|
Helbing T, Arnold L, Wiltgen G, Hirschbihl E, Gabelmann V, Hornstein A, Esser JS, Diehl P, Grundmann S, Busch HJ, Fink K, Bode C, Moser M. Endothelial BMP4 Regulates Leukocyte Diapedesis and Promotes Inflammation. Inflammation 2018; 40:1862-1874. [PMID: 28755278 DOI: 10.1007/s10753-017-0627-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Leukocyte recruitment is a fundamental event in the response of the innate immune system to injury. This process is promoted in part by the opening of endothelial cell adherens junctions that allows leukocyte extravasation through gaps between adjacent endothelial cells. VE-cadherin is a key component of endothelial cell adherens junctions and a negative regulator of leukocyte emigration. Accumulating evidence implicates bone morphogenetic protein (BMP) 4 as a critical regulator in vascular biology, but its role in leukocyte extravasation in vitro and in vivo has not been investigated so far. To assess the impact of BMP4 on leukocyte emigration in vivo, we used the thioglycollate-induced peritonitis model. C57BL/6 mice were intraperitoneally (i.p.) injected with recombinant BMP4 in addition to thioglycollate. Compared to solvent-treated controls, we observed higher accumulation of leukocytes in the peritoneal lavage of BMP4-treated mice indicating that BMP4 promotes leukocyte diapedesis into the inflamed peritoneal cavity. Endothelial cell-specific deletion of BMP4 in mice markedly diminished leukocyte diapedesis following thioglycollate administration suggesting that endothelial BMP4 is required for leukocyte recruitment. Consistent with these in vivo results, transwell migration assays with human umbilical vein endothelial cells (HUVECs) in vitro revealed that recombinant BMP4 enhanced leukocyte transmigration through the endothelial monolayer. Conversely, silencing of endothelial BMP4 by siRNA dampened leukocyte diapedesis in vitro. Mechanistic studies showed that loss of BMP4 improved endothelial junction stability by upregulation of VE-cadherin expression in vitro and in vivo. Vice versa, treatment of HUVECs with recombinant BMP4 decreased expression of VE-cadherin and impaired endothelial junction stability shown by Western blotting and immunocytochemistry. Finally, severe endothelial damage in HUVECs in response to serum of patients collected 24 h after survived cardiac arrest was accompanied by increase in leukocyte migration in transwell assays and activation of the BMP pathway most probably by upregulation of endothelial BMP4 RNA and protein expression. Collectively, the present study provides novel evidence that endothelial BMP4 controls leukocyte recruitment through a VE-cadherin-dependent mechanism and that BMP4-induced inflammation might be involved in the pathogenesis of endothelial cell damage following successful resuscitation after cardiac arrest.
Collapse
Affiliation(s)
- Thomas Helbing
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany.
| | - Linus Arnold
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Gwendoline Wiltgen
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Eva Hirschbihl
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Valentin Gabelmann
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Alexandra Hornstein
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Jennifer S Esser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Philipp Diehl
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Sebastian Grundmann
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Hans-Jörg Busch
- Department of Emergency Medicine, University Hospital of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katrin Fink
- Department of Emergency Medicine, University Hospital of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| | - Martin Moser
- Department of Cardiology and Angiology I, Heart Center Freiburg University, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
| |
Collapse
|
27
|
Yang J, Xu T, Gomez DR, Yuan X, Nguyen QN, Jeter M, Song Y, Hahn S, Liao Z. Polymorphisms in BMP2/BMP4, with estimates of mean lung dose, predict radiation pneumonitis among patients receiving definitive radiotherapy for non-small cell lung cancer. Oncotarget 2018; 8:43080-43090. [PMID: 28574846 PMCID: PMC5522129 DOI: 10.18632/oncotarget.17904] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/07/2017] [Indexed: 12/17/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) in TGFβ1 can predict the risk of radiation pneumonitis (RP) in patients with non-small cell lung cancer (NSCLC) after definitive radiotherapy. Here we investigated whether SNPs in TGFβ superfamily members BMP2 and BMP4 are associated with RP in such patients. In total, we retrospectively analyzed 663 patients given ≥ 60 Gy for NSCLC. We randomly assigned 323 patients to the training cohort and 340 patients to the validation cohort. Potentially functional and tagging SNPs of BMP2 (rs170986, rs1979855, rs1980499, rs235768, rs3178250) and BMP4 (rs17563, rs4898820, rs762642) were genotyped. The median of mean lung dose (MLD) was 17.9 Gy (range, 0.15–32.74 Gy). Higher MLD was strongly associated with increased risk of grade ≥ 2 RP (hazard ratio [HR]=2.191, 95% confidence interval [CI] = 1.680–2.856, P < 0.001) and grade ≥ 3 RP (HR = 4.253, 95% CI = 2.493–7.257, P < 0.001). In multivariate analyses, BMP2 rs235768 AT/TT was associated with higher risk of grade ≥ 2 RP (HR = 1.866, 95% CI = 1.221–2.820, P = 0.004 vs. AA) both in training cohort and validation cohort. Similar results were observed for BMP2 rs1980499. BMP2 rs3178250 CT/TT was associated with lower risk of grade ≥ 3 RP (HR = 0.406, 95% CI = 0.175–0.942, P = 0.036 vs. CC) in the pooled analysis. Adding the rs235768 and rs1980499 SNPs to a model comprising age, performance status, and MLD raised the Harrell's C for predicting grade ≥ 2 RP from 0.6117 to 0.6235 (P = 0.0105). SNPs in BMP2 can predict grade ≥ 2 or 3 RP after radiotherapy for NSCLC and improve the predictive power of MLD model. Validation is underway through an ongoing prospective trial.
Collapse
Affiliation(s)
- Ju Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China
| | - Ting Xu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Daniel R Gomez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Melenda Jeter
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yipeng Song
- Department of Radiation Oncology, Yuhuangding Hospital, Shandong, 264000, China
| | - Stephen Hahn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhongxing Liao
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
28
|
Choy KW, Lau YS, Murugan D, Vanhoutte PM, Mustafa MR. Paeonol Attenuates LPS-Induced Endothelial Dysfunction and Apoptosis by Inhibiting BMP4 and TLR4 Signaling Simultaneously but Independently. J Pharmacol Exp Ther 2017; 364:420-432. [DOI: 10.1124/jpet.117.245217] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
|
29
|
BMP-2 and icariin synergistically promote p38MAPK-mediated cardiomyocyte differentiation of mesenchymal stem cells via enhanced NOX4-driven ROS generation. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1954-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
30
|
Olsavszky V, Ulbrich F, Singh S, Diett M, Sticht C, Schmid CD, Zierow J, Wohlfeil SA, Schledzewski K, Dooley S, Gaitantzi H, Breitkopf-Heinlein K, Géraud C, Goerdt S, Koch PS. GATA4 and LMO3 balance angiocrine signaling and autocrine inflammatory activation by BMP2 in liver sinusoidal endothelial cells. Gene 2017; 627:491-499. [DOI: 10.1016/j.gene.2017.06.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/12/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022]
|
31
|
Yao J, Guihard PJ, Wu X, Blazquez-Medela AM, Spencer MJ, Jumabay M, Tontonoz P, Fogelman AM, Boström KI, Yao Y. Vascular endothelium plays a key role in directing pulmonary epithelial cell differentiation. J Cell Biol 2017; 216:3369-3385. [PMID: 28838957 PMCID: PMC5626536 DOI: 10.1083/jcb.201612122] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/26/2017] [Accepted: 07/12/2017] [Indexed: 12/19/2022] Open
Abstract
J. Yao et al. demonstrate that loss of MGP, a BMP inhibitor, causes abnormal hepatic differentiation in lungs. They find that interactions between endothelium and epithelium separate pulmonary from hepatic differentiation during development. Lack of MGP triggers hepatic differentiation in the pulmonary epithelium, as regulated by the endothelium. The vascular endothelium is critical for induction of appropriate lineage differentiation in organogenesis. In this study, we report that dysfunctional pulmonary endothelium, resulting from the loss of matrix Gla protein (MGP), causes ectopic hepatic differentiation in the pulmonary epithelium. We demonstrate uncontrolled induction of the hepatic growth factor (HGF) caused by dysregulated cross talk between pulmonary endothelium and epithelium in Mgp-null lungs. Elevated HGF induced hepatocyte nuclear factor 4 α (Hnf4a), which competed with NK2 homeobox 1 (Nkx2.1) for binding to forkhead box A2 (Foxa2) to drive hepatic differentiation in Mgp-null airway progenitor cells. Limiting endothelial HGF reduced Hnf4a, abolished interference of Hnf4a with Foxa2, and reduced hepatic differentiation in Mgp-null lungs. Together, our results suggest that endothelial–epithelial interactions, maintained by MGP, are essential in pulmonary cell differentiation.
Collapse
Affiliation(s)
- Jiayi Yao
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Pierre J Guihard
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Ana M Blazquez-Medela
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Melissa J Spencer
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Medet Jumabay
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Department of Pathology and Laboratory Medicine, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Kristina I Boström
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA .,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA .,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
32
|
Mitrofan CG, Appleby SL, Nash GB, Mallat Z, Chilvers ER, Upton PD, Morrell NW. Bone morphogenetic protein 9 (BMP9) and BMP10 enhance tumor necrosis factor-α-induced monocyte recruitment to the vascular endothelium mainly via activin receptor-like kinase 2. J Biol Chem 2017. [PMID: 28646109 PMCID: PMC5566526 DOI: 10.1074/jbc.m117.778506] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bone morphogenetic proteins 9 and 10 (BMP9/BMP10) are circulating cytokines with important roles in endothelial homeostasis. The aim of this study was to investigate the roles of BMP9 and BMP10 in mediating monocyte-endothelial interactions using an in vitro flow adhesion assay. Herein, we report that whereas BMP9/BMP10 alone had no effect on monocyte recruitment, at higher concentrations both cytokines synergized with tumor necrosis factor-α (TNFα) to increase recruitment to the vascular endothelium. The BMP9/BMP10-mediated increase in monocyte recruitment in the presence of TNFα was associated with up-regulated expression levels of E-selectin, vascular cell adhesion molecule (VCAM-1), and intercellular adhesion molecule 1 (ICAM-1) on endothelial cells. Using siRNAs to type I and II BMP receptors and the signaling intermediaries (Smads), we demonstrated a key role for ALK2 in the BMP9/BMP10-induced surface expression of E-selectin, and both ALK1 and ALK2 in the up-regulation of VCAM-1 and ICAM-1. The type II receptors, BMPR-II and ACTR-IIA were both required for this response, as was Smad1/5. The up-regulation of cell surface adhesion molecules by BMP9/10 in the presence of TNFα was inhibited by LDN193189, which inhibits ALK2 but not ALK1. Furthermore, LDN193189 inhibited monocyte recruitment induced by TNFα and BMP9/10. BMP9/10 increased basal IκBα protein expression, but did not alter p65/RelA levels. Our findings suggest that higher concentrations of BMP9/BMP10 synergize with TNFα to induce the up-regulation of endothelial selectins and adhesion molecules, ultimately resulting in increased monocyte recruitment to the vascular endothelium. This process is mediated mainly via the ALK2 type I receptor, BMPR-II/ACTR-IIA type II receptors, and downstream Smad1/5 signaling.
Collapse
Affiliation(s)
- Claudia-Gabriela Mitrofan
- From the Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ and
| | - Sarah L Appleby
- From the Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ and
| | - Gerard B Nash
- the Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Ziad Mallat
- From the Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ and
| | - Edwin R Chilvers
- From the Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ and
| | - Paul D Upton
- From the Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ and
| | - Nicholas W Morrell
- From the Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ and
| |
Collapse
|
33
|
Liu C, Gao J, Chen B, Chen L, Belguise K, Yu W, Lu K, Wang X, Yi B. Cyclooxygenase-2 promotes pulmonary intravascular macrophage accumulation by exacerbating BMP signaling in rat experimental hepatopulmonary syndrome. Biochem Pharmacol 2017. [PMID: 28642034 DOI: 10.1016/j.bcp.2017.06.117] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS One central factor in hepatopulmonary syndrome (HPS) pathogenesis is intravascular accumulation of activated macrophages in small pulmonary arteries. However, molecular mechanism underlying the macrophage accumulation in HPS is unknown. In this study, we aimed to explore whether elevated COX-2 induces the Bone morphogenic protein-2 (BMP-2)/Crossveinless-2 (CV-2) imbalance and then activation of BMP signaling pathway promotes the macrophage accumulation in Common Bile Duct Ligation (CBDL) rat lung. METHODS The COX-2/PGE2 signaling activation, the BMP-2/CV-2 imbalance and the activation of Smad1 were evaluated in CBDL rat lung and in cultured pulmonary microvascular endothelial cells (PMVECs) under the HPS serum stimulation. The effects of Parecoxib (COX-2 inhibitor), BMP-2 and CV-2 recombinant proteins on 4-week CBDL rat lung were determined, respectively. RESULTS The COX-2/PGE2 signaling pathway was activated in CBDL rat lung in vivo and PMVECs in vitro, which was due to the activation of NF-κB P65. The inhibition of COX-2 by Parecoxib reduced macrophage accumulation, decreased lung angiogenesis and improved HPS. Meanwhile, the CBDL rat lung secreted more BMP-2 but less CV-2, and the imbalance between BMP-2 and CV-2 exacerbated the BMP signaling activation thus promoting the macrophage accumulation and lung angiogenesis. The BMP-2/CV-2 imbalance is dependent on the COX-2/PGE2 signaling pathway, and thus the effects of this imbalance can be reversed by adminstration of Parecoxib. CONCLUSION Our findings indicate that inhibition of COX-2 by parecoxib can improve the HPS through the repression of BMP signaling and macrophage accumulation.
Collapse
Affiliation(s)
- Chang Liu
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Jing Gao
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Bing Chen
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China; LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Lin Chen
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Karine Belguise
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Weifeng Yu
- Department of Anesthesia, RenJi Hospital, The Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Kaizhi Lu
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Xiaobo Wang
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China; LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France.
| | - Bin Yi
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
34
|
Helbing T, Wiltgen G, Hornstein A, Brauers EZ, Arnold L, Bauer A, Esser JS, Diehl P, Grundmann S, Fink K, Patterson C, Bode C, Moser M. Bone Morphogenetic Protein-Modulator BMPER Regulates Endothelial Barrier Function. Inflammation 2017; 40:442-453. [PMID: 27995357 DOI: 10.1007/s10753-016-0490-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The endothelium serves as a selective barrier and controls the exchange of nutrients, hormones, and leukocytes between blood and tissues. Molecular mechanisms contributing to the pathogenesis of endothelial barrier dysfunction remain incompletely understood. Accumulating evidence implicates bone morphogenetic protein (BMP)-modulator BMPER as a key regulator in endothelial biology. Herein, we analyze the impact of BMPER in the control of endothelial barrier function. To assess the role of BMPER in vascular barrier function in mice, we measured the leakage of Evans blue dye from blood into interstitial lung tissue. BMPER+/- mice exhibited a significantly higher degree of vascular leak compared with wild-type siblings. In accordance with our in vivo observation, siRNA-based BMPER knockdown in human umbilical endothelial cells increased endothelial permeability measured by FITC-dextran passage in transwell assays. Mechanistically, BMPER knockdown reduced the expression of VE-cadherin, a pivotal component of endothelial adherens junctions. Conversely, recombinant human BMPER protein upregulated VE-cadherin protein levels and improved endothelial barrier function in transwell assays. The effects of BMPER knockdown on VE-cadherin expression and endothelial permeability were induced by enhanced BMP activity. Supporting this notion, activation of BMP4-Smad-Id1 signaling reduced VE-cadherin levels and impaired endothelial barrier function in vitro. In vivo, Evans blue dye accumulation was higher in the lungs of BMP4-treated C57BL/6 mice compared to controls indicating that BMP4 increased vascular permeability. High levels of BMPER antagonized BMP4-Smad5-Id1 signaling and prevented BMP4-induced downregulation of VE-cadherin and endothelial leakage, suggesting that BMPER exerts anti-BMP effects and restores endothelial barrier function. Taken together, this data demonstrates that BMPER-modulated BMP pathway activity regulates VE-cadherin expression and vascular barrier function.
Collapse
Affiliation(s)
- Thomas Helbing
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.
| | - Gwendoline Wiltgen
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Alexandra Hornstein
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Elena Z Brauers
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Linus Arnold
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Adrian Bauer
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Jennifer S Esser
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Philipp Diehl
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Sebastian Grundmann
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Katrin Fink
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Department of Emergency Medicine, University Hospital of Freiburg, Freiburg im Breisgau, Germany
| | - Cam Patterson
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- New York-Presbyterian Hospital, New York City, NY, 10065, USA
| | - Christoph Bode
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Martin Moser
- Department of Cardiology and Angiology, Heart Center Freiburg University, Hugstetter Strasse 55, 79106, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
35
|
Skulachev VP, Holtze S, Vyssokikh MY, Bakeeva LE, Skulachev MV, Markov AV, Hildebrandt TB, Sadovnichii VA. Neoteny, Prolongation of Youth: From Naked Mole Rats to “Naked Apes” (Humans). Physiol Rev 2017; 97:699-720. [DOI: 10.1152/physrev.00040.2015] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
It has been suggested that highly social mammals, such as naked mole rats and humans, are long-lived due to neoteny (the prolongation of youth). In both species, aging cannot operate as a mechanism facilitating natural selection because the pressure of this selection is strongly reduced due to 1) a specific social structure where only the “queen” and her “husband(s)” are involved in reproduction (naked mole rats) or 2) substituting fast technological progress for slow biological evolution (humans). Lists of numerous traits of youth that do not disappear with age in naked mole rats and humans are presented and discussed. A high resistance of naked mole rats to cancer, diabetes, cardiovascular and brain diseases, and many infections explains why their mortality rate is very low and almost age-independent and why their lifespan is more than 30 years, versus 3 years in mice. In young humans, curves of mortality versus age start at extremely low values. However, in the elderly, human mortality strongly increases. High mortality rates in other primates are observed at much younger ages than in humans. The inhibition of the aging process in humans by specific drugs seems to be a promising approach to prolong our healthspan. This might be a way to retard aging, which is already partially accomplished via the natural physiological phenomenon neoteny.
Collapse
Affiliation(s)
- Vladimir P. Skulachev
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Susanne Holtze
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Mikhail Y. Vyssokikh
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Lora E. Bakeeva
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Maxim V. Skulachev
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Alexander V. Markov
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Thomas B. Hildebrandt
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Viktor A. Sadovnichii
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| |
Collapse
|
36
|
Kajikawa M, Oda N, Kishimoto S, Maruhashi T, Iwamoto Y, Iwamoto A, Matsui S, Aibara Y, Mohamad Yusoff FB, Hidaka T, Kihara Y, Chayama K, Goto C, Noma K, Nakashima A, Taguchi A, Higashi Y. Increasing Risk of Osteoporotic Fracture Is Associated With Vascular Dysfunction and Abnormal Vascular Structure in Both Men and Women. Circ J 2017; 81:862-869. [DOI: 10.1253/circj.cj-16-1236] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
| | - Nozomu Oda
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Shinji Kishimoto
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Tatsuya Maruhashi
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yumiko Iwamoto
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Akimichi Iwamoto
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Shogo Matsui
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yoshiki Aibara
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Farina Binti Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Takayuki Hidaka
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University
| | - Kensuke Noma
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Ayumu Nakashima
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| | - Akira Taguchi
- Department of Oral and Maxillofacial Radiology, Matsumoto Dental University
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University
| |
Collapse
|
37
|
Angiocrine Bmp2 signaling in murine liver controls normal iron homeostasis. Blood 2016; 129:415-419. [PMID: 27903529 DOI: 10.1182/blood-2016-07-729822] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 11/23/2016] [Indexed: 01/14/2023] Open
Abstract
Microvascular endothelial cells (ECs) display a high degree of phenotypic and functional heterogeneity among different organs. Organ-specific ECs control their tissue microenvironment by angiocrine factors in health and disease. Liver sinusoidal endothelial cells (LSECs) are uniquely differentiated to fulfill important organ-specific functions in development, under homeostatic conditions, and in regeneration and liver pathology. Recently, Bmp2 has been identified by us as an organ-specific angiokine derived from LSECs. To study angiocrine Bmp2 signaling in the liver, we conditionally deleted Bmp2 in LSECs using EC subtype-specific Stab2-Cre mice. Genetic inactivation of hepatic angiocrine Bmp2 signaling in Stab2-Cre;Bmp2fl/fl (Bmp2LSECKO) mice caused massive iron overload in the liver and increased serum iron levels and iron deposition in several organs similar to classic hereditary hemochromatosis. Iron overload was mediated by decreased hepatic expression of hepcidin, a key regulator of iron homeostasis. Thus, angiocrine Bmp2 signaling within the hepatic vascular niche represents a constitutive pathway indispensable for iron homeostasis in vivo that is nonredundant with Bmp6. Notably, we demonstrate that organ-specific angiocrine signaling is essential not only for the homeostasis of the respective organ but also for the homeostasis of the whole organism.
Collapse
|
38
|
Hong OK, Yoo SJ, Son JW, Kim MK, Baek KH, Song KH, Cha BY, Jo H, Kwon HS. High glucose and palmitate increases bone morphogenic protein 4 expression in human endothelial cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2016; 20:169-75. [PMID: 26937213 PMCID: PMC4770107 DOI: 10.4196/kjpp.2016.20.2.169] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/10/2015] [Accepted: 11/13/2015] [Indexed: 11/15/2022]
Abstract
Here, we investigated whether hyperglycemia and/or free fatty acids (palmitate, PAL) aff ect the expression level of bone morphogenic protein 4 (BMP4), a proatherogenic marker, in endothelial cells and the potential role of BMP4 in diabetic vascular complications. To measure BMP4 expression, human umbilical vein endothelial cells (HUVECs) were exposed to high glucose concentrations and/or PAL for 24 or 72 h, and the effects of these treatments on the expression levels of adhesion molecules and reactive oxygen species (ROS) were examined. BMP4 loss-of-function status was achieved via transfection of a BMP4-specific siRNA. High glucose levels increased BMP4 expression in HUVECs in a dose-dependent manner. PAL potentiated such expression. The levels of adhesion molecules and ROS production increased upon treatment with high glucose and/or PAL, but this eff ect was negated when BMP4 was knocked down via siRNA. Signaling of BMP4, a proinflammatory and pro-atherogenic cytokine marker, was increased by hyperglycemia and PAL. BMP4 induced the expression of infl ammatory adhesion molecules and ROS production. Our work suggests that BMP4 plays a role in atherogenesis induced by high glucose levels and/or PAL.
Collapse
Affiliation(s)
- Oak-Kee Hong
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Soon-Jib Yoo
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Division of Endocrinology and Metabolism, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Kyunggi-do 14647, Korea
| | - Jang-Won Son
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Division of Endocrinology and Metabolism, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Kyunggi-do 14647, Korea
| | - Mee-Kyoung Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ki-Hyun Baek
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ki-Ho Song
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Bong-Yun Cha
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Korea
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, GA 30322, USA
| | - Hyuk-Sang Kwon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.; Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
39
|
Le Hiress M, Tu L, Ricard N, Phan C, Thuillet R, Fadel E, Dorfmüller P, Montani D, de Man F, Humbert M, Huertas A, Guignabert C. Proinflammatory Signature of the Dysfunctional Endothelium in Pulmonary Hypertension. Role of the Macrophage Migration Inhibitory Factor/CD74 Complex. Am J Respir Crit Care Med 2016. [PMID: 26203495 DOI: 10.1164/rccm.201402-0322oc] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Inflammation and endothelial dysfunction are considered two primary instigators of pulmonary arterial hypertension (PAH). CD74 is a receptor for the proinflammatory cytokine macrophage migration inhibitory factor (MIF). This ligand/receptor complex initiates survival pathways and cell proliferation, and it triggers the synthesis and secretion of major proinflammatory factors and cell adhesion molecules. OBJECTIVES We hypothesized that the MIF/CD74 signaling pathway is overexpressed in idiopathic PAH (iPAH) and contributes to a proinflammatory endothelial cell (EC) phenotype. METHODS Primary early passage cultures of human ECs isolated from lung tissues obtained from patients with iPAH and controls were examined for their ability to secrete proinflammatory mediators and bind inflammatory cells with or without modulation of the functional activities of the MIF/CD74 complex. In addition, we tested the efficacies of curative treatments with either the MIF antagonist ISO-1 or anti-CD74 neutralizing antibodies on the aberrant proinflammatory EC phenotype in vitro and in vivo and on the progression of monocrotaline-induced pulmonary hypertension. MEASUREMENTS AND MAIN RESULTS In human lung tissues, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin expressions are markedly up-regulated in the endothelium of distal iPAH pulmonary arteries. Circulating MIF levels are increased in the serum of patients with PAH compared with control subjects, and T-cell lymphocytes represent a source of this overabundance. In addition, CD74 is highly expressed in the endothelium of muscularized pulmonary arterioles and in cultured pulmonary ECs from iPAH, contributing to an exaggerated recruitment of peripheral blood mononuclear cells to pulmonary iPAH ECs. Finally, we found that curative treatments with the MIF antagonist ISO-1 or anti-CD74 neutralizing antibodies partially reversed development of pulmonary hypertension in rats and substantially reduced inflammatory cell infiltration. CONCLUSIONS We report here that CD74 and MIF are markedly increased and activated in patients with iPAH, contributing to the abnormal proinflammatory phenotype of pulmonary ECs in iPAH.
Collapse
Affiliation(s)
- Morane Le Hiress
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Ly Tu
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Nicolas Ricard
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Carole Phan
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Elie Fadel
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - Peter Dorfmüller
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| | - David Montani
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France.,3 AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, Kremlin-Bicêtre, France; and
| | - Frances de Man
- 4 Department of Pulmonology, VU University Medical Center/Institute of Cardiovascular Research, Amsterdam, the Netherlands
| | - Marc Humbert
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France.,3 AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, Kremlin-Bicêtre, France; and
| | - Alice Huertas
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France.,3 AP-HP, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, Kremlin-Bicêtre, France; and
| | - Christophe Guignabert
- 1 INSERM UMR_S 999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France.,2 Université Paris-Sud and Université Paris-Saclay, School of Médecine, Kremlin-Bicêtre, France
| |
Collapse
|
40
|
Ibarra Urizar A, Friberg J, Christensen DP, Lund Christensen G, Billestrup N. Inflammatory Cytokines Stimulate Bone Morphogenetic Protein-2 Expression and Release from Pancreatic Beta Cells. J Interferon Cytokine Res 2016; 36:20-9. [DOI: 10.1089/jir.2014.0199] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Adriana Ibarra Urizar
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Josefine Friberg
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Dan Ploug Christensen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen N, Denmark
| | - Gitte Lund Christensen
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Nils Billestrup
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
41
|
García de Vinuesa A, Abdelilah-Seyfried S, Knaus P, Zwijsen A, Bailly S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev 2015; 27:65-79. [PMID: 26823333 DOI: 10.1016/j.cytogfr.2015.12.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vascular system is critical for developmental growth, tissue homeostasis and repair but also for tumor development. Bone morphogenetic protein (BMP) signaling has recently emerged as a fundamental pathway of the endothelium by regulating cardiovascular and lymphatic development and by being causative for several vascular dysfunctions. Two vascular disorders have been directly linked to impaired BMP signaling: pulmonary arterial hypertension and hereditary hemorrhagic telangiectasia. Endothelial BMP signaling critically depends on the cellular context, which includes among others vascular heterogeneity, exposure to flow, and the intertwining with other signaling cascades (Notch, WNT, Hippo and hypoxia). The purpose of this review is to highlight the most recent findings illustrating the clear need for reconsidering the role of BMPs in vascular biology.
Collapse
Affiliation(s)
- Amaya García de Vinuesa
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - An Zwijsen
- VIB Center for the Biology of Disease, Leuven, Belgium; KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Médicale (INSERM, U1036), Grenoble F-38000, France; Commissariat à l'Énergie Atomique et aux Energies Alternatives, Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire Biologie du Cancer et de l'Infection, Grenoble F-38000, France; Université Grenoble-Alpes, Grenoble F-38000, France.
| |
Collapse
|
42
|
Lin CP, Huang PH, Lai CF, Chen JW, Lin SJ, Chen JS. Simvastatin Attenuates Oxidative Stress, NF-κB Activation, and Artery Calcification in LDLR-/- Mice Fed with High Fat Diet via Down-regulation of Tumor Necrosis Factor-α and TNF Receptor 1. PLoS One 2015; 10:e0143686. [PMID: 26625143 PMCID: PMC4666466 DOI: 10.1371/journal.pone.0143686] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 11/09/2015] [Indexed: 11/18/2022] Open
Abstract
Simvastatin (SIM) is anti-inflammatory. We used low density lipoprotein receptor knockout (LDLR-/-) mice and human aortic smooth muscle cells (HASMCs) as model systems to study the effect of SIM on arterial calcification and to explore the potential mechanisms contributing to this protective effect. High-fat diet (HFD) caused the LRLR -/- to develop dyslipidemia, diabetics, atherosclerosis and aortic smooth muscle calcification. SIM, N-acetyl cysteine (NAC, a ROS scavenger) and apocynin (APO, a NADPH oxidase inhibitor) did not significantly retard the development of dyslipidemia or diabetic. However, those treatments were still effective in attenuating the HFD-induced atherosclerosis and aortic smooth muscle calcification. These findings suggest that the protective effect of SIM against aortic calcification is not contributed by the cholesterol lowering effect. SIM, NAC and APO were found to attenuate the HFD induced elevation of serum TNF-α, soluble TNFR1 (sTNFR1), 3-nitro-tyrosine. We hypothesized that the pro-inflammatory cytokine, oxidative stress and TNFR1 played a role in inducing aortic calcification. We used HASMC to investigate the role of TNF-α, oxidative stress and TNFR1 in inducing aortic calcification and to elucidate the mechanism contributes the protective effect of SIM against aortic calcification. We demonstrated that treating HASMC with TNF-α induced cell Ca deposit and result in an increase in ALP, NADPH oxidase activity, NF-kB subunit p65, BMP2, MSX2, and RUNX2 expression. SIM suppressed the TNF-α induced activation of NADPH oxidase subunit p47, the above-mentioned bone markers and TNFR1 expression. Furthermore, p65, p47 and TNFR1 siRNAs inhibited the TNF-α-mediated stimulation of BMP-2, MSX2, RUNX2 expression. SIM, APO, and NAC either partially inhibit or completely block the TNF-α induced H2O2 or superoxide production. These results suggest that SIM may, independent of its cholesterol-lowering effect, suppresses the progression of vascular diseases through the inhibition of the inflammation mediators TNF-α and TNFR1.
Collapse
Affiliation(s)
- Chih-Pei Lin
- Division of Central Laboratory, Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine and Institute of Biotechnology in Medicine, Taipei, Taiwan
- * E-mail:
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | - Jaw-Wen Chen
- Division of Cardiology, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jia-Shiong Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
43
|
Varas A, Valencia J, Lavocat F, Martínez VG, Thiam NN, Hidalgo L, Fernández-Sevilla LM, Sacedón R, Vicente A, Miossec P. Blockade of bone morphogenetic protein signaling potentiates the pro-inflammatory phenotype induced by interleukin-17 and tumor necrosis factor-α combination in rheumatoid synoviocytes. Arthritis Res Ther 2015. [PMID: 26215036 PMCID: PMC4517404 DOI: 10.1186/s13075-015-0710-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Introduction Bone morphogenetic proteins (BMPs) are multifunctional secreted growth factors regulating a broad spectrum of functions in numerous systems. An increased expression and production of specific BMPs have been described in the rheumatoid arthritis (RA) synovium. The aim of this study was to analyze the involvement of the BMP signaling pathway in RA synoviocytes in response to interleukin-17 (IL-17) and tumor necrosis factor-alpha (TNF-α). Methods The expression of components of the BMP signaling pathway (BMP receptors, BMP ligands, BMP signal transducers, and BMP antagonists) was analyzed by quantitative polymerase chain reaction before and after treatment of RA synoviocytes with TNF-α or IL-17 or both. Regulation was studied in the presence of the specific BMP inhibitor DMH1 (dorsomorphin homologue 1) or an exogenous BMP ligand, BMP6. Expression and production of pro-inflammatory cytokines (IL-6 and granulocyte-macrophage colony-stimulating factor), chemokines (IL-8, CCL2, CCL5, and CXCL10), and matrix metalloproteinases (MMP-1, −2, −3, −9, and −13) were analyzed. Results RA synoviocytes express BMP receptors (mainly BMPRIA, ACTRIA, and BMPRII), signal transducers of the Smad family (Smad1 and 5 and co-Smad4), and different BMP antagonists. The modulation of the expression of the BMP target genes—Id (inhibitor of DNA-binding/differentiation) proteins and Runx (Runt-related transcription factor) transcription factors—after the addition of exogenous BMP shows that the BMP signaling pathway is active. RA synoviocytes also express BMP ligands (BMP2, BMP6, and BMP7) which are highly upregulated after activation with TNF-α and IL-17. Autocrine BMP signaling pathway can be blocked by treatment with the inhibitor DMH1, leading to an increase in the upregulated expression of pro-inflammatory cytokines, chemokines, and MMPs induced by the activation of RA synoviocytes with TNF-α and IL-17. Conversely, the additional stimulation of the BMP pathway with the exogenous addition of the BMP6 ligand decreases the expression of those pro-inflammatory and pro-destructive factors. Conclusion The results indicate that the canonical BMP pathway is functionally active in human RA synoviocytes and that the inhibition of autocrine BMP signaling exacerbates the pro-inflammatory phenotype induced in RA synoviocytes by the stimulation with IL-17 and TNF-α.
Collapse
Affiliation(s)
- Alberto Varas
- Department of Cell Biology, Faculty of Medicine, Complutense University, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| | - Jaris Valencia
- Department of Cell Biology, Faculty of Medicine, Complutense University, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| | - Fabien Lavocat
- Immunogenomics and Inflammation Research Unit and Department of Clinical Immunology and Rheumatology, Hospices Civils de Lyon, EA 4130 University of Lyon 1, Hôpital Edouard Herriot, Lyon, 69437, France.
| | - Víctor G Martínez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| | - Ndiémé Ndongo Thiam
- Immunogenomics and Inflammation Research Unit and Department of Clinical Immunology and Rheumatology, Hospices Civils de Lyon, EA 4130 University of Lyon 1, Hôpital Edouard Herriot, Lyon, 69437, France.
| | - Laura Hidalgo
- Department of Cell Biology, Faculty of Medicine, Complutense University, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| | - Lidia M Fernández-Sevilla
- Department of Cell Biology, Faculty of Medicine, Complutense University, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| | - Rosa Sacedón
- Department of Cell Biology, Faculty of Medicine, Complutense University, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| | - Angeles Vicente
- Department of Cell Biology, Faculty of Medicine, Complutense University, Plaza Ramón y Cajal s/n, Madrid, 28040, Spain.
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit and Department of Clinical Immunology and Rheumatology, Hospices Civils de Lyon, EA 4130 University of Lyon 1, Hôpital Edouard Herriot, Lyon, 69437, France.
| |
Collapse
|
44
|
Toth P, Csiszar A, Sosnowska D, Tucsek Z, Cseplo P, Springo Z, Tarantini S, Sonntag WE, Ungvari Z, Koller A. Treatment with the cytochrome P450 ω-hydroxylase inhibitor HET0016 attenuates cerebrovascular inflammation, oxidative stress and improves vasomotor function in spontaneously hypertensive rats. Br J Pharmacol 2015. [PMID: 23194285 DOI: 10.1111/bph.12079] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Hypertension increases cerebrovascular oxidative stress and inflammation and impairs vasomotor function. These pathological alterations lead to dysregulation of cerebral blood flow and exacerbate atherogenesis, increasing the morbidity of ischaemic cerebrovascular diseases and promoting vascular cognitive impairment. We aimed to test the hypothesis that increased production of the arachidonic acid metabolite 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE) contributes to hypertension-induced cerebrovascular alterations. EXPERIMENTAL APPROACH We treated male spontaneously hypertensive rats (SHR) with HET0016 (N-hydroxy-N'-(4-butyl-2-methylphenyl)-formamidine), an inhibitor of 20-HETE synthesis. In middle cerebral arteries (MCAs) of SHRs, we focused on vasomotor responses and end points that are highly relevant for cellular reactive oxygen species (ROS) production, inflammatory cytokine expression and NF-κB activation. KEY RESULTS SHRs treated with HET0016 remained hypertensive (SHR + HET0016: 149 ± 8 mmHg, Wistar-Kyoto rat: 115 ± 4 mmHg; P < 0.05.), although their systolic blood pressure was decreased compared to untreated SHRs (191 ± 6 mmHg). In MCAs of SHRs, flow-induced constriction was increased, whereas ACh- and ATP-induced dilations were impaired. This functional impairment was reversed by treatment with HET0016. Treatment with HET0016 also significantly decreased oxidative stress in MCAs of SHRs (as shown by dihydroethidium staining and analysis of vascular 5-nitrotyrosine, 4-hydroxynonenal and carbonyl content) and inhibited cerebrovascular inflammation (shown by the reduced mRNA expression of TNFα, IL-1β and IL-6). Treatment of SHRs with HET0016 also attenuated vascular NF-κB activation. In vitro treatment with 20-HETE significantly increased vascular production of ROS and promoted NF-κB activation in cultured cerebromicrovascular endothelial cells. CONCLUSIONS AND IMPLICATIONS Taken together, treatment with HET0016 confers anti-oxidative and anti-inflammatory effects in the cerebral arteries of SHRs by disrupting 20-HETE-mediated autocrine/paracrine signalling pathways in the vascular wall. It is likely that HET0016-induced decreases in blood pressure also potentiate the cerebrovascular protective effects of the drug.
Collapse
Affiliation(s)
- Peter Toth
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Luo JY, Zhang Y, Wang L, Huang Y. Regulators and effectors of bone morphogenetic protein signalling in the cardiovascular system. J Physiol 2015; 593:2995-3011. [PMID: 25952563 DOI: 10.1113/jp270207] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/27/2015] [Indexed: 12/22/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) play key roles in the regulation of cell proliferation, differentiation and apoptosis in various tissues and organs, including the cardiovascular system. BMPs signal through both Smad-dependent and -independent cascades to exert a wide spectrum of biological activities. Cardiovascular disorders such as abnormal angiogenesis, atherosclerosis, pulmonary hypertension and cardiac hypertrophy have been linked to aberrant BMP signalling. To correct the dysregulated BMP signalling in cardiovascular pathogenesis, it is essential to get a better understanding of how the regulators and effectors of BMP signalling control cardiovascular function and how the dysregulated BMP signalling contributes to cardiovascular dysfunction. We hence highlight several key regulators of BMP signalling such as extracellular regulators of ligands, mechanical forces, microRNAs and small molecule drugs as well as typical BMP effectors like direct downstream target genes, mitogen-activated protein kinases, reactive oxygen species and microRNAs. The insights into these molecular processes will help target both the regulators and important effectors to reverse BMP-associated cardiovascular pathogenesis.
Collapse
Affiliation(s)
- Jiang-Yun Luo
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Zhang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Li Wang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
46
|
Zhang M, Sara JD, Wang FL, Liu LP, Su LX, Zhe J, Wu X, Liu JH. Increased plasma BMP-2 levels are associated with atherosclerosis burden and coronary calcification in type 2 diabetic patients. Cardiovasc Diabetol 2015; 14:64. [PMID: 26003174 PMCID: PMC4450848 DOI: 10.1186/s12933-015-0214-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/08/2015] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Although Bone morphogenetic protein-2 (BMP-2) is a known mediator of bone regeneration and vascular calcification, to date no study has investigated the relationship between BMP-2 and type 2 diabetes mellitus (T2DM) and its possible role in coronary artery disease (CAD). The purpose of this study is to evaluate the relationship of BMP-2 with atherosclerosis and calcification in patients with T2DM. METHODS 124 subjects were enrolled in this study: 29 patients with T2DM and CAD; 26 patients with T2DM and without CAD; 36 patients with CAD and without T2DMand 34 without T2DM or CAD (control group). Severity of coronary lesions was assessed using coronary angiography and intravascular ultrasound (IVUS). Plasma BMP-2 levels were quantified using a commercially available ELISA kit. RESULTS Compared to the control group, the mean plasma BMP-2 level was significantly higher in T2DM patients with or without CAD (20.1 ± 1.7 or 19.3 ± 1.5 pg/ml, vs 17.2 ± 3.3 pg/ml, P < 0.001). In a multivariable linear regression analysis, both T2DM and CAD were significantly and positively associated with BMP-2 (Estimate, 0.249; standard error (SE), 0.063; p <0.0001; Estimate, 0.400; SE, 0.06; p < 0.0001). Plasma BMP-2 was also strongly correlated with glycosylated hemoglobin A1c (HbA1c) (Spearman ρ = -0.31; p = 0.0005). SYNTAX score was also significantly associated with BMP-2 (Spearman ρ = 0.46; p = 0.0002). Using the results from IVUS, plasma BMP-2 levels were shown to positively correlate with plaque burden (Spearman ρ = 0.38, P = 0.002) and plaque calcification (Spearman ρ =0.44, P = 0.0003) and to negatively correlate with lumen volume (Spearman ρ =0.31, P = 0.01). CONCLUSIONS Our study demonstrates that patients with T2DM had higher circulating levels of BMP-2 than normal controls. Plasma BMP-2 levels correlated positively with plaque burden and calcification in patients with T2DM.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, PR China.
- Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| | - Jaskanwal Deep Sara
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA.
| | - Fei-long Wang
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA.
| | - Li-Ping Liu
- Department of Nephrology, First Hospital of Tsinghua University, Beijing, China.
| | - Li-Xiao Su
- Department of Biostatistics, Rutgers School of Public Health, The State University of New Jersey, Piscataway, NJ, USA.
| | - Jing Zhe
- Department of Biostatistics, University at Buffalo, the State University of New York, Buffalo, NY, 14214, USA.
| | - Xi Wu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, PR China.
- Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| | - Jing-hua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, PR China.
| |
Collapse
|
47
|
Luna-Luna M, Medina-Urrutia A, Vargas-Alarcón G, Coss-Rovirosa F, Vargas-Barrón J, Pérez-Méndez Ó. Adipose Tissue in Metabolic Syndrome: Onset and Progression of Atherosclerosis. Arch Med Res 2015; 46:392-407. [PMID: 26009250 DOI: 10.1016/j.arcmed.2015.05.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/12/2015] [Indexed: 12/25/2022]
Abstract
Metabolic syndrome (MetS) should be considered a clinical entity when its different symptoms share a common etiology: obesity/insulin resistance as a result of a multi-organ dysfunction. The main interest in treating MetS as a clinical entity is that the addition of its components drastically increases the risk of atherosclerosis. In MetS, the adipose tissue plays a central role along with an unbalanced gut microbiome, which has become relevant in recent years. Once visceral adipose tissue (VAT) increases, dyslipidemia and endothelial dysfunction follow as additive risk factors. However, when the nonalcoholic fatty liver is present, risk of a cardiovascular event is highly augmented. Epicardial adipose tissue (EAT) seems to increase simultaneously with the VAT. In this context, the former may play a more important role in the development of the atherosclerotic plaque than the latter. Hence, EAT may act as a paracrine tissue vis-à-vis the coronary arteries favoring the local inflammation and the atheroma calcification.
Collapse
Affiliation(s)
- María Luna-Luna
- Department of Molecular Biology, Instituto Nacional de Cardiología, Mexico City, Mexico
| | | | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, Instituto Nacional de Cardiología, Mexico City, Mexico; Study Group of Atherosclerosis, Instituto Nacional de Cardiología, Mexico City, Mexico
| | | | - Jesús Vargas-Barrón
- Echocardiography, Instituto Nacional de Cardiología, Mexico City, Mexico; Study Group of Atherosclerosis, Instituto Nacional de Cardiología, Mexico City, Mexico
| | - Óscar Pérez-Méndez
- Department of Molecular Biology, Instituto Nacional de Cardiología, Mexico City, Mexico; Study Group of Atherosclerosis, Instituto Nacional de Cardiología, Mexico City, Mexico.
| |
Collapse
|
48
|
Buendía P, Montes de Oca A, Madueño JA, Merino A, Martín-Malo A, Aljama P, Ramírez R, Rodríguez M, Carracedo J. Endothelial microparticles mediate inflammation-induced vascular calcification. FASEB J 2014; 29:173-81. [PMID: 25342130 DOI: 10.1096/fj.14-249706] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Stimulation of endothelial cells (ECs) with TNF-α causes an increase in the expression of bone morphogenetic protein-2 (BMP-2) and the production of endothelial microparticles (EMPs). BMP-2 is known to produce osteogenic differentiation of vascular smooth muscle cells (VSMCs). It was found that EMPs from TNF-α-stimulated endothelial cells (HUVECs) contained a significant amount of BMP-2 and were able to enhance VSMC osteogenesis and calcification. Calcium content was greater in VSMCs exposed to EMPs from TNF-α-treated HUVECs than EMPs from nontreated HUVECs (3.56 ± 0.57 vs. 1.48 ± 0.56 µg/mg protein; P < 0.05). The increase in calcification was accompanied by up-regulation of Cbfa1 (osteogenic transcription factor) and down-regulation of SM22α (VSMC lineage marker). Inhibition of BMP-2 by small interfering RNA reduced the VSMC calcification induced by EMPs from TNF-α-treated HUVECs. Similar osteogenic capability was observed in EMPs from both patients with chronic kidney disease and senescent cells, which also presented a high level of BMP-2 expression. Labeling of EMPs with CellTracker shows that EMPs are phagocytized by VSMCs under all conditions (with or without high phosphate, control, and EMPs from TNF-α-treated HUVECs). Our data suggest that EC damage results in the release of EMPs with a high content of calcium and BMP-2 that are able to induce calcification and osteogenic differentiation of VSMCs.
Collapse
Affiliation(s)
- Paula Buendía
- Instituto Maimónides de Investigación Biomédica de Córdoba/Fundación de Investigaciones Biomédicas de Córdoba, Reina Sofía University Hospital, Córdoba, Spain; Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Addy Montes de Oca
- Instituto Maimónides de Investigación Biomédica de Córdoba/Fundación de Investigaciones Biomédicas de Córdoba, Reina Sofía University Hospital, Córdoba, Spain; Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Antonio Madueño
- Instituto Maimónides de Investigación Biomédica de Córdoba/Fundación de Investigaciones Biomédicas de Córdoba, Reina Sofía University Hospital, Córdoba, Spain
| | - Ana Merino
- Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain Laboratory of Experimental Nephrology, IDIBELL, Hospital Universitari de Bellvitge, L'Hospitalet, Barcelona, Spain
| | - Alejandro Martín-Malo
- Instituto Maimónides de Investigación Biomédica de Córdoba/Fundación de Investigaciones Biomédicas de Córdoba, Reina Sofía University Hospital, Córdoba, Spain; Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain Nephrology Unit, Reina Sofía University Hospital, Córdoba, Spain
| | - Pedro Aljama
- Instituto Maimónides de Investigación Biomédica de Córdoba/Fundación de Investigaciones Biomédicas de Córdoba, Reina Sofía University Hospital, Córdoba, Spain; Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain Nephrology Unit, Reina Sofía University Hospital, Córdoba, Spain
| | - Rafael Ramírez
- Nephrology Unit, Reina Sofía University Hospital, Córdoba, Spain; Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain Physiology Department, Alcalá de Henares University, Madrid, Spain; and
| | - Mariano Rodríguez
- Instituto Maimónides de Investigación Biomédica de Córdoba/Fundación de Investigaciones Biomédicas de Córdoba, Reina Sofía University Hospital, Córdoba, Spain; Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain Nephrology Unit, Reina Sofía University Hospital, Córdoba, Spain;
| | - Julia Carracedo
- Instituto Maimónides de Investigación Biomédica de Córdoba/Fundación de Investigaciones Biomédicas de Córdoba, Reina Sofía University Hospital, Córdoba, Spain; Redes Temáticas de Investigación Cooperativa en Salud Renal, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
49
|
Banki E, Sosnowska D, Tucsek Z, Gautam T, Toth P, Tarantini S, Tamas A, Helyes Z, Reglodi D, Sonntag WE, Csiszar A, Ungvari Z. Age-related decline of autocrine pituitary adenylate cyclase-activating polypeptide impairs angiogenic capacity of rat cerebromicrovascular endothelial cells. J Gerontol A Biol Sci Med Sci 2014; 70:665-74. [PMID: 25136000 DOI: 10.1093/gerona/glu116] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/11/2014] [Indexed: 01/16/2023] Open
Abstract
Aging impairs angiogenic capacity of cerebromicrovascular endothelial cells (CMVECs) promoting microvascular rarefaction, but the underlying mechanisms remain elusive. PACAP is an evolutionarily conserved neuropeptide secreted by endothelial cells and neurons, which confers important antiaging effects. To test the hypothesis that age-related changes in autocrine PACAP signaling contributes to dysregulation of endothelial angiogenic capacity, primary CMVECs were isolated from 3-month-old (young) and 24-month-old (aged) Fischer 344 x Brown Norway rats. In aged CMVECs, expression of PACAP was decreased, which was associated with impaired capacity to form capillary-like structures, impaired adhesiveness to collagen (assessed using electric cell-substrate impedance sensing [ECIS] technology), and increased apoptosis (caspase3 activity) when compared with young cells. Overexpression of PACAP in aged CMVECs resulted in increased formation of capillary-like structures, whereas it did not affect cell adhesion. Treatment with recombinant PACAP also significantly increased endothelial tube formation and inhibited apoptosis in aged CMVECs. In young CMVECs shRNA knockdown of autocrine PACAP expression significantly impaired tube formation capacity, mimicking the aging phenotype. Cellular and mitochondrial reactive oxygen species production (dihydroethidium and MitoSox fluorescence, respectively) were increased in aged CMVECs and were unaffected by PACAP. Collectively, PACAP exerts proangiogenic effects and age-related dysregulation of autocrine PACAP signaling may contribute to impaired angiogenic capacity of CMVECs in aging.
Collapse
Affiliation(s)
- Eszter Banki
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center. Department of Anatomy, MTA-PTE PACAP Lendulet Research Team
| | - Danuta Sosnowska
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Zsuzsanna Tucsek
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Tripti Gautam
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Peter Toth
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Stefano Tarantini
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Andrea Tamas
- Department of Anatomy, MTA-PTE PACAP Lendulet Research Team
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Szentágothai Research Center, and
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Lendulet Research Team
| | - William E Sonntag
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Anna Csiszar
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center. Department of Pathophysiology and Gerontology, Medical School and Szentagothai Research Center, University of Pécs, Hungary. Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center
| | - Zoltan Ungvari
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center. Department of Pathophysiology and Gerontology, Medical School and Szentagothai Research Center, University of Pécs, Hungary. Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center.
| |
Collapse
|
50
|
Pinocembrin protects human brain microvascular endothelial cells against fibrillar amyloid-β(1-40) injury by suppressing the MAPK/NF-κB inflammatory pathways. BIOMED RESEARCH INTERNATIONAL 2014; 2014:470393. [PMID: 25157358 PMCID: PMC4135138 DOI: 10.1155/2014/470393] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/02/2014] [Accepted: 06/15/2014] [Indexed: 11/28/2022]
Abstract
Cerebrovascular accumulation of amyloid-β (Aβ) peptides in Alzheimer's disease (AD) may contribute to disease progression through Aβ-induced microvascular endothelial pathogenesis. Pinocembrin has been shown to have therapeutic effects in AD models. These effects correlate with preservation of microvascular function, but the effect on endothelial cells under Aβ-damaged conditions is unclear. The present study focuses on the in vitro protective effect of pinocembrin on fibrillar Aβ1−40 (fAβ1−40) injured human brain microvascular endothelial cells (hBMECs) and explores potential mechanisms. The results demonstrate that fAβ1−40-induced cytotoxicity in hBMECs can be rescued by pinocembrin treatment. Pinocembrin increases cell viability, reduces the release of LDH, and relieves nuclear condensation. The mechanisms of this reversal from Aβ may be associated with the inhibition of inflammatory response, involving inhibition of MAPK activation, downregulation of phosphor-IKK level, relief of IκBα degradation, blockage of NF-κB p65 nuclear translocation, and reduction of the release of proinflammatory cytokines. Pinocembrin does not show obvious effects on regulating the redox imbalance after exposure to fAβ1−40. Together, the suppression of MAPK and the NF-κB signaling pathways play a significant role in the anti-inflammation of pinocembrin in hBMECs subjected to fAβ1−40. This may serve as a therapeutic agent for BMEC protection in Alzheimer's-related deficits.
Collapse
|