1
|
Chen H, Lai H, Chi H, Fan W, Huang J, Zhang S, Jiang C, Jiang L, Hu Q, Yan X, Chen Y, Zhang J, Yang G, Liao B, Wan J. Multi-modal transcriptomics: integrating machine learning and convolutional neural networks to identify immune biomarkers in atherosclerosis. Front Cardiovasc Med 2024; 11:1397407. [PMID: 39660117 PMCID: PMC11628520 DOI: 10.3389/fcvm.2024.1397407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
Background Atherosclerosis, a complex chronic vascular disorder with multifactorial etiology, stands as the primary culprit behind consequential cardiovascular events, imposing a substantial societal and economic burden. Nevertheless, our current understanding of its pathogenesis remains imprecise. In this investigation, our objective is to establish computational models elucidating molecular-level markers associated with atherosclerosis. This endeavor involves the integration of advanced machine learning techniques and comprehensive bioinformatics analyses. Materials and methods Our analysis incorporated data from three publicly available the Gene Expression Omnibus (GEO) datasets: GSE100927 (104 samples, 30,558 genes), which includes atherosclerotic lesions and control arteries from carotid, femoral, and infra-popliteal arteries of deceased organ donors; GSE43292 (64 samples, 23,307 genes), consisting of paired carotid endarterectomy samples from 32 hypertensive patients, comparing atheroma plaques and intact tissues; and GSE159677 (30,498 single cells, 33,538 genes), examining single-cell transcriptomes of calcified atherosclerotic core plaques and adjacent carotid artery tissues from patients undergoing carotid endarterectomy. Utilizing single-cell sequencing, highly variable atherosclerotic monocyte subpopulations were systematically identified. We analyzed cellular communication patterns with temporal dynamics. The bioinformatics approach Weighted Gene Co-expression Network Analysis (WGCNA) identified key modules, constructing a Protein-Protein Interaction (PPI) network from module-associated genes. Three machine-learning models derived marker genes, formulated through logistic regression and validated via convolutional neural network(CNN) modeling. Subtypes were clustered based on Gene Set Variation Analysis (GSVA) scores, validated through immunoassays. Results Three pivotal atherosclerosis-associated genes-CD36, S100A10, CSNK1A1-were unveiled, offering valuable clinical insights. Profiling based on these genes delineated two distinct isoforms: C2 demonstrated potent microbicidal activity, while C1 engaged in inflammation regulation, tissue repair, and immune homeostasis. Molecular docking analyses explored therapeutic potential for Estradiol, Zidovudine, Indinavir, and Dronabinol for clinical applications. Conclusion This study introduces three signature genes for atherosclerosis, shaping a novel paradigm for investigating clinical immunological medications. It distinguishes the high biocidal C2 subtype from the inflammation-modulating C1 subtype, utilizing identified signature gene as crucial targets.
Collapse
Affiliation(s)
- Haiqing Chen
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Haotian Lai
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Hao Chi
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Wei Fan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jinbang Huang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Shengke Zhang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chenglu Jiang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Qingwen Hu
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Xiuben Yan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Yemeng Chen
- New York College of Traditional Chinese Medicine, Mineola, NY, United States
| | - Jieying Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Bin Liao
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Juyi Wan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Wiesli MG, Huber MW, Weisse B, Zboray R, Kiderlen S, González-Vázquez A, Maniura-Weber K, Rottmar M, Lackington WA. Immunomodulation Using BMP-7 and IL-10 to Enhance the Mineralization Capacity of Bone Progenitor Cells in a Fracture Hematoma-Like Environment. Adv Healthc Mater 2024:e2400077. [PMID: 38599586 DOI: 10.1002/adhm.202400077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Following biomaterial implantation, a failure to resolve inflammation during the formation of a fracture hematoma can significantly limit the biomaterial's ability to facilitate bone regeneration. This study aims to combine the immunomodulatory and osteogenic effects of BMP-7 and IL-10 with the regenerative capacity of collagen-hydroxyapatite (CHA) scaffolds to enhance in vitro mineralization in a hematoma-like environment. Incubation of CHA scaffolds with human whole blood leads to rapid adsorption of fibrinogen, significant stiffening of the scaffold, and the formation of a hematoma-like environment characterized by a limited capacity to support the infiltration of human bone progenitor cells, a significant upregulation of inflammatory cytokines and acute phase proteins, and significantly reduced osteoconductivity. CHA scaffolds functionalized with BMP-7 and IL-10 significantly downregulate the production of key inflammatory cytokines, including IL-6, IL-8, and leptin, creating a more permissive environment for mineralization, ultimately enhancing the biomaterial's osteoconductivity. In conclusion, targeting the onset of inflammation in the early phase of bone healing using BMP-7 and IL-10 functionalized CHA scaffolds is a promising approach to effectively downregulate inflammatory processes, while fostering a more permissive environment for bone regeneration.
Collapse
Affiliation(s)
- Matthias Guido Wiesli
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, 9014, Switzerland
| | - Matthias Werner Huber
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, 9014, Switzerland
| | - Bernhard Weisse
- Laboratory for Mechanical Systems Engineering, Empa, Dübendorf, 8600, Switzerland
| | - Robert Zboray
- Center of X-ray Analytics, Empa, Dübendorf, 8600, Switzerland
| | | | - Arlyng González-Vázquez
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, 9014, Switzerland
| | - Markus Rottmar
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, 9014, Switzerland
| | - William Arthur Lackington
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, 9014, Switzerland
| |
Collapse
|
3
|
Jawich K, Hadakie R, Jamal S, Habeeb R, Al Fahoum S, Ferlin A, De Toni L. Emerging Role of Non-collagenous Bone Proteins as Osteokines in Extraosseous Tissues. Curr Protein Pept Sci 2024; 25:215-225. [PMID: 37937553 DOI: 10.2174/0113892037268414231017074054] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 11/09/2023]
Abstract
Bone is a unique tissue, composed of various types of cells embedded in a calcified extracellular matrix (ECM), whose dynamic structure consists of organic and inorganic compounds produced by bone cells. The main inorganic component is represented by hydroxyapatite, whilst the organic ECM is primarily made up of type I collagen and non-collagenous proteins. These proteins play an important role in bone homeostasis, calcium regulation, and maintenance of the hematopoietic niche. Recent advances in bone biology have highlighted the importance of specific bone proteins, named "osteokines", possessing endocrine functions and exerting effects on nonosseous tissues. Accordingly, osteokines have been found to act as growth factors, cell receptors, and adhesion molecules, thus modifying the view of bone from a static tissue fulfilling mobility to an endocrine organ itself. Since bone is involved in a paracrine and endocrine cross-talk with other tissues, a better understanding of bone secretome and the systemic roles of osteokines is expected to provide benefits in multiple topics: such as identification of novel biomarkers and the development of new therapeutic strategies. The present review discusses in detail the known osseous and extraosseous effects of these proteins and the possible respective clinical and therapeutic significance.
Collapse
Affiliation(s)
- Kenda Jawich
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syrian Arab Republic
- Department of Biochemistry, Faculty of Pharmacy, International University of Science and Technology, Darrah, Syrian Arab Republic
| | - Rana Hadakie
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syrian Arab Republic
| | - Souhaib Jamal
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syrian Arab Republic
| | - Rana Habeeb
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syrian Arab Republic
- Department of Biochemistry, Faculty of Pharmacy, International University of Science and Technology, Darrah, Syrian Arab Republic
| | - Sahar Al Fahoum
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Damascus University, Damascus, Syrian Arab Republic
| | - Alberto Ferlin
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
| | - Luca De Toni
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
| |
Collapse
|
4
|
Cassuto J, Folestad A, Göthlin J, Malchau H, Kärrholm J. The importance of BMPs and TGF-βs for endochondral bone repair - A longitudinal study in hip arthroplasty patients. Bone Rep 2023; 19:101723. [PMID: 38047271 PMCID: PMC10690547 DOI: 10.1016/j.bonr.2023.101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/14/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Osseointegration of hip implants, although a decade-long process, shows striking similarities with the four major phases of endochondral bone repair. In the current study we investigated the spatiotemporal involvement of bone morphogenic proteins (BMPs) and transforming growth factor betas (TGF-βs) throughout the process of bone repair leading to successfully osseointegrated hip implants. Materials and methods Twenty-four patients that had undergone primary total hip arthroplasty (THA) due to one-sided osteoarthritis (OA) were investigated during a period of 18 years (Y) with repeated measurements of plasma biomarkers as well as clinical and radiological variables. All implants were clinically and radiographically well anchored throughout the follow-up. Eighty-one healthy donors divided in three gender- and age-matched groups and twenty OA patients awaiting THA, served as controls. Plasma was analyzed for BMP-1, -2, -3, -4, -6, -7 -9 and TGF-β1, -β2, -β3 by use of a high-sensitivity and wide dynamic range electrochemiluminescence technique allowing for detection of minor changes. Results Spatiotemporal changes during the follow-up are presented in the context of the four phases of endochondral bone repair shown in earlier studies and transposed to the current study based on similarities in biomarker responses. Phase 1: Primary proinflammatory phase lasting from surgery until day 7, Phase 2: Chondrogenic phase from day 7 until 18 months postsurgery, Phase 3: Secondary proinflammatory and cartilage remodeling phase lasting from 18 months until 7Y, Phase 4: coupled bone remodeling from 7Y until 18Y postsurgery. BMP-1 increased sharply shortly after surgery and remained significantly above healthy during the chondrocyte recruitment, proliferation, and hypertrophy phases with a subsequent return to control level at 5Y postsurgery. BMP-2 was above healthy controls before surgery and 1 day after surgery before decreasing to control level and remaining there throughout the follow-up. BMP-3 was at control level from presurgery until 6M after surgery when it increased to a peak at 2Y during the cartilage hypertrophy phase followed by a gradual decrease to control level at 10Y during the phase of bone formation. In the following, BMP-3 decreased below controls to a nadir 15Y postsurgery during coupled bone remodeling. BMP-4 was at control level from presurgery until 10Y postsurgery when it increased to a sharp peak at 15Y after surgery followed by a return to the level of healthy at 18Y. BMP-6 did not differ from healthy during the follow-up. BMP-7 was at control level from presurgery until 1Y postsurgery before gradually increasing to a peak at 10Y during the early phase of osteogenesis with a gradual return to control level at 18Y during the phase of coupled bone remodeling. BMP-9 was above OA before surgery followed by a decrease to basal level on day 1 after surgery and a renewed increase to a plateau above controls lasting from 6 W until returning to the level of healthy at 18Y postsurgery, i.e., throughout the phases of cartilage formation, cartilage hypertrophy and remodeling, bone formation and coupled bone remodeling. TGF-β1 was above controls presurgery before decreasing to baseline shortly after surgery followed by a renewed increase at 6 M to a peak at 2Y during cartilage hypertrophy/remodeling followed by a gradual return to baseline at 10Y during early osteoblastogenesis. TGF-β2 was at control level from presurgery until the phase of cartilage remodeling at 5Y when it increased sharply to a peak at 7Y with a gradual return to baseline at 18Y postsurgery. TGF-β3 remained at control level throughout the study. Conclusion This study shows that the involvement of BMPs and TGF-βs in endochondral bone repair is a process of stepwise recruitment of individual biomarkers characterized by distinct, yet overlaping, spatiotemporal patterns that extend from the early phase of pre-chondrocyte recruitment until the late phase of coupled bone remodeling.
Collapse
Affiliation(s)
- Jean Cassuto
- Orthopedic Research Unit & Department of Orthopedic Surgery, Sahlgrenska University Hospital, Mölndal, Sweden
- Institution of Clinical Sciences, Göteborg University, Göteborg, Sweden
| | - Agnetha Folestad
- Department of Orthopedics, CapioLundby Hospital, Göteborg, Sweden
| | - Jan Göthlin
- Department of Radiology, Sahlgrenska University Hospital, Mölndal, Sweden
- Institution of Clinical Sciences, Göteborg University, Göteborg, Sweden
| | - Henrik Malchau
- Orthopedic Research Unit & Department of Orthopedic Surgery, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Orthopedic Surgery, Harvard Medical School, Boston, USA
| | - Johan Kärrholm
- Orthopedic Research Unit & Department of Orthopedic Surgery, Sahlgrenska University Hospital, Mölndal, Sweden
- Institution of Clinical Sciences, Göteborg University, Göteborg, Sweden
| |
Collapse
|
5
|
Stakišaitis D, Kapočius L, Kilimaitė E, Gečys D, Šlekienė L, Balnytė I, Palubinskienė J, Lesauskaitė V. Preclinical Study in Mouse Thymus and Thymocytes: Effects of Treatment with a Combination of Sodium Dichloroacetate and Sodium Valproate on Infectious Inflammation Pathways. Pharmaceutics 2023; 15:2715. [PMID: 38140056 PMCID: PMC10747708 DOI: 10.3390/pharmaceutics15122715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/17/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
The research presents data from a preclinical study on the anti-inflammatory effects of a sodium dichloroacetate and sodium valproate combination (DCA-VPA). The 2-week treatment with a DCA 100 mg/kg/day and VPA 150 mg/kg/day combination solution in drinking water's effects on the thymus weight, its cortex/medulla ratio, Hassall's corpuscles (HCs) number in the thymus medulla, and the expression of inflammatory and immune-response-related genes in thymocytes of male Balb/c mice were studied. Two groups of mice aged 6-7 weeks were investigated: a control (n = 12) and a DCA-VPA-treated group (n = 12). The treatment did not affect the body weight gain (p > 0.05), the thymus weight (p > 0.05), the cortical/medulla ratio (p > 0.05), or the number of HCs (p > 0.05). Treatment significantly increased the Slc5a8 gene expression by 2.1-fold (p < 0.05). Gene sequence analysis revealed a significant effect on the expression of inflammation-related genes in thymocytes by significantly altering the expression of several genes related to the cytokine activity pathway, the inflammatory response pathway, and the Il17 signaling pathway in thymocytes. Data suggest that DCA-VPA exerts an anti-inflammatory effect by inhibiting the inflammatory mechanisms in the mouse thymocytes.
Collapse
Affiliation(s)
- Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania
| | - Linas Kapočius
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Evelina Kilimaitė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Dovydas Gečys
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu Ave., 50161 Kaunas, Lithuania;
| | - Lina Šlekienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Jolita Palubinskienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (L.Š.); (I.B.); (J.P.)
| | - Vaiva Lesauskaitė
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu Ave., 50161 Kaunas, Lithuania;
| |
Collapse
|
6
|
Smoljan I, Detel D, Buljevic S, Erjavec I, Marić I. Therapeutic Potential of BMP7 in the Treatment of Osteoporosis Caused by the Interaction between Inflammation and Corticosteroids in Inflammatory Bowel Disease. Biomedicines 2023; 11:2161. [PMID: 37626658 PMCID: PMC10452398 DOI: 10.3390/biomedicines11082161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Individuals with inflammatory bowel disease (IBD) have an increased risk of bone impairment, which is a process controlled by the RANKL/RANK/OPG system, mostly due to chronic inflammation and corticosteroid treatment. Bone morphogenic protein 7 (BMP7) has a complex role in maintaining inflammation and bone remodeling but little is known about its anti-inflammatory potential in chronic colitis. We investigated the effect of systemically administered BMP7 and corticosteroids on the severity of inflammation, macrophage differentiation, and bone regeneration in a chronic IBD model. METHODS Chronic colitis was induced in male Sprague Dawley rats via weekly administration of 2,4,6-trinitrobenzenesulfonic acid over 21 days following BMP7 or corticosteroid treatment for five days. The levels of serum and colon tissue inflammatory cytokines, RANKL/OPG system, as well as markers of macrophage polarization, were detected using RT-PCR, ELISA, or immunohistochemistry. Long bone and spine analyses were performed using microcomputed tomography (micro-CT). RESULTS The administration of BMP7 reduced the adverse effects of colitis and led to elevated OPG and RANK in the colon with a simultaneous decrease in TNF-α and an increase in IL-10 and TGF-β. Decreased expression of the M2 macrophage marker CD163 was found in the BMP7-treated rats compared with the colitis group, whereas the number of M1 marker iNOS-positive cells did not differ between the groups. As a result of the BMP7 treatment, morphometric parameters of trabecular bone increased, and increased trabecular separation noted in the colitis group did not appear. CONCLUSIONS We showed that BMP7 suppressed the inflammatory response in chronic colitis, mainly by shifting the cytokine balance and by triggering alterations in the RANKL/OPG system rather than through a macrophage polarization imbalance. In addition, considering the demonstrated effect of BMP7 on bone morphology and structure, it can be suggested that BMP7 plays a role in the managing of osteoporosis in chronic colitis, and thus, its therapeutic potential in the treatment of IBD should be further evaluated.
Collapse
Affiliation(s)
- Ivana Smoljan
- Department of Internal Medicine, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia;
- Department of Cardiovascular Diseases, Clinical Hospital Center Rijeka, Kresimirova 42, 51000 Rijeka, Croatia
| | - Dijana Detel
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia;
| | - Suncica Buljevic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia;
| | - Igor Erjavec
- Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Ivana Marić
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia;
| |
Collapse
|
7
|
Narasimhulu CA, Singla DK. BMP-7 Attenuates Sarcopenia and Adverse Muscle Remodeling in Diabetic Mice via Alleviation of Lipids, Inflammation, HMGB1, and Pyroptosis. Antioxidants (Basel) 2023; 12:331. [PMID: 36829889 PMCID: PMC9952667 DOI: 10.3390/antiox12020331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Diabetic myopathy involves hyperglycemia, oxidative stress, and inflammation. However, the role of hypercholesterolemia-induced inflammation-mediated pathological mechanisms leading to fibrosis, sarcopenia, deterioration of muscle, and muscle dysfunction in diabetes is not well understood. In this study, we investigated the novel role of bone morphogenetic protein-7 (BMP-7) in ameliorating metabolic alterations, inflammation, pyroptosis, TGF-β/SMAD cell signaling mechanisms, and progression of diabetic myopathy. C57BL/6J mice were treated with saline, streptozotocin (STZ), or STZ+BMP-7. Diabetes was confirmed by increased fasting glucose levels and a glucose tolerance test. Gastrocnemius muscle and blood samples were collected for lipid and tissue analysis using various methods. A significant increase in hyperglycemia resulted in an increase in lipid accumulation, monocyte infiltration, and inflammation, as well as an increase in pyroptotic markers and signaling markers in diabetic muscle myocytes. A structural analysis showed significant muscle loss, and increased muscle deterioration and fibrosis leading to muscle dysfunction. BMP-7 attenuated pathological processes that resulted in significantly improved muscle function. We report, for the first time, that increased hyperlipidemia aggravates inflammation-induced pyroptosis, resulting in significant muscle loss, sarcopenia, and adverse skeletal muscle remodeling in diabetic muscle myopathy. Interventional treatment with BMP-7 attenuates hypercholesterolemia-induced inflammation-mediated sarcopenia and adverse muscle remodeling, suggesting BMP-7 could be a potential treatment option for diabetic muscle myopathy.
Collapse
Affiliation(s)
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
8
|
Ye D, Liu Y, Pan H, Feng Y, Lu X, Gan L, Wan J, Ye J. Insights into bone morphogenetic proteins in cardiovascular diseases. Front Pharmacol 2023; 14:1125642. [PMID: 36909186 PMCID: PMC9996008 DOI: 10.3389/fphar.2023.1125642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are secretory proteins belonging to the transforming growth factor-β (TGF-β) superfamily. These proteins play important roles in embryogenesis, bone morphogenesis, blood vessel remodeling and the development of various organs. In recent years, as research has progressed, BMPs have been found to be closely related to cardiovascular diseases, especially atherosclerosis, vascular calcification, cardiac remodeling, pulmonary arterial hypertension (PAH) and hereditary hemorrhagic telangiectasia (HHT). In this review, we summarized the potential roles and related mechanisms of the BMP family in the cardiovascular system and focused on atherosclerosis and PAH.
Collapse
Affiliation(s)
- Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yinghui Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liren Gan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
9
|
Xie Y, Chen H, Qu P, Qiao X, Guo L, Liu L. Novel insight on the role of Macrophages in atherosclerosis: Focus on polarization, apoptosis and efferocytosis. Int Immunopharmacol 2022; 113:109260. [DOI: 10.1016/j.intimp.2022.109260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/13/2022] [Indexed: 11/05/2022]
|
10
|
Bernhard JC, Marolt Presen D, Li M, Monforte X, Ferguson J, Leinfellner G, Heimel P, Betti SL, Shu S, Teuschl-Woller AH, Tangl S, Redl H, Vunjak-Novakovic G. Effects of Endochondral and Intramembranous Ossification Pathways on Bone Tissue Formation and Vascularization in Human Tissue-Engineered Grafts. Cells 2022; 11:cells11193070. [PMID: 36231032 PMCID: PMC9564153 DOI: 10.3390/cells11193070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
Bone grafts can be engineered by differentiating human mesenchymal stromal cells (MSCs) via the endochondral and intramembranous ossification pathways. We evaluated the effects of each pathway on the properties of engineered bone grafts and their capacity to drive bone regeneration. Bone-marrow-derived MSCs were differentiated on silk scaffolds into either hypertrophic chondrocytes (hyper) or osteoblasts (osteo) over 5 weeks of in vitro cultivation, and were implanted subcutaneously for 12 weeks. The pathways' constructs were evaluated over time with respect to gene expression, composition, histomorphology, microstructure, vascularization and biomechanics. Hypertrophic chondrocytes expressed higher levels of osteogenic genes and deposited significantly more bone mineral and proteins than the osteoblasts. Before implantation, the mineral in the hyper group was less mature than that in the osteo group. Following 12 weeks of implantation, the hyper group had increased mineral density but a similar overall mineral composition compared with the osteo group. The hyper group also displayed significantly more blood vessel infiltration than the osteo group. Both groups contained M2 macrophages, indicating bone regeneration. These data suggest that, similar to the body's repair processes, endochondral pathway might be more advantageous when regenerating large defects, whereas intramembranous ossification could be utilized to guide the tissue formation pattern with a scaffold architecture.
Collapse
Affiliation(s)
- Jonathan C. Bernhard
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Ming Li
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xavier Monforte
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria
| | - James Ferguson
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Gabriele Leinfellner
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Patrick Heimel
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- School of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Susanna L. Betti
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sharon Shu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Andreas H. Teuschl-Woller
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria
| | - Stefan Tangl
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- School of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Correspondence: (H.R.); (G.V.-N.); Tel.: +43-(0)-59393-41961 (H.R.); +1-212-305-2304 (G.V.-N.); Fax: +43-(0)-59393-41982 (H.R.); +1-212-305-4692 (G.V.-N.)
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
- College of Dental Medicine, Columbia University, New York, NY 10032, USA
- Correspondence: (H.R.); (G.V.-N.); Tel.: +43-(0)-59393-41961 (H.R.); +1-212-305-2304 (G.V.-N.); Fax: +43-(0)-59393-41982 (H.R.); +1-212-305-4692 (G.V.-N.)
| |
Collapse
|
11
|
Jia Y, Li D, Yu J, Jiang W, Liao X, Zhao Q. Potential diabetic cardiomyopathy therapies targeting pyroptosis: A mini review. Front Cardiovasc Med 2022; 9:985020. [PMID: 36061533 PMCID: PMC9433721 DOI: 10.3389/fcvm.2022.985020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Pyroptosis is primarily considered a pro-inflammatory class of caspase-1- and gasdermin D (GSDMD)-dependent programmed cell death. Inflammasome activation promotes the maturation and release of interleukin (IL)-1β and IL-18, cleavage of GSDMD, and development of pyroptosis. Recent studies have reported that NLRP3 inflammasome activation-mediated pyroptosis aggravates the formation and development of diabetes cardiomyopathy (DCM). These studies provide theoretical mechanisms for exploring a novel approach to treat DCM-associated cardiac dysfunction. Accordingly, this review aims to summarize studies that investigated possible DCM therapies targeting pyroptosis and elucidate the molecular mechanisms underlying NLRP3 inflammasome-mediated pyroptosis, and its potential association with the pathogenesis of DCM. This review may serve as a basis for the development of potential pharmacological agents as novel and effective treatments for managing and treating DCM.
Collapse
Affiliation(s)
- Yu Jia
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dongze Li
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, Chengdu, China
| | - Jing Yu
- Department of Emergency Medicine and National Clinical Research Center for Geriatrics, Disaster Medicine Center, West China Hospital, Sichuan University West China School of Medicine, Chengdu, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoyang Liao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Zhao
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qian Zhao,
| |
Collapse
|
12
|
Therapeutic Benefit in Rheumatoid Cachexia Illustrated Using a Novel Primary Human Triple Cell Coculture Model. Int J Inflam 2022; 2022:1524913. [PMID: 35693848 PMCID: PMC9184217 DOI: 10.1155/2022/1524913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 11/21/2022] Open
Abstract
Background The loss of muscle mass in rheumatoid arthritis (RA), termed rheumatoid cachexia, is predicted to result from the complex interactions between different cell types involved in the maintenance of skeletal muscle mass, namely, myoblasts, fibroblasts, and macrophages. The complexity within the muscle is further highlighted by the incidence of nonresponsiveness to current RA treatment strategies. Method This study aimed at determining differences in the cellular responses in a novel human primary cell triple coculture model exposed to serum collected from nonarthritic controls (NC), RA treatment naïve (RATN), and RA treatment-nonresponding (RATNR) patients. Bone morphogenetic protein-7 (BMP-7) was investigated as a treatment option. Results Plasma analysis indicated that samples were indeed representative of healthy and RA patients—notably, the RATNR patients additionally exhibited dysregulated IL-6/IL-10 correlations. Coculture exposure to serum from RATNR patients demonstrated increased cellular growth (p < 0.001), while both hepatocyte growth factor (p < 0.01) and follistatin (p < 0.001) were reduced when compared to NC. Furthermore, decreased concentration of markers of extracellular matrix formation, transforming growth factor-β (TGF-β; p < 0.05) and fibronectin (p < 0.001), but increased collagen IV (p < 0.01) was observed following RATNR serum exposure. Under healthy conditions, BMP-7 exhibited potentially beneficial results in reducing fibrosis-generating TGF-β (p < 0.05) and fibronectin (p < 0.05). BMP-7 further exhibited protective potential in the RA groups through reversing the aberrant tendencies observed especially in the RATNR serum-exposed group. Conclusion Exposure of the triple coculture to RATN and RATNR serum resulted in dysregulated myoblast proliferation and growth, and ECM impairment, which was reversed by BMP-7 treatment.
Collapse
|
13
|
Kim BY, Choi SH, Kim JY, Ko J, Yook JI, Kim HS, Lee EJ, Kikkawa DO, Yoon JS. Potential Therapeutic Role of Bone Morphogenic Protein 7 (BMP7) in the Pathogenesis of Graves' Orbitopathy. Invest Ophthalmol Vis Sci 2022; 63:7. [PMID: 35671049 PMCID: PMC9187939 DOI: 10.1167/iovs.63.6.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose We investigated a role of bone morphogenic protein 7 (BMP7), a member of the TGF-β superfamily on pathogenic mechanism of Graves' orbitopathy (GO). The therapeutic effects of BMP7 on inflammation and fibrosis were evaluated in cultured Graves' orbital fibroblasts. Methods Expression of BMP7 was compared in cultured orbital tissue explants from GO (n = 12) and normal control (n = 12) subjects using real-time PCR. Orbital fibroblasts were cultured from orbital connective tissues obtained from GO (n = 3) and normal control patients (n = 3). Cells were pretreated with recombinant human BMP7 (rhBMP7) before stimulation with TGF-β, IL-1β, and TNF-α. Fibrosis-related proteins and inflammatory cytokines were analyzed by Western blotting. The activation of signaling molecules in inflammation and fibrosis was also analyzed. Results The expressions of BMP7 mRNA were lower in GO orbital tissues than control. Fibrosis-related proteins, fibronectin, collagen 1α, and α-SMA induced by TGF-β were suppressed by treating rhBMP7, and rhBMP7 upregulated TGF-β induced SMAD1/5/8 protein expression, whereas downregulated SMAD2/3. Increased pro-inflammatory molecules, IL-6, IL-8, and intercellular adhesion molecule-1 (ICAM-1) by IL-1β or TNF-α were blocked by rhBMP7 treatment, and the expression of phosphorylated NFκB and Akt was suppressed by rhBMP7 treatment. Conclusions BMP7 transcript levels were downregulated in Graves' orbital tissues. Exogenous BMP7 treatment showed inhibitory effects on the production of profibrotic proteins and proinflammatory cytokines in orbital fibroblasts. Our results provide a molecular basis of BMP7 as a new potential therapeutic agent through the opposing mechanism of profibrotic TGF-β/SMAD signaling and proinflammatory cytokine production.
Collapse
Affiliation(s)
- Bo Yi Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Hyun Choi
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Ji-Young Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - JaeSang Ko
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| | - Jong In Yook
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Hyun Sil Kim
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Eun Jig Lee
- Department of Endocrinology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Don O Kikkawa
- Department of Ophthalmology, Division of Oculofacial Plastic and Reconstructive Surgery, University of California San Diego, La Jolla, California, United States
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Zhang Y, Xu Y, Jing X, Lu W, Zhang F, Qin C. Moscatilin suppresses the inflammation from macrophages and T cells. Open Med (Wars) 2022; 17:756-767. [PMID: 35509689 PMCID: PMC9008319 DOI: 10.1515/med-2022-0456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/06/2022] [Accepted: 02/18/2022] [Indexed: 11/15/2022] Open
Abstract
In this study, we aim to investigate moscatilin in alleviating symptoms of autoimmune liver disease (ALD) in a concanavalin A (ConA)-induced liver injury mouse model and elucidate the underlying mechanisms. ALD mouse models were constructed by intravenous injection of ConA (20 mg/kg) and the serum level of alanine aminotransferase (ALT) was measured using an enzyme-linked immunosorbent assay. Moscatilin in various doses was administered for two days starting from a day before the ConA injection. We showed that moscatilin dose-dependently decreased ALT levels in liver tissue of ALD mouse models. Ifng and Tnfa also showed significant downregulation in liver tissues. Macrophages only showed significant Tnfa downregulation and CD4+ T cells only showed significant Ifng downregulation at high moscatilin doses. In vivo administration of moscatilin induced interleukin-37 upregulation in hepatic tissues. In vitro, moscatilin also induced IL-37 upregulation in hepatic stellate cell line JS-1 rather than immune cells represented by RAW264.7 and CTLL-2 cell lines, suggesting that the hepatic stellate cell is majorly responsive to moscatilin treatment in terms of interleukin (IL)-37 upregulation. Our data indicate that moscatilin could alleviate liver injury in ConA-induced ALD mouse models through anti-inflammatory activities, warranting further development of moscatilin as a new drug in treating ALD.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Hepatobiliary Surgery, Taian City Central Hospital, Taian 271000, Shandong, China
| | - Yugang Xu
- Department of General Surgery, Taian City Central Hospital, Taian 271000, Shandong, China
| | - Xiujie Jing
- Department of Pediatrics, Taian City Central Hospital, Taian 271000, Shandong, China
| | - Wenkui Lu
- Department of General Surgery, Dongping People's Hospital, Dongping 271500, China
| | - Fusen Zhang
- Department of Critical Care Unit, Taian City Central Hospital, Taian 271000, Shandong, China
| | - Chengkun Qin
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, No. 324 Jingwuwei Road No.7, Jinan 250021, Shandong, China
| |
Collapse
|
15
|
Sconocchia T, Sconocchia G. Regulation of the Immune System in Health and Disease by Members of the Bone Morphogenetic Protein Family. Front Immunol 2021; 12:802346. [PMID: 34925388 PMCID: PMC8674571 DOI: 10.3389/fimmu.2021.802346] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/17/2021] [Indexed: 12/17/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are potent signaling molecules initially described as osteopromoting proteins. BMPs represent one of the members of the larger TGFβ family and today are recognized for their important role in numerous processes. Among the wide array of functions recently attributed to them, BMPs were also described to be involved in the regulation of components of the innate and adaptive immune response. This review focuses on the signaling pathway of BMPs and highlights the effects of BMP signaling on the differentiation, activation, and function of the main cell types of the immune system.
Collapse
Affiliation(s)
| | - Giuseppe Sconocchia
- Institute of Translational Pharmacology, National Research Council (CNR), Rome, Italy
| |
Collapse
|
16
|
Ollewagen T, Powrie YSL, Myburgh KH, Smith C. Unresolved intramuscular inflammation, not diminished skeletal muscle regenerative capacity, is at the root of rheumatoid cachexia: insights from a rat CIA model. Physiol Rep 2021; 9:e15119. [PMID: 34806343 PMCID: PMC8606867 DOI: 10.14814/phy2.15119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 11/24/2022] Open
Abstract
Rheumatoid arthritis targets numerous organs in patients, including the skeletal muscle, resulting in rheumatoid cachexia. In the muscle niche, satellite cells, macrophages, and myofibroblasts may be affected and the factors they release altered. This study aimed to assess these cell types, cytokines, and growth factors and their relationships to muscle fiber size and number in a rodent collagen-induced arthritis (CIA) model, in order to identify new therapeutic targets. Fiber cross-sectional area (CSA) was 57% lower in CIA than controls (p < 0.0001), thus smaller but more fibers visible per field of view. Immunostaining indicated the increased presence of satellite cells, macrophages, myofibroblasts, and myonuclei per field of view in CIA (p < 0.01), but this finding was not maintained when taking fiber number into consideration. Western blots of gastrocnemius samples indicated that tumor necrosis factor-α was significantly elevated (p < 0.01) while interleukin-10 (IL-10) was decreased (p < 0.05) in CIA. This effect was maintained (and heightened for IL-10) when expressed per fiber number. Myogenic regulatory factors (MyoD and myogenin), transforming growth factor-β and inhibitor of differentiation were significantly elevated in CIA muscle and levels correlated significantly with CSA. Several of these factors remained elevated, but bone morphogenetic protein-7 decreased when considering fiber number per area. In conclusion, CIA-muscle demonstrated a good regenerative response. Myoblast numbers per fiber were not elevated, suggesting their activity results from the persistent inflammatory signaling which also significantly hampered maintenance of muscle fiber size. A clearer picture of signaling events at cellular level in arthritis muscle may be derived from expressing data per fiber.
Collapse
Affiliation(s)
- Tracey Ollewagen
- Department Physiological SciencesScience FacultyStellenbosch UniversityStellenboschSouth Africa
| | - Yigael S. L. Powrie
- Division of Clinical PharmacologyDepartment of MedicineFaculty of Medicine and Health SciencesStellenbosch UniversityStellenboschSouth Africa
| | - Kathryn H. Myburgh
- Department Physiological SciencesScience FacultyStellenbosch UniversityStellenboschSouth Africa
| | - Carine Smith
- Division of Clinical PharmacologyDepartment of MedicineFaculty of Medicine and Health SciencesStellenbosch UniversityStellenboschSouth Africa
| |
Collapse
|
17
|
Elmadbouh I, Singla DK. BMP-7 Attenuates Inflammation-Induced Pyroptosis and Improves Cardiac Repair in Diabetic Cardiomyopathy. Cells 2021; 10:2640. [PMID: 34685620 PMCID: PMC8533936 DOI: 10.3390/cells10102640] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 01/15/2023] Open
Abstract
In the present study, we investigated a novel signaling target in diabetic cardiomyopathy where inflammation induces caspase-1-dependent cell death, pyroptosis, involving Nek7-GBP5 activators to activate the NLRP3 inflammasome, destabilizes cardiac structure and neovascularization. Furthermore, we explored the therapeutic ability of bone morphogenetic protein-7 (BMP-7) to attenuate these adverse effects. C57BL/6J mice (n = 16 mice/group) were divided into: control (200 mg/kg, 0.9% saline intraperitoneal injection, i.p.); Streptozotocin (STZ) and STZ-BMP-7 groups (STZ, 200 mg/kg, i.p. injection). After 6 weeks, heart function was examined with echocardiography, and mice were sacrificed. Immunostaining, Western blotting, H&E, and Masson's trichrome staining was performed on heart tissues. STZ-induced diabetic cardiomyopathy significantly increased inflammasome formation (TLR4, NLRP3, Nek7, and GBP5), pyroptosis markers (caspase-1, IL-1β, and IL-18), inflammatory cytokines (IL-6 and TNF-α), MMP9, and infiltration of monocytes (CD14), macrophage (iNOS), and dendritic cells (CD11b and CD11c) (p < 0.05). Moreover, a significant endothelial progenitor cells (EPCs) dysfunction (c-Kit/FLk-1, CD31), adverse cardiac remodeling, and reduction in left ventricular (LV) heart function were observed in STZ versus control (p < 0.05). Treatment with BMP-7 significantly reduced inflammasome formation, pyroptosis, and inflammatory cytokines and infiltrated inflammatory cells. In addition, BMP-7 treatment enhanced EPC markers and neovascularization and subsequently improved cardiac remodeling in a diabetic heart. Moreover, a significant improvement in LV heart function was achieved after BMP-7 administration relative to diabetic mice (p < 0.05). In conclusion, BMP-7 attenuated inflammation-induced pyroptosis, adverse cardiac remodeling, and improved heart function via the TLR4-NLRP3 inflammasome complex activated by novel signaling Nek7/GBP5. Our BMP-7 pre-clinical studies of mice could have significant potential as a future therapy for diabetic patients.
Collapse
Affiliation(s)
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
18
|
Kosyreva A, Dzhalilova D, Lokhonina A, Vishnyakova P, Fatkhudinov T. The Role of Macrophages in the Pathogenesis of SARS-CoV-2-Associated Acute Respiratory Distress Syndrome. Front Immunol 2021; 12:682871. [PMID: 34040616 PMCID: PMC8141811 DOI: 10.3389/fimmu.2021.682871] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophages are cells that mediate both innate and adaptive immunity reactions, playing a major role in both physiological and pathological processes. Systemic SARS-CoV-2-associated complications include acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation syndrome, edema, and pneumonia. These are predominantly effects of massive macrophage activation that collectively can be defined as macrophage activation syndrome. In this review we focus on the role of macrophages in COVID-19, as pathogenesis of the new coronavirus infection, especially in cases complicated by ARDS, largely depends on macrophage phenotypes and functionalities. We describe participation of monocytes, monocyte-derived and resident lung macrophages in SARS-CoV-2-associated ARDS and discuss possible utility of cell therapies for its treatment, notably the use of reprogrammed macrophages with stable pro- or anti-inflammatory phenotypes.
Collapse
Affiliation(s)
- Anna Kosyreva
- Department of Neuromorphology, Science Research Institute of Human Morphology, Moscow, Russia
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
| | - Dzhuliia Dzhalilova
- Department of Immunomorphology of Inflammation, Science Research Institute of Human Morphology, Moscow, Russia
| | - Anastasia Lokhonina
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Polina Vishnyakova
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Timur Fatkhudinov
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Growth and Development, Science Research Institute of Human Morphology, Moscow, Russia
| |
Collapse
|
19
|
Sconocchia T, Hochgerner M, Schwarzenberger E, Tam-Amersdorfer C, Borek I, Benezeder T, Bauer T, Zyulina V, Painsi C, Passegger C, Wolf P, Sibilia M, Strobl H. Bone morphogenetic protein signaling regulates skin inflammation via modulating dendritic cell function. J Allergy Clin Immunol 2021; 147:1810-1822.e9. [PMID: 33250156 DOI: 10.1016/j.jaci.2020.09.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/22/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Bone morphogenetic proteins (BMPs) are members of the TGF-β family that signal via the BMP receptor (BMPR) signaling cascade, distinct from canonical TGF-β signaling. BMP downstream signaling is strongly induced within epidermal keratinocytes in cutaneous psoriatic lesions, and BMP7 instructs monocytic cells to acquire characteristics of psoriasis-associated Langerhans dendritic cells (DCs). Regulatory T (Treg)-cell numbers strongly increase during psoriatic skin inflammation and were recently shown to limit psoriatic skin inflammation. However, the factors mediating Treg-cell accumulation in psoriatic skin currently remain unknown. OBJECTIVE We sought to investigate the role of BMP signaling in Treg-cell accumulation in psoriasis. METHODS The following methods were used: immunohistology of patients and healthy controls; ex vivo models of Treg-cell generation in the presence or absence of Langerhans cells; analysis of BMP versus canonical TGF-β signaling in DCs and Treg cells; and modeling of psoriatic skin inflammation in mice lacking the BMPR type 1a in CD11c+ cells. RESULTS We here demonstrated a positive correlation between Treg-cell numbers and epidermal BMP7 expression in cutaneous psoriatic lesions and show that unlike Treg cells from healthy skin, a portion of inflammation-associated Treg cells exhibit constitutive-active BMP signaling. We further found that BMPR signaling licenses inflammation-associated Langerhans cell/DC to gain an enhanced capacity to promote Treg cells via BMPR-mediated CD25 induction and that this effect is associated with reduced skin inflammation. CONCLUSIONS Psoriatic lesions are marked by constitutive high BMP7/BMPR signaling in keratinocytes, which instructs inflammatory DCs to gain enhanced Treg-cell-stimulatory activity. Locally secreted BMP7 can directly promote Treg-cell generation through the BMP signaling cascade.
Collapse
Affiliation(s)
- Tommaso Sconocchia
- Otto Loewi Research Center, Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Mathias Hochgerner
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Elke Schwarzenberger
- Otto Loewi Research Center, Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Carmen Tam-Amersdorfer
- Otto Loewi Research Center, Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Izabela Borek
- Otto Loewi Research Center, Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Theresa Benezeder
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Thomas Bauer
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Victoria Zyulina
- Otto Loewi Research Center, Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Clemens Painsi
- Department of Dermatology, State Hospital Klagenfurt, Klagenfurt, Austria
| | - Christina Passegger
- Otto Loewi Research Center, Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Peter Wolf
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Otto Loewi Research Center, Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
20
|
Aluganti Narasimhulu C, Singla DK. Amelioration of diabetes-induced inflammation mediated pyroptosis, sarcopenia, and adverse muscle remodelling by bone morphogenetic protein-7. J Cachexia Sarcopenia Muscle 2021; 12:403-420. [PMID: 33463042 PMCID: PMC8061343 DOI: 10.1002/jcsm.12662] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/14/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Diabetic myopathy involves hyperglycaemia and inflammation that causes skeletal muscle dysfunction; however, the potential cellular mechanisms that occur between hyperglycaemia and inflammation, which induces sarcopenia, and muscle dysfunction remain unknown. In this study, we investigated hyperglycaemia-induced inflammation mediating high-mobility group box 1 activation, which is involved in a novel form of cell death, pyroptosis, diabetic sarcopenia, atrophy, and adverse muscle remodelling. Furthermore, we investigated the therapeutic potential of bone morphogenetic protein-7 (BMP-7), an osteoporosis drug, to treat pyroptosis, and diabetic muscle myopathy. METHODS C57BL6 mice were treated with saline (control), streptozotocin (STZ), or STZ + BMP-7 to generate diabetic muscle myopathy. Diabetes was established by determining the increased levels of glucose. Then, muscle function was examined, and animals were sacrificed. Gastrocnemius muscle or blood samples were analysed for inflammation, pyroptosis, weight loss, muscle atrophy, and adverse structural remodelling of gastrocnemius muscle using histology, enzyme-linked immunosorbent assay, immunohistochemistry, western blotting, and reverse transcription polymerase chain reaction. RESULTS A significant (P < 0.05) increase in hyperglycaemia leads to an increase in inflammasome (high-mobility group box 1, toll-like receptor-4, and nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing protein 3) formation in diabetic muscle cells. Further analysis showed an up-regulation of the downstream pyroptotic pathway with significant (P < 0.05) number of positive muscle cells expressing pyroptosis-specific markers [caspase-1, interleukin (IL)-1β, IL-18, and gasdermin-D]. Pyroptotic cell death is involved in further increasing inflammation by releasing pro-inflammatory cytokine IL-6. Structural analysis showed the loss of muscle weight, decreased myofibrillar area, and increased fibrosis leading to muscle dysfunction. Consistent with this finding, BMP-7 attenuated hyperglycaemia (~50%), pyroptosis, inflammation, and diabetic adverse structural modifications as well as improved muscle function. CONCLUSIONS In conclusion, we report for the first time that increased hyperglycaemia and inflammation involve cellular pyroptosis that induces significant muscle cell loss and adverse remodelling in diabetic myopathy. We also report that targeting pyroptosis with BMP-7 improves diabetic muscle pathophysiology and muscle function. These findings suggest that BMP-7 could be a potential therapeutic option to treat diabetic myopathy.
Collapse
Affiliation(s)
- Chandrakala Aluganti Narasimhulu
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
21
|
Gu X, Li SY, DeFalco T. Immune and vascular contributions to organogenesis of the testis and ovary. FEBS J 2021; 289:2386-2408. [PMID: 33774913 PMCID: PMC8476657 DOI: 10.1111/febs.15848] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/07/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Gonad development is a highly regulated process that coordinates cell specification and morphogenesis to produce sex-specific organ structures that are required for fertility, such as testicular seminiferous tubules and ovarian follicles. While sex determination occurs within specialized gonadal supporting cells, sexual differentiation is evident throughout the entire organ, including within the interstitial compartment, which contains immune cells and vasculature. While immune and vascular cells have been traditionally appreciated for their supporting roles during tissue growth and homeostasis, an increasing body of evidence supports the idea that these cell types are critical drivers of sexually dimorphic morphogenesis of the gonad. Myeloid immune cells, such as macrophages, are essential for multiple aspects of gonadogenesis and fertility, including for forming and maintaining gonadal vasculature in both sexes at varying stages of life. While vasculature is long known for supporting organ growth and serving as an export mechanism for gonadal sex steroids in utero, it is also an important component of fetal testicular morphogenesis and differentiation; additionally, it is vital for ovarian corpus luteal function and maintenance of pregnancy. These findings point toward a new paradigm in which immune cells and blood vessels are integral components of sexual differentiation and organogenesis. In this review, we discuss the state of the field regarding the diverse roles of immune and vascular cells during organogenesis of the testis and ovary and highlight outstanding questions in the field that could stimulate new research into these previously underappreciated constituents of the gonad.
Collapse
Affiliation(s)
- Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
22
|
Cao H, Quan S, Zhang L, Chen Y, Jiao G. BMPR2 expression level is correlated with low immune infiltration and predicts metastasis and poor survival in osteosarcoma. Oncol Lett 2021; 21:391. [PMID: 33777214 PMCID: PMC7988701 DOI: 10.3892/ol.2021.12652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most common malignant bone tumor in adolescents and young adults, and identifying biomarkers for prognosis and therapy is necessary. Bone morphogenetic protein receptor 2 (BMPR2) is involved in various cellular functions, including cell adhesion, proliferation and invasion, inflammation, apoptosis and metastatic spread. However, the correlation between BMPR2 expression levels and prognosis and tumor-infiltrating immune cells in osteosarcoma is not well understood. In the present study, the expression level of BMPR2 was investigated using the Oncomine and R2 databases. The association between the expression level of BMPR2 and the clinical prognosis of patients with cancer was analyzed using the R2 database. The relationship between the expression level of BMPR2 and immune cell infiltration in the stroma of osteosarcoma was assessed using the Tumor Immune Estimation Resource (TIMER) and CIBERSORT. The correlations between BMPR2 expression level and infiltrated immune cell gene marker sets in osteosarcoma were validated in the TIMER and R2 databases. Analysis of a cohort of patients with osteosarcoma revealed that BMPR2 expression was significantly higher in osteosarcoma compared with in normal tissue and was correlated with poor prognosis. M0 macrophages, M2 macrophages, resting mast, γ δ T and CD8+ T cells were the top five immune cells with the highest degrees of infiltration in osteosarcoma. In addition, BMPR2 expression level showed a significant negative correlation with the gene markers of CD8+ T cells, monocytes and M2 macrophages. Low levels of infiltrating CD8+ T cells, monocytes or M2 macrophages in osteosarcoma was significantly associated with poor survival. These data suggested that CD8+ T cells, monocytes and M2 macrophages play significant roles in the establishment of the immune microenvironment of osteosarcoma. High BMPR2 expression was associated with poor prognosis and low infiltration of CD8+ T cells, monocytes and M2 macrophages in osteosarcoma. Hence, BMPR2 can be considered a biomarker of the immune infiltration, metastasis and prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Hongxin Cao
- Department of Medical Oncology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Key Laboratory of Chemical Biology, Ministry of Education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Shuang Quan
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lu Zhang
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Spine and Spinal Cord Disease Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yunzhen Chen
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Spine and Spinal Cord Disease Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Guangjun Jiao
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China.,Spine and Spinal Cord Disease Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
23
|
Caron MMJ, Ripmeester EGJ, van den Akker G, Wijnands NKAP, Steijns J, Surtel DAM, Cremers A, Emans PJ, van Rhijn LW, Welting TJM. Discovery of bone morphogenetic protein 7-derived peptide sequences that attenuate the human osteoarthritic chondrocyte phenotype. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:247-261. [PMID: 33850953 PMCID: PMC8022858 DOI: 10.1016/j.omtm.2021.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/09/2021] [Indexed: 01/18/2023]
Abstract
Treatment of osteoarthritis (OA) is mainly symptomatic by alleviating pain to postpone total joint replacement. Bone morphogenetic protein 7 (BMP7) is a candidate morphogen for experimental OA treatment that favorably alters the chondrocyte and cartilage phenotype. Intra-articular delivery and sustained release of a recombinant growth factor for treating OA are challenging, whereas the use of peptide technology potentially circumvents many of these challenges. In this study, we screened a high-resolution BMP7 peptide library and discovered several overlapping peptide sequences from two regions in BMP7 with nanomolar bioactivity that attenuated the pathological OA chondrocyte phenotype. A single exposure of OA chondrocytes to peptides p[63-82] and p[113-132] ameliorated the OA chondrocyte phenotype for up to 8 days, and peptides were bioactive on chondrocytes in OA synovial fluid. Peptides p[63-82] and p[113-132] required NKX3-2 for their bioactivity on chondrocytes and provoke changes in SMAD signaling activity. The bioactivity of p[63-82] depended on specific evolutionary conserved sequence elements common to BMP family members. Intra-articular injection of a rat medial meniscal tear (MMT) model with peptide p[63-82] attenuated cartilage degeneration. Together, this study identified two regions in BMP7 from which bioactive peptides are able to attenuate the OA chondrocyte phenotype. These BMP7-derived peptides provide potential novel disease-modifying treatment options for OA.
Collapse
Affiliation(s)
- Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Ellen G J Ripmeester
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Guus van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Nina K A P Wijnands
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Jessica Steijns
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Don A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Pieter J Emans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.,Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Lodewijk W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.,Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.,Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| |
Collapse
|
24
|
Kim JY, Lim S, Lim HS, Kim YS, Eun KM, Khalmuratova R, Seo Y, Kim JK, Kim YS, Kim MK, Jin S, Han SC, Pyo S, Hong SN, Park JW, Shin HW, Kim DW. Bone morphogenetic protein-2 as a novel biomarker for refractory chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2021; 148:461-472.e13. [PMID: 33667477 DOI: 10.1016/j.jaci.2021.02.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Bone morphogenetic proteins (BMPs), which are members of the TGF-β superfamily, regulate bone remodeling by stimulating osteoblasts and osteoclasts. Although the association between osteitis and poor surgical outcomes is well known in patients with chronic rhinosinusitis (CRS), BMPs have not been fully investigated as potential biomarkers for the prognosis of CRS. OBJECTIVE Our aim was to investigate the role of BMPs in osteitis in patients with CRS with nasal polyps (NPs) (CRSwNPs), as well as associations between BMPs and inflammatory markers in sinonasal tissues from patients with CRSwNP. METHODS We investigated the expression of 6 BMPs (BMP-2, BMP-4, BMP-6, BMP-7, BMP-9, and BMP-10) and their cellular origins in NPs of human subjects by using immunohistochemistry and ELISA of NP tissues. Exploratory factor analysis was performed to identify associations between BMPs and inflammatory markers. Air-liquid interface cell culture of human nasal epithelial cells was performed to evaluate the induction of the epithelial-mesenchymal transition by BMPs. RESULTS Of the 6 BMPs studied, BMP-2 and BMP-7 were associated with refractoriness. Only BMP-2 concentrations were higher in patients with severe osteitis and advanced disease extent according to the computed tomography findings. Eosinophils and some macrophages were identified as cellular sources of BMP-2 in immunofluorescence analysis. An in vitro experiment revealed that BMP-2 induced epithelial-mesenchymal transition in air-liquid interface-cultured human nasal epithelial cells, particularly in a TH2 milieu. CONCLUSION BMP-2 could reflect the pathophysiology of mucosa and bone remodeling and may be a novel biomarker for refractory CRSwNP.
Collapse
Affiliation(s)
- Jin Youp Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea; Interdisciplinary Program of Medical Informatics, Seoul National University College of Medicine, Seoul, Korea
| | - Suha Lim
- Obstructive Upper Airway Research Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hee-Suk Lim
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Yi-Sook Kim
- Obstructive Upper Airway Research Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Kyoung Mi Eun
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Roza Khalmuratova
- Obstructive Upper Airway Research Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea
| | - Yuju Seo
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Joon Kon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Young Seok Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Kyung Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Siyeon Jin
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Cheol Han
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Suyeon Pyo
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Seung-No Hong
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Obstructive Upper Airway Research Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Woo Shin
- Obstructive Upper Airway Research Laboratory, Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea; Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, Korea; Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Korea.
| | - Dae Woo Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
25
|
Descamps E, Molto A, Borderie D, Lories R, Richard CM, Pons M, Roux C, Briot K. Changes in bone formation regulator biomarkers in early axial spondyloarthritis. Rheumatology (Oxford) 2021; 60:1185-1194. [PMID: 32888036 DOI: 10.1093/rheumatology/keaa296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 03/24/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The hallmark of advanced axial SpA (axSpA) is spine ankylosis due to excessive ectopic bone formation. This prospective study aimed to describe the changes in serum levels of different regulators [sclerostin, dickkopf-1 (DKK-1)] and markers of bone formation [bone morphogenetic protein 7 (BMP-7)] over 5 years in early axSpA patients and to assess determinants of such changes. METHODS The DEvenir des Spondyloarthropathies Indifférenciées Récentes cohort is a prospective, multicentre French study of 708 patients with early (>3 months-<3 years) inflammatory back pain suggestive of axSpA. Serum levels of BMP-7, sclerostin and DKK-1 were assessed at baseline and after 2 and 5 years. Changes in bone formation regulators over time were analysed using mixed linear models. RESULTS Serum BMP-7 significantly increased over time, with a median relative change of 223.7% [interquartile range (IQR) 0-10 700 (0.17 pg/ml/month), P < 0.001]. Serum sclerostin significantly increased over time, with a median relative change of 14.8% [IQR -7.9-41.4% (0.001 ng/ml/month), P < 0.001]. Serum DKK-1 did not significantly change over time. Serum BMP-7 increased over time in active disease (Ankylosing Spondylitis Disease Activity Score with CRP ≥1.3, P = 0.01), but the increase was less pronounced with TNF inhibitor (TNFi) use (P < 0.001). No determinant was associated with serum sclerostin change. CONCLUSION Serum BMP-7 change over 5 years was related with inflammation; it was increased in active disease, but the increase was low with TNFi use. Serum sclerostin levels significantly increased over time, but to a lesser degree than for serum BMP-7. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/, NCT01648907.
Collapse
Affiliation(s)
- Elise Descamps
- Department of Rheumatology, Cochin Hospital and Epidemiology and Biostatistics Unit, Sorbonne Paris Cite Research Center, Paris Descartes University, INSERM U1153, Paris, France
| | - Anna Molto
- Department of Rheumatology, Cochin Hospital and Epidemiology and Biostatistics Unit, Sorbonne Paris Cite Research Center, Paris Descartes University, INSERM U1153, Paris, France
| | - Didier Borderie
- Department of Biology, Cochin Hospital, Paris Descartes University, INSERM U1153, Paris, France
| | - Rik Lories
- KU Leuven and Division of Rheumatology, Skeletal Biology and Engineering Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Corinne Miceli Richard
- Department of Rheumatology, Cochin Hospital and Epidemiology and Biostatistics Unit, Sorbonne Paris Cite Research Center, Paris Descartes University, INSERM U1153, Paris, France
| | - Marion Pons
- Department of Rheumatology, Cochin Hospital and Epidemiology and Biostatistics Unit, Sorbonne Paris Cite Research Center, Paris Descartes University, INSERM U1153, Paris, France
| | - Christian Roux
- Department of Rheumatology, Cochin Hospital and Epidemiology and Biostatistics Unit, Sorbonne Paris Cite Research Center, Paris Descartes University, INSERM U1153, Paris, France
| | - Karine Briot
- Department of Rheumatology, Cochin Hospital and Epidemiology and Biostatistics Unit, Sorbonne Paris Cite Research Center, Paris Descartes University, INSERM U1153, Paris, France
| |
Collapse
|
26
|
Dessouki FBA, Kukreja RC, Singla DK. Stem Cell-Derived Exosomes Ameliorate Doxorubicin-Induced Muscle Toxicity through Counteracting Pyroptosis. Pharmaceuticals (Basel) 2020; 13:ph13120450. [PMID: 33316945 PMCID: PMC7764639 DOI: 10.3390/ph13120450] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 12/21/2022] Open
Abstract
Doxorubicin (Dox)-induced muscle toxicity (DIMT) is a common occurrence in cancer patients; however, the cause of its development and progression is not established. We tested whether inflammation-triggered cell death, “pyroptosis” plays a role in DIMT. We also examined the potential role of exosomes derived from embryonic stem cells (ES-Exos) in attenuating DIMT. C57BL/6J mice (10 ± 2 wks age) underwent the following treatments: Control (saline), Dox, Dox+ES-Exos, and Dox+MEF-Exos (mouse-embryonic fibroblast-derived exosomes, negative control). Our results demonstrated that Dox significantly reduced muscle function in mice, which was associated with a significant increase in NLRP3 inflammasome and initiation marker TLR4 as compared with controls. Pyroptosis activator, ASC, was significantly increased compared to controls with an upregulation of specific markers (caspase-1, IL-1β, and IL-18). Treatment with ES-Exos but not MEF-Exos showed a significant reduction in inflammasome and pyroptosis along with improved muscle function. Additionally, we detected a significant increase in pro-inflammatory cytokines (TNF-α and IL-6) and inflammatory M1 macrophages in Dox-treated animals. Treatment with ES-Exos decreased M1 macrophages and upregulated anti-inflammatory M2 macrophages. Furthermore, ES-Exos showed a significant reduction in muscular atrophy and fibrosis. In conclusion, these results suggest that DIMT is mediated through inflammation and pyroptosis, which is attenuated following treatment with ES-Exos.
Collapse
Affiliation(s)
- Fatima Bianca A. Dessouki
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
| | - Rakesh C. Kukreja
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
- Correspondence: ; Tel.: +1-401-823-0953
| |
Collapse
|
27
|
Cortez MA, Masrorpour F, Ivan C, Zhang J, Younes AI, Lu Y, Estecio MR, Barsoumian HB, Menon H, Caetano MDS, Ramapriyan R, Schoenhals JE, Wang X, Skoulidis F, Wasley MD, Calin G, Hwu P, Welsh JW. Bone morphogenetic protein 7 promotes resistance to immunotherapy. Nat Commun 2020; 11:4840. [PMID: 32973129 PMCID: PMC7519103 DOI: 10.1038/s41467-020-18617-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/26/2020] [Indexed: 11/24/2022] Open
Abstract
Immunotherapies revolutionized cancer treatment by harnessing the immune system to target cancer cells. However, most patients are resistant to immunotherapies and the mechanisms underlying this resistant is still poorly understood. Here, we report that overexpression of BMP7, a member of the TGFB superfamily, represents a mechanism for resistance to anti-PD1 therapy in preclinical models and in patients with disease progression while on immunotherapies. BMP7 secreted by tumor cells acts on macrophages and CD4+ T cells in the tumor microenvironment, inhibiting MAPK14 expression and impairing pro-inflammatory responses. Knockdown of BMP7 or its neutralization via follistatin in combination with anti-PD1 re-sensitizes resistant tumors to immunotherapies. Thus, we identify the BMP7 signaling pathway as a potential immunotherapeutic target in cancer. The mechanisms underlying resistance to immunotherapy are still poorly understood. Here, the authors show that BMP7, a molecule part of the TGF-beta superfamily, suppresses proinflammatory antitumor responses and may represent a target for overcoming resistance to PD1 inhibitors.
Collapse
Affiliation(s)
- Maria Angelica Cortez
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Fatemeh Masrorpour
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristina Ivan
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie Zhang
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmed I Younes
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yue Lu
- Epigenetic and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcos R Estecio
- Epigenetic and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hampartsoum B Barsoumian
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hari Menon
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mauricio da Silva Caetano
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rishab Ramapriyan
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jonathan E Schoenhals
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaohong Wang
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ferdinandos Skoulidis
- Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark D Wasley
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George Calin
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick Hwu
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
28
|
Hellwinkel JE, Miclau T, Provencher MT, Bahney CS, Working ZM. The Life of a Fracture: Biologic Progression, Healing Gone Awry, and Evaluation of Union. JBJS Rev 2020; 8:e1900221. [PMID: 32796195 PMCID: PMC11147169 DOI: 10.2106/jbjs.rvw.19.00221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
New knowledge about the molecular biology of fracture-healing provides opportunities for intervention and reduction of risk for specific phases that are affected by disease and medications. Modifiable and nonmodifiable risk factors can prolong healing, and the informed clinician should optimize each patient to provide the best chance for union. Techniques to monitor progression of fracture-healing have not changed substantially over time; new objective modalities are needed.
Collapse
Affiliation(s)
- Justin E Hellwinkel
- Department of Orthopedic Surgery, New York Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Theodore Miclau
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Matthew T Provencher
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
| | - Chelsea S Bahney
- Center for Regenerative Sports Medicine, The Steadman Clinic and Steadman Philippon Research Institute, Vail, Colorado
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
| | - Zachary M Working
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF) and Zuckerberg San Francisco General Hospital (ZSFG), San Francisco, California
- Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
29
|
Aluganti Narasimhulu C, Singla DK. The Role of Bone Morphogenetic Protein 7 (BMP-7) in Inflammation in Heart Diseases. Cells 2020; 9:cells9020280. [PMID: 31979268 PMCID: PMC7073173 DOI: 10.3390/cells9020280] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
Bone morphogenetic protein-7 is (BMP-7) is a potent anti-inflammatory growth factor belonging to the Transforming Growth Factor Beta (TGF-β) superfamily. It plays an important role in various biological processes, including embryogenesis, hematopoiesis, neurogenesis and skeletal morphogenesis. BMP-7 stimulates the target cells by binding to specific membrane-bound receptor BMPR 2 and transduces signals through mothers against decapentaplegic (Smads) and mitogen activated protein kinase (MAPK) pathways. To date, rhBMP-7 has been used clinically to induce the differentiation of mesenchymal stem cells bordering the bone fracture site into chondrocytes, osteoclasts, the formation of new bone via calcium deposition and to stimulate the repair of bone fracture. However, its use in cardiovascular diseases, such as atherosclerosis, myocardial infarction, and diabetic cardiomyopathy is currently being explored. More importantly, these cardiovascular diseases are associated with inflammation and infiltrated monocytes where BMP-7 has been demonstrated to be a key player in the differentiation of pro-inflammatory monocytes, or M1 macrophages, into anti-inflammatory M2 macrophages, which reduces developed cardiac dysfunction. Therefore, this review focuses on the molecular mechanisms of BMP-7 treatment in cardiovascular disease and its role as an anti-fibrotic, anti-apoptotic and anti-inflammatory growth factor, which emphasizes its potential therapeutic significance in heart diseases.
Collapse
|
30
|
Meyer JG, Garcia TY, Schilling B, Gibson BW, Lamba DA. Proteome and Secretome Dynamics of Human Retinal Pigment Epithelium in Response to Reactive Oxygen Species. Sci Rep 2019; 9:15440. [PMID: 31659173 PMCID: PMC6817852 DOI: 10.1038/s41598-019-51777-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/04/2019] [Indexed: 12/22/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in developed countries, and is characterized by slow retinal degeneration linked to chronic reactive oxygen species (ROS) in the retinal pigmented epithelium (RPE). The molecular mechanisms leading to RPE dysfunction in response to ROS are unclear. Here, human stem cell-derived RPE samples were stressed with ROS for 1 or 3 weeks, and both intracellular and secreted proteomes were quantified by mass spectrometry. ROS increased glycolytic proteins but decreased mitochondrial complex I subunits, as well as membrane proteins required for endocytosis. RPE secreted over 1,000 proteins, many of which changed significantly due to ROS. Notably, secreted APOE is decreased 4-fold, and urotensin-II, the strongest known vasoconstrictor, doubled. Furthermore, secreted TGF-beta is increased, and its cognate signaler BMP1 decreased in the secretome. Together, our results paint a detailed molecular picture of the retinal stress response in space and time.
Collapse
Affiliation(s)
- Jesse G Meyer
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Department of Chemistry, Department of Biomolecular Chemistry, National Center for Quantitative Biology of Complex Systems, University of Wisconsin - Madison, Madison, WI, 53706, USA.
| | - Thelma Y Garcia
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Bradford W Gibson
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Discovery Attribute Sciences, Research, Amgen, South San Francisco, CA, 94080, USA
| | - Deepak A Lamba
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Department of Ophthalmology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California - San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
31
|
Lahr CA, Wagner F, Shafiee A, Rudert M, Hutmacher DW, Holzapfel BM. Recombinant Human Bone Morphogenetic Protein 7 Exerts Osteo-Catabolic Effects on Bone Grafts That Outweigh Its Osteo-Anabolic Capacity. Calcif Tissue Int 2019; 105:331-340. [PMID: 31214730 DOI: 10.1007/s00223-019-00574-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/07/2019] [Indexed: 11/29/2022]
Abstract
This study aimed to investigate the effects of recombinant human bone morphogenetic protein (rhBMP-7) on human cancellous bone grafts (BGs) while differentiating between anabolic and catabolic events. Human BGs alone or supplemented with rhBMP-7 were harvested 14 weeks after subcutaneous implantation into NOD/Scid mice, and studied via micro-CT, histomorphometry, immunohistochemistry and flow cytometry. Immunohistochemical staining for human-specific proteins made it possible to differentiate between grafted human bone and newly formed murine bone. Only BGs implanted with rhBMP-7 formed an ossicle containing a functional hematopoietic compartment. The total ossicle volume in the BMP+ group was higher than in the BMP- group (835 mm3 vs. 365 mm3, respectively, p < 0.001). The BMP+ group showed larger BM spaces (0.47 mm vs. 0.28 mm, p = 0.002) and lower bone volume-to-total volume ratio (31% vs. 47%, p = 0.002). Immunohistochemical staining for human-specific proteins confirmed a higher ratio of newly formed bone area (murine) to total area (0.12 vs. 0.001, p < 0.001) in the BMP+ group, while the ratio of grafted bone (human) area to total area was smaller (0.14 vs. 0.34, p = 0.004). The results demonstrate that rhBMP-7 induces BG resorption at a higher rate than new bone formation while creating a haematopoietic niche. Clinicians therefore need to consider the net catabolic effect when rhBMP-7 is used with BGs. Overall, this model indicates its promising application to further decipher BMPs action on BGs and its potential in complex bone tissue regeneration.
Collapse
Affiliation(s)
- Christoph A Lahr
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia
- Department of Orthopaedic Surgery, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstrasse 11, 97074, Wuerzburg, Germany
| | - Ferdinand Wagner
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Abbas Shafiee
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Maximilian Rudert
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Dietmar W Hutmacher
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Boris Michael Holzapfel
- Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, QLD, 4059, Australia.
- Department of Orthopaedic Surgery, University of Wuerzburg, Koenig-Ludwig-Haus, Brettreichstrasse 11, 97074, Wuerzburg, Germany.
| |
Collapse
|
32
|
Acute Lymphoblastic Leukaemia Cells Impair Dendritic Cell and Macrophage Differentiation: Role of BMP4. Cells 2019; 8:cells8070722. [PMID: 31337120 PMCID: PMC6679123 DOI: 10.3390/cells8070722] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/06/2019] [Accepted: 07/13/2019] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells and macrophages are common components of the tumour immune microenvironment and can contribute to immune suppression in both solid and haematological cancers. The Bone Morphogenetic Protein (BMP) pathway has been reported to be involved in cancer, and more recently in leukaemia development and progression. In the present study, we analyse whether acute lymphoblastic leukaemia (ALL) cells can affect the differentiation of dendritic cells and macrophages and the involvement of BMP pathway in the process. We show that ALL cells produce BMP4 and that conditioned media from ALL cells promote the generation of dendritic cells with immunosuppressive features and skew M1-like macrophage polarization towards a less pro-inflammatory phenotype. Likewise, BMP4 overexpression in ALL cells potentiates their ability to induce immunosuppressive dendritic cells and favours the generation of M2-like macrophages with pro-tumoral features. These results suggest that BMP4 is in part responsible for the alterations in dendritic cell and macrophage differentiation produced by ALL cells.
Collapse
|
33
|
Li L, Jiang Y, Lin H, Shen H, Sohn J, Alexander PG, Tuan RS. Muscle injury promotes heterotopic ossification by stimulating local bone morphogenetic protein-7 production. J Orthop Translat 2019; 18:142-153. [PMID: 31508317 PMCID: PMC6718974 DOI: 10.1016/j.jot.2019.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 02/01/2023] Open
Abstract
Background Heterotopic ossification (HO) is a pathological condition of abnormal bone formation in soft tissue, which causes pain and restricted range of motion in patients. There are two broad categories of HO, hereditary and acquired. Although different types of HO do not use identical mechanistic pathways of pathogenesis, muscle injury appears to be a unifying feature for all types of HO. However, little is known about the mechanisms by which muscle injury facilitates HO formation. Objective and method This study aimed to explore the cellular and molecular mechanisms linking muscle injury to HO by using cardiotoxin to induce muscle injury in a bone morphogenetic protein-2 (BMP-2)-induced HO mouse model. Results We found that muscle injury augmented HO formation and that this effect was correlated with BMP signalling activation and upregulation of BMP-7 expression at the early phase of HO progression. We further demonstrated that inhibition of BMP-7 activity in vitro suppressed the osteogenesis-promoting effect of conditioned medium derived from injured muscle tissue and in vivo reduced the volume of HO formation. We also showed that antiinflammatory drug treatment reduced the volume of HO with concomitant reduction in BMP-7 production. Conclusion In summary, our study has identified BMP-7 as a key osteoinductive factor in injured muscle that facilitates HO formation. The translational potential of this article Our results provide a candidate mechanistic rationale for the use of antiinflammatory drugs in the prevention of HO.
Collapse
Affiliation(s)
- La Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Cellular and Molecular Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yangzi Jiang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - He Shen
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jihee Sohn
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peter G. Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Cellular and Molecular Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Corresponding author. Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
34
|
West J, Chen X, Yan L, Gladson S, Loyd J, Rizwan H, Talati M. Adverse effects of BMPR2 suppression in macrophages in animal models of pulmonary hypertension. Pulm Circ 2019; 10:2045894019856483. [PMID: 31124398 PMCID: PMC7074495 DOI: 10.1177/2045894019856483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/17/2019] [Indexed: 01/11/2023] Open
Abstract
Inflammatory cells contribute to irreversible damage in pulmonary arterial hypertension (PAH). We hypothesized that in PAH, dysfunctional BMPR2 signaling in macrophages contributes to pulmonary vascular injury and phenotypic changes via proinflammatory cytokine production. Studies were conducted in: (1) Rosa26-rtTA2 3 X TetO7-Bmpr2delx4 FVB/N mice (mutant Bmpr2 is universally expressed, BMPR2delx4 mice) given a weekly intra-tracheal liposomal clodronate injections for four weeks; and (2) LysM-Cre X floxed BMPR2 X floxed eGFP monocyte lineage-specific BMPR2 knockout (KO) mouse model (Bmpr2 gene expression knockdown in monocytic lineage cells) (BMPR2KO) following three weeks of sugen/hypoxia treatment. In the BMPR2delx4 mice, increased right ventricular systolic pressure (RVSP; P < 0.05) was normalized by clodronate, and in monocyte lineage-specific BMPR2KO mice sugen hypoxia treatment increased (P < 0.05) RVSP compared to control littermates, suggesting that suppressed BMPR2 in macrophages modulate RVSP in animal models of PH. In addition, in these mouse models, muscularized pulmonary vessels were increased (P < 0.05) and surrounded by an increased number of macrophages. Elimination of macrophages in BMPR2delx4 mice reduced the number of muscularized pulmonary vessels and macrophages surrounding these vessels. Further, in monocyte lineage-specific BMPR2KO mice, there was significant increase in proinflammatory cytokines, including C-X-C Motif Chemokine Ligand 12 (CXCL12), complement component 5 a (C5a), Interleukin-16 (IL-16), and secretory ICAM. C5a positive inflammatory cells present in and around the pulmonary vessels in the PAH lung could potentially be involved in pulmonary vessel remodeling. In summary, our data indicate that, in BMPR2-related PAH, macrophages with dysfunctional BMPR2 influence pulmonary vascular remodeling and phenotypic outcomes via proinflammatory cytokine production.
Collapse
Affiliation(s)
- James West
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Xinping Chen
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ling Yan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Santhi Gladson
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James Loyd
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hamid Rizwan
- Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Megha Talati
- Division of Respiratory and Critical Care, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
35
|
IL-37 suppresses the sustained hepatic IFN-γ/TNF-α production and T cell-dependent liver injury. Int Immunopharmacol 2019; 69:184-193. [DOI: 10.1016/j.intimp.2019.01.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/11/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
|
36
|
Exosome-integrated titanium oxide nanotubes for targeted bone regeneration. Acta Biomater 2019; 86:480-492. [PMID: 30630122 DOI: 10.1016/j.actbio.2019.01.006] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 01/02/2019] [Accepted: 01/06/2019] [Indexed: 12/13/2022]
Abstract
Exosomes are extracellular nanovesicles that play an important role in cellular communication. The modulatory effects of bone morphogenetic protein 2 (BMP2) on macrophages have encouraged the functionalization of scaffolds through the integration of the exosomes from the BMP2-stimulated macrophages to avoid ectopic bone formation and reduce adverse effects. To determine the functionality of exosomal nanocarriers from macrophages after BMP2 stimulation, we isolated the exosomes from Dulbecco's modified Eagle's medium (DMEM)- or BMP2-stimulated macrophages and evaluated their effects on osteogenesis. Morphological characterization of the exosomes derived from DMEM- or BMP2-treated macrophages revealed no significant differences, and the bone marrow-derived mesenchymal stromal cells showed similar cellular uptake patterns for both exosomes. In vitro study using BMP2/macrophage-derived exosomes indicated their beneficial effects on osteogenic differentiation. To improve the bio-functionality for titanium implants, BMP2/macrophage-derived exosomes were used to modify titanium nanotube implants to favor osteogenesis. The incorporation of BMP2/macrophage-derived exosomes dramatically increased the expression of early osteoblastic differentiation markers, alkaline phosphatase (ALP) and BMP2, indicative of the pro-osteogenic role of the titanium nanotubes incorporated with BMP2/macrophage-derived exosomes. The titanium nanotubes functionalized with BMP2/macrophage-derived exosomes activated autophagy during osteogenic differentiation. In conclusion, the exosome-integrated titanium nanotube may serve as an emerging functional material for bone regeneration. STATEMENT OF SIGNIFICANCE: The clinical application of bone morphogenetic protein 2 (BMP2) is often limited by its side effects. Exosomes are naturally secreted nanosized vesicles derived from cells and play an important role in intercellular communication. The contributions of this study include (1) the demonstration of the potential regulatory role of BMP2/macrophage-derived exosomes on the osteogenic differentiation of mesenchymal stromal cells (MSCs); (2) fabrication of titanium nanotubes incorporated with exosomes; (3) new insights into the application of titanium nanotube-based materials for the safe use of BMP2.
Collapse
|
37
|
M2 Macrophages as a Potential Target for Antiatherosclerosis Treatment. Neural Plast 2019; 2019:6724903. [PMID: 30923552 PMCID: PMC6409015 DOI: 10.1155/2019/6724903] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammation course, which could induce life-threatening diseases such as stroke and myocardial infarction. Optimal medical treatments for atherosclerotic risk factors with current antihypertensive and lipid-lowering drugs (for example, statins) are widely used in clinical practice. However, many patients with established disease still continue to have recurrent cardiovascular events in spite of treatment with a state-of-the-art therapy. Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of mortality worldwide. Hence, current treatment of atherosclerosis is still far from being satisfactory. Recently, M2 macrophages have been found associated with atherosclerosis regression. The M2 phenotype can secrete anti-inflammatory factors such as IL-10 and TGF-β, promote tissue remodeling and repairing through collagen formation, and clear dying cells and debris by efferocytosis. Therefore, modulators targeting macrophages' polarization to the M2 phenotype could be another promising treatment strategy for atherosclerosis. Two main signaling pathways, the Akt/mTORC/LXR pathway and the JAK/STAT6 pathway, are found playing important roles in M2 polarization. In addition, researchers have reported several potential approaches to modulate M2 polarization. Inhibiting or activating some kinds of enzymes, affecting transcription factors, or acting on several membrane receptors could regulate the polarization of the M2 phenotype. Besides, biomolecules, for example vitamin D, were found to affect the process of M2 polarization. Pomegranate juice could promote M2 polarization via unclear mechanism. In this review, we will discuss how M2 macrophages affect atherosclerosis regression, signal transduction in M2 polarization, and outline potential targets and compounds that affect M2 polarization, thus controlling the progress of atherosclerosis.
Collapse
|
38
|
Lu L, Zhu J, Zhang Y, Wang Y, Zhang S, Xia A. Febuxostat inhibits TGF‑β1‑induced epithelial‑mesenchymal transition via downregulation of USAG‑1 expression in Madin‑Darby canine kidney cells in vitro. Mol Med Rep 2019; 19:1694-1704. [PMID: 30628645 PMCID: PMC6390060 DOI: 10.3892/mmr.2019.9806] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/04/2018] [Indexed: 01/06/2023] Open
Abstract
Our previous study demonstrated that febuxostat, a xanthine oxidase inhibitor, can alleviate kidney dysfunction and ameliorate renal tubulointerstitial fibrosis in a rat unilateral ureteral obstruction (UUO) model; however, the underlying mechanisms remain unknown. Increasing evidence has revealed that epithelial-mesenchymal transition (EMT) is one of the key mechanisms mediating the progression of renal tubulointerstitial fibrosis in chronic kidney disease (CKD). Uterine sensitization-associated gene-1 (USAG-1), a kidney-specific bone morphogenetic protein antagonist, is involved in the development of numerous types of CKDs. The present study aimed to investigate the role of febuxostat in the process of EMT in Madin-Darby canine kidney (MDCK) cells in vitro. Western blotting, reverse transcription-semiquantitative polymerase chain reaction analysis and immunofluorescence staining were used to evaluate the expression levels of bone morphogenetic protein 7, USAG-1, α-smooth muscle actin (α-SMA) and E-cadherin, respectively. The results demonstrated that the expression of USAG-1 and α-SMA increased, and that of E-cadherin decreased significantly in MDCK cells following treatment with transforming growth factor-β1 (TGF-β1). The application of small interfering RNA-USAG-1 potently inhibited TGF-β1-induced EMT. Subsequently, the effects of febuxostat on TGF-β1-induced EMT was investigated. The results demonstrated that febuxostat downregulated the expression of USAG-1, and reversed TGF-β1-induced EMT in MDCK cells. Furthermore, pretreatment with febuxostat significantly restored the decreased expression levels of phosphorylated Smad1/5/8 induced by TGF-β1 in MDCK cells. The results of the present study suggested that USAG-1 may be involved in the EMT process of MDCK cells induced by TGF-β1, and febuxostat inhibited EMT by activating the Smad1/5/8 signaling pathway via downregulating the expression of USAG-1 in MDCK cells.
Collapse
Affiliation(s)
- Linghong Lu
- Department of Pharmacology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jiajun Zhu
- Department of Anesthesiology, Guanyun County People's Hospital, Lianyungang, Jiangsu 222200, P.R. China
| | - Yaqian Zhang
- Department of Pharmacology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Yanxia Wang
- Department of Pharmacology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Shu Zhang
- Department of Pharmacology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Anzhou Xia
- Department of Pharmacology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
39
|
Shoulders H, Garner KH, Singla DK. Macrophage depletion by clodronate attenuates bone morphogenetic protein-7 induced M2 macrophage differentiation and improved systolic blood velocity in atherosclerosis. Transl Res 2019; 203:1-14. [PMID: 30107156 PMCID: PMC6314201 DOI: 10.1016/j.trsl.2018.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022]
Abstract
Bone morphogenetic protein-7 (BMP-7) affects the presence of macrophage subtypes in vitro and in vivo at an early stage of atherosclerosis (ATH); however, it remains unknown whether BMP-7 treatment affects the development and progression of ATH at a mid-stage of the disease. We therefore performed a Day 28 (D28) study to examine BMP-7's potential to affect monocyte differentiation. Atherosclerosis was developed in ApoE KO mice, and these animals were treated with intravenous injections of BMP-7 and/or liposomal clodronate (LC). BMP-7 significantly (P < 0.05) lowers plaque formation following induction of atherosclerosis. However, upon macrophage depletion, BMP-7 fails to significantly alter plaque progression suggesting a direct role of BMP-7 on macrophages. Immunohistochemical staining of carotid arteries was performed to determine BMP-7's effect on pro-inflammatory M1 inducible nitric oxide synthase and anti-inflammatory M2 (cluster of differentiation [CD]206, Arginase-1) macrophages, and monocytes ( CD14). BMP-7 significantly reduced pro-inflammatory M1 macrophages and increased anti-inflammatory M2 macrophages at D28, while BMP-7 showed no effect on M2 macrophage differentiation in animals treated with LC. Enzyme-linked immunosorbent assay data showed significant reduction in proinflammatory cytokines (Interleukin-6 [IL-6]), monocyte chemoattractant protein-1, and tumor necrosis factor-α) and a significant increase in anti-inflammatory cytokine (IL-10) in BMP-7 treated mice (P < 0.05).Western blot analysis of arterial tissue confirms a significant increase in pro-survival kinases extracellular-signal regulated kinase and SMAD and a reduction in pro-inflammatory kinases p38 and c-Jun N-terminal kinase in BMP-7 treated mice (P < 0.05). Overall, this study suggests that clodronate treatment inhibits BMP-7 induced differentiation of monocytes into M2 macrophages and improved systolic blood velocity.
Collapse
Affiliation(s)
- Heidi Shoulders
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Kaley H Garner
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida.
| |
Collapse
|
40
|
Johnson TA, Singla DK. PTEN inhibitor VO-OHpic attenuates inflammatory M1 macrophages and cardiac remodeling in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol 2018; 315:H1236-H1249. [PMID: 30095997 PMCID: PMC6297808 DOI: 10.1152/ajpheart.00121.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 02/06/2023]
Abstract
Doxorubicin (Doxo) is an effective agent commonly used in cancer therapeutics. Unfortunately, Doxo treatment can stimulate cardiomyopathy and subsequent heart failure, limiting the use of this drug. The role of phosphatase and tensin homolog (PTEN) in apoptosis has been documented in Doxo-induced cardiomyopathy (DIC) and heart failure models. However, whether direct inhibition of PTEN attenuates apoptosis, cardiac remodeling, and inflammatory M1 macrophages in the DIC model remains elusive. Therefore, the present study was designed to understand the effects of VO-OHpic (VO), a potent inhibitor of PTEN, in reducing apoptosis and cardiac remodeling. At day 56, echocardiography was performed, which showed that VO treatment significantly ( P < 0.05) improved heart function. Immunohistochemistry, TUNEL, and histological staining were used to determine apoptosis, proinflammatory M1 macrophages, anti-inflammatory M2 macrophages, and cardiac remodeling. Our data show a significant increase in apoptosis, hypertrophy, fibrosis, and proinflammatory M1 macrophages with Doxo treatment, whereas VO treatment significantly reduced apoptosis, adverse cardiac remodeling, and proinflammatory M1 macrophages significantly ( P < 0.05) compared with the Doxo-treated group. Western blot analysis confirmed the reduction of phosphorylated PTEN and increase in phosphorylated AKT protein expression in the Doxo + VO-treated group. Moreover, VO administration increased anti-inflammatory M2 macrophages. Collectively, our data suggest that VO treatment attenuates apoptosis and adverse cardiac remodeling, a process that is mediated through the PTEN/AKT pathway, resulting in improved heart function in DIC. NEW & NOTEWORTHY Doxorubicin-induced cardiomyopathy (DIC) is still a major issue in patients with cancer. These novel findings on the phosphatase and tensin homolog inhibitor VO-OHpic in DIC is the first report, as per the best of our knowledge, that VO-OHpic significantly decreases apoptosis, fibrosis, hypertrophy, adverse cardiac remodeling, and proinflammatory M1 macrophages and increases anti-inflammatory M2 macrophages along with significantly improved cardiac function. VO-OHpic could be a future therapeutic agent for patients with DIC.
Collapse
Affiliation(s)
- Taylor A Johnson
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida , Orlando, Florida
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida , Orlando, Florida
| |
Collapse
|
41
|
Adamson SE, Polanowska-Grabowska R, Marqueen K, Griffiths R, Angdisen J, Breevoort SR, Schulman IG, Leitinger N. Deficiency of Dab2 (Disabled Homolog 2) in Myeloid Cells Exacerbates Inflammation in Liver and Atherosclerotic Plaques in LDLR (Low-Density Lipoprotein Receptor)-Null Mice-Brief Report. Arterioscler Thromb Vasc Biol 2018; 38:1020-1029. [PMID: 29599136 PMCID: PMC5920703 DOI: 10.1161/atvbaha.117.310467] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 03/06/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Inflammatory macrophages promote the development of atherosclerosis. We have identified the adaptor protein Dab2 (disabled homolog 2) as a regulator of phenotypic polarization in macrophages. The absence of Dab2 in myeloid cells promotes an inflammatory phenotype, but the impact of myeloid Dab2 deficiency on atherosclerosis has not been shown. APPROACH AND RESULTS To determine the role of myeloid Dab2 in atherosclerosis, Ldlr-/- mice were reconstituted with either Dab2-positive or Dab2-deficient bone marrow and fed a western diet. Consistent with our previous finding that Dab2 inhibits NFκB (nuclear factor κ-light-chain-enhancer of activated B cells) signaling in macrophages, Ldlr-/- mice reconstituted with Dab2-deficient bone marrow had increased systemic inflammation as evidenced by increased serum IL-6 (interleukin-6) levels and increased inflammatory cytokine expression levels in liver. Serum lipid levels were significantly lower in Ldlr-/- mice reconstituted with Dab2-deficient bone marrow, and further examination of livers from these mice revealed drastically increased inflammatory tissue damage and massive infiltration of immune cells. Surprisingly, the atherosclerotic lesion burden in Ldlr-/- mice reconstituted with Dab2-deficient bone marrow was decreased compared with Ldlr-/- mice reconstituted with wild-type bone marrow. Further analysis of aortic root sections revealed increased macrophage content and evidence of increased apoptosis in lesions from Ldlr-/- mice reconstituted with Dab2-deficient bone marrow but no difference in collagen or α-smooth muscle actin content. CONCLUSIONS Dab2 deficiency in myeloid cells promotes inflammation in livers and atherosclerotic plaques in a mouse model of atherosclerosis. Nevertheless, decreased serum lipids as a result of massive inflammatory liver damage may preclude an appreciable increase in atherosclerotic lesion burden in mice reconstituted with Dab2-deficient bone marrow.
Collapse
Affiliation(s)
- Samantha E Adamson
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| | - Renata Polanowska-Grabowska
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| | - Kathryn Marqueen
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Rachael Griffiths
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| | - Jerry Angdisen
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Sarah R Breevoort
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Ira G Schulman
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
| | - Norbert Leitinger
- From the Department of Pharmacology (S.E.A., R.P.-G., K.M., R.G., J.A., S.R.B., I.G.S., N.L.)
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville (S.E.A., R.P.-G., R.G., N.L.)
| |
Collapse
|
42
|
Mast Cells Exert Anti-Inflammatory Effects in an IL10 -/- Model of Spontaneous Colitis. Mediators Inflamm 2018; 2018:7817360. [PMID: 29849494 PMCID: PMC5932457 DOI: 10.1155/2018/7817360] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/27/2018] [Accepted: 03/04/2018] [Indexed: 12/14/2022] Open
Abstract
Mast cells are well established as divergent modulators of inflammation and immunosuppression, but their role in inflammatory bowel disease (IBD) remains to be fully defined. While previous studies have demonstrated a proinflammatory role for mast cells in acute models of chemical colitis, more recent investigations have shown that mast cell deficiency can exacerbate inflammation in spontaneous colitis models, thus suggesting a potential anti-inflammatory role of mast cells in IBD. Here, we tested the hypothesis that in chronic, spontaneous colitis, mast cells are protective. We compared colitis and intestinal barrier function in IL10−/− mice to mast cell deficient/IL10−/− (double knockout (DKO): KitWsh/Wsh × IL10−/−) mice. Compared with IL10−/− mice, DKO mice exhibited more severe colitis as assessed by increased colitis scores, mucosal hypertrophy, intestinal permeability, and colonic cytokine production. PCR array analyses demonstrated enhanced expression of numerous cytokine and chemokine genes and downregulation of anti-inflammatory genes (e.g., Tgfb2, Bmp2, Bmp4, Bmp6, and Bmp7) in the colonic mucosa of DKO mice. Systemic reconstitution of DKO mice with bone marrow-derived mast cells resulted in significant amelioration of IL10−/−-mediated colitis and intestinal barrier injury. Together, the results presented here demonstrate that mast cells exert anti-inflammatory properties in an established model of chronic, spontaneous IBD. Given the previously established proinflammatory role of mast cells in acute chemical colitis models, the present findings provide new insight into the divergent roles of mast cells in modulating inflammation during different stages of colitis. Further investigation of the mechanism of the anti-inflammatory role of the mast cells may elucidate novel therapies.
Collapse
|
43
|
Wei F, Zhou Y, Wang J, Liu C, Xiao Y. The Immunomodulatory Role of BMP-2 on Macrophages to Accelerate Osteogenesis. Tissue Eng Part A 2018; 24:584-594. [DOI: 10.1089/ten.tea.2017.0232] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Fei Wei
- The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| | - Yinghong Zhou
- The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| | - Jing Wang
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Changsheng Liu
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Yin Xiao
- The Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- The Australia-China Center for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
44
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
45
|
Curaj A, Wu Z, Rix A, Gresch O, Sternkopf M, Alampour-Rajabi S, Lammers T, van Zandvoort M, Weber C, Koenen RR, Liehn EA, Kiessling F. Molecular Ultrasound Imaging of Junctional Adhesion Molecule A Depicts Acute Alterations in Blood Flow and Early Endothelial Dysregulation. Arterioscler Thromb Vasc Biol 2017; 38:40-48. [PMID: 29191926 DOI: 10.1161/atvbaha.117.309503] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 11/17/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The junctional adhesion molecule A (JAM-A) is physiologically located in interendothelial tight junctions and focally redistributes to the luminal surface of blood vessels under abnormal shear and flow conditions accompanying atherosclerotic lesion development. Therefore, JAM-A was evaluated as a target for molecularly targeted ultrasound imaging of transient endothelial dysfunction under acute blood flow variations. APPROACH AND RESULTS Flow-dependent endothelial dysfunction was induced in apolipoprotein E-deficient mice (n=43) by carotid partial ligation. JAM-A expression was investigated by molecular ultrasound using antibody-targeted poly(n-butyl cyanoacrylate) microbubbles and validated with immunofluorescence. Flow disturbance and arterial remodeling were assessed using functional ultrasound. Partial ligation led to an immediate drop in perfusion at the ligated side and a direct compensatory increase at the contralateral side. This was accompanied by a strongly increased JAM-A expression and JAM-A-targeted microbubbles binding at the partially ligated side and by a moderate and temporary increase in the contralateral artery (≈14× [P<0.001] and ≈5× [P<0.001] higher than control, respectively), both peaking after 2 weeks. Subsequently, although JAM-A expression and JAM-A-targeted microbubbles binding persisted at a higher level at the partially ligated side, it completely normalized within 4 weeks at the contralateral side. CONCLUSIONS Temporary blood flow variations induce endothelial rearrangement of JAM-A, which can be visualized using JAM-A-targeted microbubbles. Thus, JAM-A may be considered as a marker of acute endothelial activation and dysfunction. Its imaging may facilitate the early detection of cardiovascular risk areas, and it enables the therapeutic prevention of their progression toward an irreversible pathological state.
Collapse
Affiliation(s)
- Adelina Curaj
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Zhuojun Wu
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Anne Rix
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Oliver Gresch
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Marieke Sternkopf
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Setareh Alampour-Rajabi
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Twan Lammers
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Marc van Zandvoort
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Christian Weber
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Rory R Koenen
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Elisa A Liehn
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.)
| | - Fabian Kiessling
- From the Institute for Molecular Cardiovascular Research (IMCAR) (A.C., Z.W., M.S., S.A.-R., M.v.Z., E.A.L.), and Institute for Experimental Molecular Imaging (ExMI) (A.C., Z.W., A.R., T.L., F.K.), University Hospital Aachen, RWTH Aachen, Germany; Victor Babes National Institute of Pathology, Bucharest, Romania (A.C.); AYOXXA Biosystems GmbH, Cologne, Germany (O.G.); Department of Targeted Therapeutics, University of Twente, Enschede, The Netherlands (T.L.); Department of Genetics and Molecular Cell Biology, School for Oncology and Developmental Biology (GROW), Maastricht University, The Netherlands (M.v.Z., R.R.K.); Department of Biochemistry, School for Cardiovascular Diseases (CARIM), Maastricht University, The Netherlands (M.v.Z., C.W.); German Centre for Cardiovascular Research, partner site Munich Heart Alliance (DZHK), Germany (C.W.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Munich, Germany (C.W.); and Human Genetic Laboratory, University for Medicine and Pharmacy, Craiova, Romania (E.A.L.).
| |
Collapse
|
46
|
Identification of Key Pathways and Genes in Advanced Coronary Atherosclerosis Using Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4323496. [PMID: 29226137 PMCID: PMC5684517 DOI: 10.1155/2017/4323496] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/17/2017] [Indexed: 01/02/2023]
Abstract
Background Coronary artery atherosclerosis is a chronic inflammatory disease. This study aimed to identify the key changes of gene expression between early and advanced carotid atherosclerotic plaque in human. Methods Gene expression dataset GSE28829 was downloaded from Gene Expression Omnibus (GEO), including 16 advanced and 13 early stage atherosclerotic plaque samples from human carotid. Differentially expressed genes (DEGs) were analyzed. Results 42,450 genes were obtained from the dataset. Top 100 up- and downregulated DEGs were listed. Functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) identification were performed. The result of functional and pathway enrichment analysis indicted that the immune system process played a critical role in the progression of carotid atherosclerotic plaque. Protein-protein interaction (PPI) networks were performed either. Top 10 hub genes were identified from PPI network and top 6 modules were inferred. These genes were mainly involved in chemokine signaling pathway, cell cycle, B cell receptor signaling pathway, focal adhesion, and regulation of actin cytoskeleton. Conclusion The present study indicated that analysis of DEGs would make a deeper understanding of the molecular mechanisms of atherosclerosis development and they might be used as molecular targets and diagnostic biomarkers for the treatment of atherosclerosis.
Collapse
|
47
|
Liu G, Liu Q, Shen Y, Kong D, Gong Y, Tao B, Chen G, Guo S, Li J, Zuo S, Yu Y, Yin H, Zhang L, Zhou B, Funk CD, Zhang J, Yu Y. Early treatment with Resolvin E1 facilitates myocardial recovery from ischaemia in mice. Br J Pharmacol 2017; 175:1205-1216. [PMID: 28925017 DOI: 10.1111/bph.14041] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/21/2017] [Accepted: 09/06/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE An appropriate inflammatory response is necessary for cardiac healing after acute myocardial infarction (MI). Resolvin E1 (RvE1) is an anti-inflammatory and pro-resolution lipid mediator derived from eicosapentaenoic acid. Here we have investigated the effects of RvE1 on the recovery of cardiac function after MI in mice. EXPERIMENTAL APPROACH Acute MI was induced by surgical ligation of the left anterior descending artery in male C57BL/6 mice. RvE1 (5 ng·g-1 ·day-1 ; i.p.) was given to mice at different times following MI. Cardiac function was monitored by transthoracic echocardiography at days 3, 7 and 14 after MI. Effects of RvE1 on the migration of subpopulations of monocytes/macrophages (Mos/Mps, Ly6Chi and Ly6Clow ) were examined by flow cytometry and transwell assay. KEY RESULTS RvE1 administration from days 1 to 7 post-MI improved cardiac function, whereas treatment from days 7 to 14 markedly inhibited recovery of cardiac function. Early treatment with RvE1 post-MI suppressed the infiltration of dominant Ly6Chi Mos/Mps and secretion of pro-inflammatory cytokines in injured hearts, which protected cardiomyocytes against apoptosis in the peri-infarct zones. Contrastingly, treatment with RvE1 1 week after MI decreased infiltration of Ly6Clow Mos/Mps and expression of pro-angiogenic factors in cardiac tissue, consequently reducing neovascularization in the peri-infarct zones. Additionally, RvE1 inhibited Mp migration by activating ChemR23 receptors. CONCLUSION AND IMPLICATIONS Treatment with RvE1 during the initial 7 days after MI facilitated cardiac healing by suppressing pro-inflammatory cytokine secretion, indicating that RvE1 may serve as an early therapeutic agent for acute MI. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Guizhu Liu
- Key Laboratory of Food Safety Research, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qian Liu
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yujun Shen
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Deping Kong
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yanjun Gong
- Key Laboratory of Food Safety Research, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Bo Tao
- Key Laboratory of Food Safety Research, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Guilin Chen
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shumin Guo
- Key Laboratory of Food Safety Research, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Juanjuan Li
- Department of Gastroenterology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shengkai Zuo
- Key Laboratory of Food Safety Research, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Yu
- Key Laboratory of Food Safety Research, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Colin D Funk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L3N6, Canada
| | - Jian Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ying Yu
- Department of Pharmacology, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
48
|
Dietary teasaponin ameliorates alteration of gut microbiota and cognitive decline in diet-induced obese mice. Sci Rep 2017; 7:12203. [PMID: 28939875 PMCID: PMC5610180 DOI: 10.1038/s41598-017-12156-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/30/2017] [Indexed: 12/14/2022] Open
Abstract
A high-fat (HF) diet alters gut microbiota and promotes obesity related inflammation and cognitive impairment. Teasaponin is the major active component of tea, and has been associated with anti-inflammatory effects and improved microbiota composition. However, the potential protective effects of teasaponin, against HF diet-induced obesity and its associated alteration of gut microbiota, inflammation and cognitive decline have not been studied. In this study, obesity was induced in C57BL/6 J male mice by feeding a HF diet for 8 weeks, followed by treatment with oral teasaponin (0.5%) mixed in HF diet for a further 6 weeks. Teasaponin treatment prevented the HF diet-induced recognition memory impairment and improved neuroinflammation, gliosis and brain-derived neurotrophic factor (BDNF) deficits in the hippocampus. Furthermore, teasaponin attenuated the HF diet-induced endotoxemia, pro-inflammatory macrophage accumulation in the colon and gut microbiota alterations. Teasaponin also improved glucose tolerance and reduced body weight gain in HF diet-induced obese mice. The behavioral and neurochemical improvements suggest that teasaponin could limit unfavorable gut microbiota alterations and cognitive decline in HF diet-induced obesity.
Collapse
|
49
|
Chen F, Chen R, Liu H, Sun R, Huang J, Huang Z, Jian G. BMP-7 ameliorates cobalt alloy particle-induced inflammation by suppressing Th17 responses. APMIS 2017; 125:880-887. [PMID: 28736908 DOI: 10.1111/apm.12730] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/07/2017] [Indexed: 02/05/2023]
Abstract
Metal wear debris has been shown to activate an aseptic osteolytic process that causes failure in total joint arthroplasty (TJA). This osteolysis is characterized by a proinflammatory, self-propagating immune response involving primarily macrophages, dendritic cells, and activated osteoclasts, as well as T cells and B cells. The human bone morphogenic protein (BMP)-7, on the other hand, was shown to promote osteoblast survival, and reversed the downregulation of anabolic Smad proteins and Runx2 following cobalt injury. Therefore, we investigated the effect and mechanism of BMP-7 on the proinflammatory immune responses in osteoarthritis patients with previous TJA. Cobalt-treated monocytes/macrophages presented significantly elevated levels of interleukin 6 (IL-6) and tumor necrosis factor (TNF), both of which were suppressed by the addition of exogenous BMP-7. In patients with TJA, the serum BMP-7 level was inversely associated with the level of IL-6 and TNF secreted by monocytes/macrophages. Cobalt-treated monocytes/macrophages effectively supported Th17 inflammation, by an IL-6-dependent but not TNF-dependent mechanism. BMP-7, however, significantly suppressed cobalt-induced Th17 inflammation. In patients with TJA, the risk of osteolysis development was positively associated with the frequency of Th17 cells and negatively associated with the level of BMP-7. Together, these results demonstrated that BMP-7 could serve as a therapeutic agent in treating patients with metal wear debris-induced inflammation.
Collapse
Affiliation(s)
- Fengrong Chen
- Department of Orthopedics, Chenggong Hospital, Xiamen University, Xiamen, China
| | - Ruisong Chen
- Department of Orthopedics, Chenggong Hospital, Xiamen University, Xiamen, China
| | - Haoyuan Liu
- Department of Orthopedics, Chenggong Hospital, Xiamen University, Xiamen, China
| | - Rupeng Sun
- Department of Orthopedics, Chenggong Hospital, Xiamen University, Xiamen, China
| | - Jianming Huang
- Department of Orthopedics, Chenggong Hospital, Xiamen University, Xiamen, China
| | - Zheyuan Huang
- Department of Orthopedics, Chenggong Hospital, Xiamen University, Xiamen, China
| | - Guojian Jian
- Department of Orthopedics, Chenggong Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
50
|
Peiró C, Lorenzo Ó, Carraro R, Sánchez-Ferrer CF. IL-1β Inhibition in Cardiovascular Complications Associated to Diabetes Mellitus. Front Pharmacol 2017; 8:363. [PMID: 28659798 PMCID: PMC5468794 DOI: 10.3389/fphar.2017.00363] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 05/26/2017] [Indexed: 01/15/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic disease that affects nowadays millions of people worldwide. In adults, type 2 diabetes mellitus (T2DM) accounts for the majority of all diagnosed cases of diabetes. The course of the T2DM is characterized by insulin resistance and a progressive loss of β-cell mass. DM is associated with a number of related complications, among which cardiovascular complications and atherosclerosis are the main cause of morbidity and mortality in patients suffering from the disease. DM is acknowledged as a low-grade chronic inflammatory state characterized by the over-secretion of pro-inflammatory cytokines, including interleukin (IL)-1β, which reinforce inflammatory signals thus contributing to the development of complications. In this context, the pharmacological approaches to treat diabetes should not only correct hyperglycaemia, but also attenuate inflammation and prevent the development of metabolic and cardiovascular complications. Over the last years, novel biological drugs have been developed to antagonize the pathophysiological actions of IL-1β. The drugs currently used in clinical practice are anakinra, a recombinant form of the naturally occurring IL-1 receptor antagonist, the soluble decoy receptor rilonacept and the monoclonal antibodies canakinumab and gevokizumab. This review will summarize the main experimental and clinical findings obtained with pharmacological IL-1β inhibitors in the context of the cardiovascular complications of DM, and discuss the perspectives of IL-1β inhibitors as novel therapeutic tools for treating these patients.
Collapse
Affiliation(s)
- Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ)Madrid, Spain
| | - Óscar Lorenzo
- Department of Medicine, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Fundación Jiménez DíazMadrid, Spain
| | - Raffaele Carraro
- Department of Medicine, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Service of Endocrinology, Hospital de La PrincesaMadrid, Spain.,Instituto de Investigación Sanitaria Hospital de La PrincesaMadrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ)Madrid, Spain
| |
Collapse
|