1
|
Sun C, Ding Z, Li B, Chen S, Li E, Yang Q. New insights into Gremlin-1: A tumour microenvironment landscape re-engineer and potential therapeutic target. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119962. [PMID: 40250712 DOI: 10.1016/j.bbamcr.2025.119962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Gremlin-1 (GREM1), a well-known bone morphogenetic protein (BMP) antagonist, is highly expressed in various malignant tumours. However, the specific role of GREM1 in tumours remains controversial and may be attributed to the heterogeneity and complexity of the tumour microenvironment (TME). It is currently believed that GREM1 regulates the complex landscape of the TME, primarily by antagonising BMP signalling or BMP-independent pathways. Both GREM1 and BMP play dual roles in tumour progression. Therefore, the mutual crosstalk between tumour cells and tumour-associated fibroblasts and the regulation of various secreted factors in the TME affect the secretion level of GREM1, which in turn regulates the amplitude balance between GREM1 and BMP, affecting tumour progression. The inhibition of GREM1 activity in the TME can disrupt this amplitude balance and prevent the formation of a tumour-supportive microenvironment, demonstrating that GREM1 is a potential therapeutic target. In this study, we reviewed the specific signalling pathways via which GREM1 in the TME regulates epithelial-mesenchymal transition, construction of the tumour immune microenvironment, and maintenance of tumour cell stemness via BMP-dependent and BMP-independent regulation, and also summarised the latest clinical progress of GREM1.
Collapse
Affiliation(s)
- Chengpeng Sun
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang 330006, China; HuanKui Academy, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zijun Ding
- School of Ophthalmology and Optometry, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Benjie Li
- Queen Mary School, Jiangxi Medical college, Nanchang University, Nanchang 330031, China
| | - Sihong Chen
- Queen Mary School, Jiangxi Medical college, Nanchang University, Nanchang 330031, China
| | - Enliang Li
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang 330006, China; Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, China.
| | - Qingping Yang
- Department of Reproductive Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17, Yongwai zheng Street, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
2
|
Jin Z, Cao Y. Gremlin1: a BMP antagonist with therapeutic potential in Oncology. Invest New Drugs 2024; 42:716-727. [PMID: 39347850 DOI: 10.1007/s10637-024-01474-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
Gremlins, originating from early 20th-century Western folklore, are mythical creatures known for causing mechanical malfunctions and electronic failures, aptly dubbed "little devils". Analogously, GREM1 acts like a horde of these mischievous entities by antagonizing the bone morphogenetic protein (BMP signaling) pathway or through other non-BMP dependent mechanisms (such as binding to Fibroblast Growth Factor Receptor 1and Epidermal Growth Factor Receptor) contributing to the malignant progression of various cancers. The overexpression of GREM1 promotes tumor cell growth and survival, enhances angiogenesis within the tumor microenvironment, and creates favorable conditions for tumor development and dissemination. Consequently, inhibiting the activity of GREM1 or blocking its interaction with BMP presents a promising strategy for suppressing tumor growth and metastasis. However, the role of GREM1 in cancer remains a subject of debate, with evidence suggesting both oncogenic and tumor-suppressive functions. Currently, several pharmaceutical companies are researching the GREM1 target, with some advancing to Phase I/II clinical trials. This article will provide a detailed overview of the GREM1 target and explore its potential role in cancer therapy.
Collapse
Affiliation(s)
- Zhao Jin
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
3
|
Unal Kocabas G, Furundaoturan O, Degırmencı C, Ozgur S, Kocamanoglu M, Arslan CG, Pehlivan Koroglu E, Afrashı F, Akcay Y, Yildirim Simsir I, Sarer Yurekli B. Changes in retinal microvasculature and serum Gremlin-1 levels in acromegaly: A case-control study. Photodiagnosis Photodyn Ther 2024; 50:104415. [PMID: 39592020 DOI: 10.1016/j.pdpdt.2024.104415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVE There are controversial results about the effects of GH/IGF-1 on retinal vasculature. Gremlin-1 has roles in both pituitary tumorigenesis and angiogenesis. Therefore, we aimed to detect retinal micro-vascular changes in acromegaly with OCTA and examine its association with serum Gremlin-1 levels. DESIGN A cross-sectional, case-control study METHODS: We included 53 acromegaly patients and age/gender-matched 31 healthy controls. To evaluate retinal structure, optical coherence tomography angiography (OCTA) imaging was performed. Gremlin -1 levels were measured by the ELISA method. RESULTS Acromegaly patients had significantly lower superficial total density (p< 0.001), superficial parafoveal density (p = 0.002), superficial perifoveal density (p < 0.001); deep total density (p < 0.001), deep parafoveal density (p < 0.001), deep perifoveal density (p < 0.001). Total thickness and perifoveal thickness were significantly lower in the acromegaly group. Gremlin-1 levels were significantly lower in acromegaly patients (1.22(0.13) vs 1.38(0.29); p = 0.015, for acromegaly and control groups, respectively). We assessed study groups based on glucose tolerance status. Normal glucose tolerance (NGT) acromegaly patients had significantly lower superficial total density (p = 0.036), superficial perifoveal density (p = 0.003), deep total density (p = 0.023), and deep perifoveal density (p = 0.023) compared to NGT controls. According to backward linear regression analysis, the presence of acromegaly itself consistently showed a significant negative impact on all types of vascular density. CONCLUSION Patients with acromegaly have decreased vascular density and lower levels of Gremlin-1 independent of glucose tolerance status. Acromegaly may cause a reduction in gremlin-1 as a compensatory mechanism due to high IGF-1 levels known as an angiogenic factor, which in turn leads to the decrease in vascular density, or gremlin-1 may already have shown a decline in response to chronic inflammation and endothelial dysfunction in acromegaly resulting in a reduction in vascular density.
Collapse
Affiliation(s)
- Gokcen Unal Kocabas
- Ege University School of Medicine, Internal Medicine Department, Endocrinology and Metabolism Unit, Turkey.
| | | | | | - Su Ozgur
- Regional Hub for Cancer Registration in Northern Africa, Central and Western Asia, WHO/IARC GICR, Turkey
| | - Meltem Kocamanoglu
- Ege University School of Medicine, Medical Biochemistry Department, Turkey
| | | | - Esma Pehlivan Koroglu
- Ege University School of Medicine, Internal Medicine Department, Endocrinology and Metabolism Unit, Turkey
| | - Filiz Afrashı
- Ege University School of Medicine, Ophtalmology Department, Turkey
| | - Yasemin Akcay
- Ege University School of Medicine, Medical Biochemistry Department, Turkey
| | - Ilgin Yildirim Simsir
- Ege University School of Medicine, Internal Medicine Department, Endocrinology and Metabolism Unit, Turkey
| | - Banu Sarer Yurekli
- Ege University School of Medicine, Internal Medicine Department, Endocrinology and Metabolism Unit, Turkey
| |
Collapse
|
4
|
Kuprytė M, Lesauskaitė V, Siratavičiūtė V, Utkienė L, Jusienė L, Pangonytė D. Expression of Osteopontin and Gremlin 1 Proteins in Cardiomyocytes in Ischemic Heart Failure. Int J Mol Sci 2024; 25:8240. [PMID: 39125809 PMCID: PMC11311846 DOI: 10.3390/ijms25158240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
A relevant role of osteopontin (OPN) and gremlin 1 (Grem1) in regulating cardiac tissue remodeling and formation of heart failure (HF) are documented, with the changes of OPN and Grem1 levels in blood plasma due to acute ischemia, ischemic heart disease-induced advanced HF or dilatative cardiomyopathy being the primary focus in most of these studies. However, knowledge on the early OPN and Grem1 proteins expression changes within cardiomyocytes during remodeling due to chronic ischemia remains insufficient. The aim of this study was to determine the OPN and Grem1 proteins expression changes in human cardiomyocytes at different stages of ischemic HF. A semi-quantitative immunohistochemical analysis was performed in 105 myocardial tissue samples obtained from the left cardiac ventricles. Increased OPN immunostaining intensity was already detected in the stage A HF group, compared to the control group (p < 0.001), and continued to increase in the stage B HF (p < 0.001), achieving the peak of immunostaining in the stages C/D HF group (p < 0.001). Similar data of Grem1 immunostaining intensity changes in cardiomyocytes were documented. Significantly positive correlations were detected between OPN, Grem1 expression in cardiomyocytes and their diameter as well as the length, in addition to positive correlation between OPN and Grem1 expression changes within cardiomyocytes. These novel findings suggest that OPN and Grem1 contribute significantly to reorganization of cellular geometry from the earliest stage of cardiomyocyte remodeling, providing new insights into the ischemic HF pathogenesis.
Collapse
Affiliation(s)
- Milda Kuprytė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Vaiva Lesauskaitė
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania;
| | - Vitalija Siratavičiūtė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Lina Utkienė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Lina Jusienė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| | - Dalia Pangonytė
- Laboratory of Cardiac Pathology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania; (M.K.); (V.S.); (L.U.); (L.J.)
| |
Collapse
|
5
|
Lusta KA, Summerhill VI, Khotina VA, Sukhorukov VN, Glanz VY, Orekhov AN. The Role of Bacterial Extracellular Membrane Nanovesicles in Atherosclerosis: Unraveling a Potential Trigger. Curr Atheroscler Rep 2024; 26:289-304. [PMID: 38805145 DOI: 10.1007/s11883-024-01206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE OF REVIEW In this review, we explore the intriguing and evolving connections between bacterial extracellular membrane nanovesicles (BEMNs) and atherosclerosis development, highlighting the evidence on molecular mechanisms by which BEMNs can promote the athero-inflammatory process that is central to the progression of atherosclerosis. RECENT FINDINGS Atherosclerosis is a chronic inflammatory disease primarily driven by metabolic and lifestyle factors; however, some studies have suggested that bacterial infections may contribute to the development of both atherogenesis and inflammation in atherosclerotic lesions. In particular, the participation of BEMNs in atherosclerosis pathogenesis has attracted special attention. We provide some general insights into how the immune system responds to potential threats such as BEMNs during the development of atherosclerosis. A comprehensive understanding of contribution of BEMNs to atherosclerosis pathogenesis may lead to the development of targeted interventions for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Konstantin A Lusta
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Volha I Summerhill
- Department of Research and Development, Institute for Atherosclerosis Research, Moscow, 121609, Russia.
| | - Victoria A Khotina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Vasily N Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Victor Y Glanz
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Alexander N Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia.
- Department of Research and Development, Institute for Atherosclerosis Research, Moscow, 121609, Russia.
| |
Collapse
|
6
|
Wu X, Qin B, Cheng R, Zhou R, Wang X, Zhang Z, Mao X, Xie Z, Chen M, Jiang L, Xie P, Ji J, Zhang W, Yuan S, Hu Z, Liu Q. Angiogenic and Fibrogenic Dual-effect of Gremlin1 on Proliferative Diabetic Retinopathy. Int J Biol Sci 2024; 20:897-915. [PMID: 38250154 PMCID: PMC10797694 DOI: 10.7150/ijbs.85735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/24/2023] [Indexed: 01/23/2024] Open
Abstract
Ocular angiogenic diseases, such as proliferative diabetic retinopathy (PDR), are often characterized by pathological new vessels and fibrosis formation. Anti-vascular endothelial growth factor (VEGF) therapy, despite of its efficiency to inhibit new vessels, has limitations, including drug resistance and retinal fibrosis. Here, we identified that Gremlin1, a novel angiogenesis and fibrosis inducer, was secreted from Müller glial cells, and its expression increased in the vitreous fluid from patients with PDR. Mechanistically, Gremlin1 triggered angiogenesis by promoting endothelial-mesenchymal transition via the EGFR/RhoA/ROCK pathway. In addition, Gremlin1 activated microglia to present profibrotic and fibrogenic properties. Further, anti-Gremlin1 antibody inhibited ocular angiogenesis and microglia fibrosis in mouse models. Collectively, Gremlin1 could be a potential therapeutic target in the treatment of ocular angiogenic diseases.
Collapse
Affiliation(s)
- Xinjing Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Bing Qin
- Department of Ophthalmology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian 223800, China
| | - Ruiwen Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ru Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Ophthalmology, People's Hospital of Yangzhong City, Yangzhong 212200, China
| | - Xingxing Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhengyu Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiying Mao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhan Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mingkang Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lin Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiangdong Ji
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Weiwei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zizhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
7
|
Harm T, Rath D, Kreisselmeier KP, Baas L, Prang C, Gekeler S, Schröder S, Gawaz MP, Geisler T, Müller II, Müller KAL. Thrombus architecture is influenced by the antiplatelet loading treatment in patients with acute myocardial infarction. Thromb Res 2023; 230:45-54. [PMID: 37634310 DOI: 10.1016/j.thromres.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Intracoronary thrombus formation is a main cause of acute myocardial infarction triggered by platelet activation. However, there are no data on the impact of different treatment strategies with antiplatelet agents before percutaneous coronary intervention (PCI) on histological characteristics of thrombus formation. OBJECTIVE In this study, we investigate the impact of preinterventional administration of the P2Y12-inhibitors clopidogrel and prasugrel on thrombus composition, highlighting significant changes associated with the antiplatelet pre-treatment. METHODS We prospectively enrolled 104 consecutive patients with ST-segment elevation myocardial infarction (STEMI) undergoing immediate PCI and thrombus aspiration by immunohistochemical staining along with RNA-sequencing employing Nanostring analysis. Fifty-two patients were treated with either prasugrel loading (60 mg) or clopidogrel loading (600 mg) prior to PCI, respectively. RESULTS In Patients with STEMI, intracoronary thrombus architecture was significantly altered between patients pre-treated with prasugrel when compared to clopidogrel. Fibrin content of thrombi was significantly decreased (41.8 % versus 66.7 %, p = 0.009) after pre-treatment with prasugrel compared to clopidogrel. Furthermore, levels of MPO positive cells in intracoronary thrombi were significantly decreased in patients with prasugrel pre-treatment (90.5 versus 201.1, p = 0.014) indicating an association of antiplatelet pre-treatment and the inflammatory responses during thrombus formation. Most strikingly, we observed significant differences among both pre-treatment groups regarding altered RNA expression and signaling pathways of thrombo-inflammatory processes within the thrombotic material, which were independently associated with antiplatelet strategies. CONCLUSIONS Our study elucidates the impact of antiplatelet pre-treatment on thrombus remodeling and architecture, thereby lowering the risk of recurrent adverse cardiovascular events in prasugrel-treated patients.
Collapse
Affiliation(s)
- Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Klaus-Peter Kreisselmeier
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Livia Baas
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Carolin Prang
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Sarah Gekeler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Stephen Schröder
- Department of Cardiology and Angiology, Klinik am Eichert, Eichertstraße 3, 73035 Göppingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany.
| | - Iris Irmgard Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| |
Collapse
|
8
|
Mthunzi L, Rowan SC, Kostyunina DS, Baugh JA, Knaus UG, McLoughlin P. Gremlin 1 is required for macrophage M2 polarization. Am J Physiol Lung Cell Mol Physiol 2023; 325:L270-L276. [PMID: 37401390 DOI: 10.1152/ajplung.00163.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023] Open
Abstract
Pro-proliferative, M2-like polarization of macrophages is a critical step in the development of fibrosis and remodeling in chronic lung diseases such as pulmonary fibrosis and pulmonary hypertension. Macrophages in healthy and diseased lungs express gremlin 1 (Grem1), a secreted glycoprotein that acts in both paracrine and autocrine manners to modulate cellular function. Increased Grem1 expression plays a central role in pulmonary fibrosis and remodeling, however, the role of Grem1 in M2-like polarization of macrophages has not previously been explored. The results reported here show that recombinant Grem1 potentiated M2-like polarization of mouse macrophages and bone marrow-derived macrophages (BMDMs) in response to the Th2 cytokines IL4 and IL13. Genetic depletion of Grem1 in BMDMs inhibited M2 polarization while exogenous gremlin 1 could partially rescue this effect. Taken together, these findings reveal that gremlin 1 is required for M2-like polarization of macrophages.NEW & NOTEWORTHY We show here that gremlin 1 potentiated M2 polarization of mouse bone marrow-derived macrophages (BMDMs) in response to the Th2 cytokines IL4 and IL13. Genetic depletion of Grem1 in BMDMs inhibited M2 polarization while exogenous gremlin 1 partially rescued this effect. Taken together, these findings reveal a previously unknown requirement for gremlin 1 in M2 polarization of macrophages and suggest a novel cellular mechanism promoting fibrosis and remodeling in lung diseases.
Collapse
Affiliation(s)
- Liberty Mthunzi
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - Simon C Rowan
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - Daria S Kostyunina
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - John A Baugh
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - Ulla G Knaus
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- School of Medicine, Health Sciences Centre, University College Dublin, Dublin, Ireland
| |
Collapse
|
9
|
Sun R, Han R, McCornack C, Khan S, Tabor GT, Chen Y, Hou J, Jiang H, Schoch KM, Mao DD, Cleary R, Yang A, Liu Q, Luo J, Petti A, Miller TM, Ulrich JD, Holtzman DM, Kim AH. TREM2 inhibition triggers antitumor cell activity of myeloid cells in glioblastoma. SCIENCE ADVANCES 2023; 9:eade3559. [PMID: 37172094 PMCID: PMC10181199 DOI: 10.1126/sciadv.ade3559] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 04/07/2023] [Indexed: 05/14/2023]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) plays important roles in brain microglial function in neurodegenerative diseases, but the role of TREM2 in the GBM TME has not been examined. Here, we found that TREM2 is highly expressed in myeloid subsets, including macrophages and microglia in human and mouse GBM tumors and that high TREM2 expression correlates with poor prognosis in patients with GBM. TREM2 loss of function in human macrophages and mouse myeloid cells increased interferon-γ-induced immunoactivation, proinflammatory polarization, and tumoricidal capacity. In orthotopic mouse GBM models, mice with chronic and acute Trem2 loss of function exhibited decreased tumor growth and increased survival. Trem2 inhibition reprogrammed myeloid phenotypes and increased programmed cell death protein 1 (PD-1)+CD8+ T cells in the TME. Last, Trem2 deficiency enhanced the effectiveness of anti-PD-1 treatment, which may represent a therapeutic strategy for patients with GBM.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Rowland Han
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Colin McCornack
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Saad Khan
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - G. Travis Tabor
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yun Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jinchao Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Haowu Jiang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kathleen M. Schoch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA
| | - Diane D. Mao
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryan Cleary
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Alicia Yang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Qin Liu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Allegra Petti
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy M. Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA
| | - Jason D. Ulrich
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA
- Center for Science and Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA
| | - Albert H. Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Grillo E, Ravelli C, Colleluori G, D'Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev 2023; 69:51-60. [PMID: 36155165 DOI: 10.1016/j.cytogfr.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Francesco D'Agostino
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Domenichini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
11
|
Kim JE, Li B, Fei L, Horne R, Lee D, Loe AK, Miyake H, Ayar E, Kim DK, Surette MG, Philpott DJ, Sherman P, Guo G, Pierro A, Kim TH. Gut microbiota promotes stem cell differentiation through macrophage and mesenchymal niches in early postnatal development. Immunity 2022; 55:2300-2317.e6. [PMID: 36473468 DOI: 10.1016/j.immuni.2022.11.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/15/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Intestinal stem cell maturation and development coincide with gut microbiota exposure after birth. Here, we investigated how early life microbial exposure, and disruption of this process, impacts the intestinal stem cell niche and development. Single-cell transcriptional analysis revealed impaired stem cell differentiation into Paneth cells and macrophage specification upon antibiotic treatment in early life. Mouse genetic and organoid co-culture experiments demonstrated that a CD206+ subset of intestinal macrophages secreted Wnt ligands, which maintained the mesenchymal niche cells important for Paneth cell differentiation. Antibiotics and reduced numbers of Paneth cells are associated with the deadly infant disease, necrotizing enterocolitis (NEC). We showed that colonization with Lactobacillus or transfer of CD206+ macrophages promoted Paneth cell differentiation and reduced NEC severity. Together, our work defines the gut microbiota-mediated regulation of stem cell niches during early postnatal development.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Bo Li
- General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Lijiang Fei
- Center for Stem Cell and Regenerative Medicine, Zhejiang University of School of Medicine, Hangzhou 310058, China
| | - Rachael Horne
- Program in Cell Biology, Division of Gastroenterology, Hepatology & Nutrition, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dorothy Lee
- General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Adrian Kwan Loe
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hiromu Miyake
- General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Eda Ayar
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dae-Kyum Kim
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, Department of Medicine, McMaster University, 1280 Main St. W, Hamilton, ON L8S 4L8, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Philip Sherman
- Program in Cell Biology, Division of Gastroenterology, Hepatology & Nutrition, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University of School of Medicine, Hangzhou 310058, China
| | - Agostino Pierro
- General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
12
|
A set of common buccal CpGs that predict epigenetic age and associate with lifespan-regulating genes. iScience 2022; 25:105304. [PMID: 36304118 PMCID: PMC9593711 DOI: 10.1016/j.isci.2022.105304] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/11/2022] [Accepted: 10/02/2022] [Indexed: 11/23/2022] Open
Abstract
Epigenetic aging clocks are computational models that use DNA methylation sites to predict age. Since cheek swabs are non-invasive and painless, collecting DNA from buccal tissue is highly desirable. Here, we review 11 existing clocks that have been applied to buccal tissue. Two of these were exclusively trained on adults and, while moderately accurate, have not been used to capture health-relevant differences in epigenetic age. Using 130 common CpGs utilized by two or more existing buccal clocks, we generate a proof-of-concept predictor in an adult methylomic dataset. In addition to accurately estimating age (r = 0.95 and mean absolute error = 3.88 years), this clock predicted that Down syndrome subjects were significantly older relative to controls. A literature and database review of CpG-associated genes identified numerous genes (e.g., CLOCK, ELOVL2, and VGF) and molecules (e.g., alpha-linolenic acid, glycine, and spermidine) reported to influence lifespan and/or age-related disease in model organisms. 130 CpGs have been used by two or more aging clocks applied to human buccal tissue Common CpG genes are linked to the adaptive immune system and telomere maintenance Common CpGs can be used to build a novel, proof-of-concept epigenetic aging clock Several compounds associated with common CpG genes regulate lifespan in animals
Collapse
|
13
|
Brandhofer M, Hoffmann A, Blanchet X, Siminkovitch E, Rohlfing AK, El Bounkari O, Nestele JA, Bild A, Kontos C, Hille K, Rohde V, Fröhlich A, Golemi J, Gokce O, Krammer C, Scheiermann P, Tsilimparis N, Sachs N, Kempf WE, Maegdefessel L, Otabil MK, Megens RTA, Ippel H, Koenen RR, Luo J, Engelmann B, Mayo KH, Gawaz M, Kapurniotu A, Weber C, von Hundelshausen P, Bernhagen J. Heterocomplexes between the atypical chemokine MIF and the CXC-motif chemokine CXCL4L1 regulate inflammation and thrombus formation. Cell Mol Life Sci 2022; 79:512. [PMID: 36094626 PMCID: PMC9468113 DOI: 10.1007/s00018-022-04539-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/31/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022]
Abstract
To fulfil its orchestration of immune cell trafficking, a network of chemokines and receptors developed that capitalizes on specificity, redundancy, and functional selectivity. The discovery of heteromeric interactions in the chemokine interactome has expanded the complexity within this network. Moreover, some inflammatory mediators, not structurally linked to classical chemokines, bind to chemokine receptors and behave as atypical chemokines (ACKs). We identified macrophage migration inhibitory factor (MIF) as an ACK that binds to chemokine receptors CXCR2 and CXCR4 to promote atherogenic leukocyte recruitment. Here, we hypothesized that chemokine–chemokine interactions extend to ACKs and that MIF forms heterocomplexes with classical chemokines. We tested this hypothesis by using an unbiased chemokine protein array. Platelet chemokine CXCL4L1 (but not its variant CXCL4 or the CXCR2/CXCR4 ligands CXCL8 or CXCL12) was identified as a candidate interactor. MIF/CXCL4L1 complexation was verified by co-immunoprecipitation, surface plasmon-resonance analysis, and microscale thermophoresis, also establishing high-affinity binding. We next determined whether heterocomplex formation modulates inflammatory/atherogenic activities of MIF. Complex formation was observed to inhibit MIF-elicited T-cell chemotaxis as assessed by transwell migration assay and in a 3D-matrix-based live cell-imaging set-up. Heterocomplexation also blocked MIF-triggered migration of microglia in cortical cultures in situ, as well as MIF-mediated monocyte adhesion on aortic endothelial cell monolayers under flow stress conditions. Of note, CXCL4L1 blocked binding of Alexa-MIF to a soluble surrogate of CXCR4 and co-incubation with CXCL4L1 attenuated MIF responses in HEK293-CXCR4 transfectants, indicating that complex formation interferes with MIF/CXCR4 pathways. Because MIF and CXCL4L1 are platelet-derived products, we finally tested their role in platelet activation. Multi-photon microscopy, FLIM-FRET, and proximity-ligation assay visualized heterocomplexes in platelet aggregates and in clinical human thrombus sections obtained from peripheral artery disease (PAD) in patients undergoing thrombectomy. Moreover, heterocomplexes inhibited MIF-stimulated thrombus formation under flow and skewed the lamellipodia phenotype of adhering platelets. Our study establishes a novel molecular interaction that adds to the complexity of the chemokine interactome and chemokine/receptor-network. MIF/CXCL4L1, or more generally, ACK/CXC-motif chemokine heterocomplexes may be target structures that can be exploited to modulate inflammation and thrombosis.
Collapse
Affiliation(s)
- Markus Brandhofer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Adrian Hoffmann
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.,Department of Anesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Xavier Blanchet
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany
| | - Elena Siminkovitch
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Omar El Bounkari
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Jeremy A Nestele
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Alexander Bild
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Christos Kontos
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Kathleen Hille
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Vanessa Rohde
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Adrian Fröhlich
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Jona Golemi
- Systems Neuroscience Group, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Ozgun Gokce
- Systems Neuroscience Group, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany
| | - Christine Krammer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Patrick Scheiermann
- Department of Anesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Nikolaos Tsilimparis
- Department of Vascular Surgery, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Nadja Sachs
- Department for Vascular and Endovascular Surgery, Klinikum Rechts Der Isar, Technische Universität München (TUM), 81675, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany
| | - Wolfgang E Kempf
- Department for Vascular and Endovascular Surgery, Klinikum Rechts Der Isar, Technische Universität München (TUM), 81675, Munich, Germany
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum Rechts Der Isar, Technische Universität München (TUM), 81675, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany
| | - Michael K Otabil
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Hans Ippel
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Rory R Koenen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Junfu Luo
- Vascular Biology and Pathology, Institute of Laboratory Medicine, Ludwig-Maximilians-Universität, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Bernd Engelmann
- Vascular Biology and Pathology, Institute of Laboratory Medicine, Ludwig-Maximilians-Universität, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, 81377, Munich, Germany
| | - Kevin H Mayo
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands.,Department of Biochemistry, Molecular Biology and Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Aphrodite Kapurniotu
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), 85354, Freising, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany.,Munich Heart Alliance, 80802, Munich, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER, Maastricht, The Netherlands
| | - Philipp von Hundelshausen
- Institute for Cardiovascular Prevention (IPEK), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Pettenkofer Straße 8a/9, 80336, Munich, Germany. .,Munich Heart Alliance, 80802, Munich, Germany.
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), 81377, Munich, Germany. .,Munich Heart Alliance, 80802, Munich, Germany.
| |
Collapse
|
14
|
Song S, Xiao Z, Dekker FJ, Poelarends GJ, Melgert BN. Macrophage migration inhibitory factor family proteins are multitasking cytokines in tissue injury. Cell Mol Life Sci 2022; 79:105. [PMID: 35091838 PMCID: PMC8799543 DOI: 10.1007/s00018-021-04038-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
The family of macrophage migration inhibitory factor (MIF) proteins in humans consist of MIF, its functional homolog D-dopachrome tautomerase (D-DT, also known as MIF-2) and the relatively unknown protein named DDT-like (DDTL). MIF is a pleiotropic cytokine with multiple properties in tissue homeostasis and pathology. MIF was initially found to associate with inflammatory responses and therefore established a reputation as a pro-inflammatory cytokine. However, increasing evidence demonstrates that MIF influences many different intra- and extracellular molecular processes important for the maintenance of cellular homeostasis, such as promotion of cellular survival, antioxidant signaling, and wound repair. In contrast, studies on D-DT are scarce and on DDTL almost nonexistent and their functions remain to be further investigated as it is yet unclear how similar they are compared to MIF. Importantly, the many and sometimes opposing functions of MIF suggest that targeting MIF therapeutically should be considered carefully, taking into account timing and severity of tissue injury. In this review, we focus on the latest discoveries regarding the role of MIF family members in tissue injury, inflammation and repair, and highlight the possibilities of interventions with therapeutics targeting or mimicking MIF family proteins.
Collapse
|
15
|
Elemam NM, Malek AI, Mahmoud EE, El-Huneidi W, Talaat IM. Insights into the Role of Gremlin-1, a Bone Morphogenic Protein Antagonist, in Cancer Initiation and Progression. Biomedicines 2022; 10:301. [PMID: 35203511 PMCID: PMC8869528 DOI: 10.3390/biomedicines10020301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
The bone morphogenic protein (BMP) antagonist Gremlin-1 is a biologically significant regulator known for its crucial role in tissue differentiation and embryonic development. Nevertheless, it has been reported that Gremlin-1 can exhibit its function through BMP dependent and independent pathways. Gremlin-1 has also been reported to be involved in organ fibrosis, which has been correlated to the development of other diseases, such as renal inflammation and diabetic nephropathy. Based on growing evidence, Gremlin-1 has recently been implicated in the initiation and progression of different types of cancers. Further, it contributes to the stemness state of cancer cells. Herein, we explore the recent findings on the role of Gremlin-1 in various cancer types, including breast, cervical, colorectal, and gastric cancers, as well as glioblastomas. Additionally, we highlighted the impact of Gremlin-1 on cellular processes and signaling pathways involved in carcinogenesis. Therefore, it was suggested that Gremlin-1 might be a promising prognostic biomarker and therapeutic target in cancers.
Collapse
Affiliation(s)
- Noha Mousaad Elemam
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdullah Imadeddin Malek
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
| | - Esraa Elaraby Mahmoud
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
| | - Waseem El-Huneidi
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Iman M. Talaat
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| |
Collapse
|
16
|
Sauter M, Sauter RJ, Nording H, Lin C, Olbrich M, Autenrieth S, Gleissner C, Thunemann M, Otero N, Lutgens E, Aherrahrou Z, Wolf D, Zender L, Meuth S, Feil R, Langer HF. Apolipoprotein E derived from CD11c + cells ameliorates atherosclerosis. iScience 2022; 25:103677. [PMID: 35036868 PMCID: PMC8749187 DOI: 10.1016/j.isci.2021.103677] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/01/2021] [Accepted: 12/20/2021] [Indexed: 11/24/2022] Open
Abstract
Atherosclerosis is studied in models with dysfunctional lipid homeostasis—predominantly the ApoE−/− mouse. The role of antigen-presenting cells (APCs) for lipid homeostasis is not clear. Using a LacZ reporter mouse, we showed that CD11c+ cells were enriched in aortae of ApoE−/− mice. Systemic long-term depletion of CD11c+ cells in ApoE−/− mice resulted in significantly increased plaque formation associated with reduced serum ApoE levels. In CD11ccre+ApoEfl/fl and Albumincre+ApoEfl/fl mice, we could show that ≈70% of ApoE is liver-derived and ≈25% originates from CD11c+ cells associated with significantly increased atherosclerotic plaque burden in both strains. Exposure to acLDL promoted cholesterol efflux from CD11c+ cells and cell-specific deletion of ApoE resulted in increased inflammation reflected by increased IL-1β serum levels. Our results determined for the first time the level of ApoE originating from CD11c+ cells and demonstrated that CD11c+ cells ameliorate atherosclerosis by the secretion of ApoE. CD11c+ cells are enriched in aortae of high cholesterol-fed ApoE−/- mice Depletion of CD11c+ cells increases plaque size in ApoE−/- mice ≈ 20% of serum ApoE derives from CD11c+ cells ApoE from CD11c+ cells contributes to protection from atherosclerosis
Collapse
Affiliation(s)
- Manuela Sauter
- Department of Cardiology, University Hospital, Medical Clinic II, University Heart Center Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | - Reinhard J Sauter
- Department of Cardiology, University Hospital, Medical Clinic II, University Heart Center Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | - Henry Nording
- Department of Cardiology, University Hospital, Medical Clinic II, University Heart Center Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, 23562 Luebeck, Germany
| | - Chaolan Lin
- Department of Cardiology, University Hospital, Medical Clinic II, University Heart Center Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| | - Marcus Olbrich
- University Hospital, Department of Cardiology, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Stella Autenrieth
- University Hospital, Department of Hematology and Oncology, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Christian Gleissner
- University Hospital, Department of Cardiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Martin Thunemann
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Nadia Otero
- Philipps University Marburg, Faculty of Medicine, 35043 Marburg, Germany
| | - Esther Lutgens
- University Hospital Munich, Institute for Prophylaxis and Epidemiology of Circulatory Diseases, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Zouhair Aherrahrou
- University of Luebeck, Institute of Cardiogenetics, 23538 Luebeck, Germany
| | - Dennis Wolf
- University Hospital, Department of Cardiology and Angiology, University Heart Center Freiburg - Bad Krozingen, 79106 Freiburg, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, 72076 Tuebingen, Germany.,DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), University of Tuebingen, 72076 Tuebingen, Germany.,German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sven Meuth
- University Hospital, Department of Neurology, University of Duesseldorf, 40225 Duesseldorf, Germany
| | - Robert Feil
- Interfaculty Institute of Biochemistry, University of Tuebingen, 72076 Tuebingen, Germany
| | - Harald F Langer
- Department of Cardiology, University Hospital, Medical Clinic II, University Heart Center Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany
| |
Collapse
|
17
|
Davis JM, Cheng B, Drake MM, Yu Q, Yang B, Li J, Liu C, Younes M, Zhao X, Bailey JM, Shen Q, Ko TC, Cao Y. Pancreatic stromal Gremlin 1 expression during pancreatic tumorigenesis. Genes Dis 2022; 9:108-115. [PMID: 35005111 PMCID: PMC8720668 DOI: 10.1016/j.gendis.2020.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/07/2020] [Accepted: 05/08/2020] [Indexed: 12/28/2022] Open
Abstract
Chronic pancreatitis (CP) is a major risk factor of pancreatic ductal adenocarcinoma (PDAC). How CP promotes pancreatic oncogenesis remains unclear. A characteristic feature of PDAC is its prominent desmoplasia in the tumor microenvironment, composed of activated fibroblasts and macrophages. Macrophages can be characterized as M1 or M2, with tumor-inhibiting or -promoting functions, respectively. We reported that Gremlin 1 (GREM1), a key pro-fibrogenic factor, is upregulated in the stroma of CP. The current study aimed to investigate the expression of GREM1 and correlation between GREM1 and macrophages within the pancreas during chronic inflammation and the development of PDAC. By mRNA in situ hybridization, we detected GREM1 mRNA expression within α-smooth muscle actin (SMA)-positive fibroblasts of the pancreatic stroma. These designated FibroblastsGrem1+ marginally increased from CP to pancreatic intraepithelial neoplasia (PanIN) and PDAC. Within PDAC, FibroblastsGrem1+ increased with higher pathological tumor stages and in a majority of PDAC subtypes screened. Additionally, FibroblastsGrem1+ positively correlated with total macrophages (MacCD68+) and M2 macrophages (M2CD163+) in PDAC. To begin exploring potential molecular links between FibroblastsGrem1+ and macrophages in PDAC, we examined the expression of macrophage migration inhibitory factor (MIF), an endogenous counteracting molecule of GREM1 and an M1 macrophage promoting factor. By IHC staining of MIF, we found MIF to be expressed by tumor cells, positively correlated with GREM1; by IHC co-staining, we found MIF to be negatively correlated with M2CD163+ expression. Our findings suggest that GREM1 expression by activated fibroblasts may promote PDAC development, and GREM1/MIF may play an important role in macrophage phenotype.
Collapse
Affiliation(s)
- Joy M. Davis
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Binglu Cheng
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Madeline M. Drake
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Qiang Yu
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Baibing Yang
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jing Li
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Chunhui Liu
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mamoun Younes
- Department of Pathology & Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiurong Zhao
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jennifer M. Bailey
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Qiang Shen
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Tien C. Ko
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yanna Cao
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
18
|
O'Reilly S. Gremlin: a complex molecule regulating wound healing and fibrosis. Cell Mol Life Sci 2021; 78:7917-7923. [PMID: 34731251 PMCID: PMC11071963 DOI: 10.1007/s00018-021-03964-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/27/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Gremlin-1 is part of the TGF-β superfamily and is a BMP antagonist that blocks BMP signalling to precisely control BMP gradients. Gremlin-1 is primarily involved in organogenesis and limb patterning however, has recently been described as being involved in fibrotic diseases. Initially described as a key factor involved in diabetic kidney fibrosis due to being induced by high glucose, it has now been described as being associated with lung, liver, eye, and skin fibrosis. This suggests that it is a key conserved molecule mediating fibrotic events irrespective of organ. It appears that Gremlin-1 may have effects mediated by BMP-dependent and independent pathways. The aim of this review is to evaluate the role of Gremlin-1 in fibrosis, its mechanisms and if this can be targeted therapeutically in fibrotic diseases, which currently have very limited treatment options and are highly prevalent.
Collapse
|
19
|
Platelet-Derived PCSK9 Is Associated with LDL Metabolism and Modulates Atherothrombotic Mechanisms in Coronary Artery Disease. Int J Mol Sci 2021; 22:ijms222011179. [PMID: 34681838 PMCID: PMC8538687 DOI: 10.3390/ijms222011179] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/03/2023] Open
Abstract
Platelets play a significant role in atherothrombosis. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is critically involved in the regulation of LDL metabolism and interacts with platelet function. The effect of PCSK9 in platelet function is poorly understood. The authors of this article sought to characterize platelets as a major source of PCSK9 and PCSK9's role in atherothrombosis. In a large cohort of patients with coronary artery disease (CAD), platelet count, platelet reactivity, and platelet-derived PCSK9 release were analyzed. The role of platelet PCSK9 on platelet and monocyte function was investigated in vitro. Platelet count and hyper-reactivity correlated with plasma LDL in CAD. The circulating platelets express on their surface and release substantial amounts of PCSK9. Release of PCSK9 augmented platelet-dependent thrombosis, monocyte migration, and differentiation into macrophages/foam cells. Platelets and PCSK9 accumulated in tissue derived from atherosclerotic carotid arteries in areas of macrophages. PCSK9 inhibition reduced platelet activation and platelet-dependent thrombo-inflammation. The authors identified platelets as a source of PCSK9 in CAD, which may have an impact on LDL metabolism. Furthermore, platelet-derived PCSK9 contributes to atherothrombosis, and inhibition of PCSK9 attenuates thrombo-inflammation, which may contribute to the reported beneficial clinical effects.
Collapse
|
20
|
Müller II, Schneider M, Müller KAL, Lunov O, Borst O, Simmet T, Gawaz M. Protective role of Gremlin-1 in myocardial function. Eur J Clin Invest 2021; 51:e13539. [PMID: 33729579 DOI: 10.1111/eci.13539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/26/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Gremlin-1 is a cystine knot protein and is expressed in organs developing fibrosis. Transient ischaemia leads to myocardial fibrosis, a major determinant of impaired myocardial function. MATERIALS AND METHODS Expression of Gremlin-1 was investigated in infarcted myocardium by real-time PCR, Western blot analysis, histological and immunohistochemistry staining. We further elaborated the colocalization of Gremlin-1 and TGF-β proteins by confocal microscopy and co-immunoprecipitation experiments. The interaction between Gremlin-1 and TGF-β was analysed by photon correlation spectroscopy. Gremlin-1 modulation of the TGF-β-dependent collagen I synthesis in fibroblasts was investigated using ELISA and immunohistochemistry experiments. The effect of prolonged administration of recombinant Gremlin-1 on myocardial function following ischaemia/reperfusion was accessed by echocardiography and immunohistochemistry. RESULTS Gremlin-1 is expressed in myocardial tissue and infiltrating cells after transient myocardial ischaemia (P < .05). Gremlin-1 colocalizes with the pro-fibrotic cytokine transforming growth factor-β (TGF-β) expressed in fibroblasts and inflammatory cell infiltrates (P < .05). Gremlin-1 reduces TGF-β-induced collagen production of myocardial fibroblasts by approximately 20% (P < .05). We found that Gremlin-1 binds with high affinity to TGF-β (KD = 54 nmol/L) as evidenced by photon correlation spectroscopy and co-immunoprecipitation. intravenous administration of m Gremlin-1-Fc, but not of equivalent amount of Fc control, significantly reduced infarct size by approximately 20%. In the m Gremlin-1-Fc group, infarct area was reduced by up to 30% in comparison with mice treated with Fc control (I/LV: 4.8 ± 1.2% vs 6.0 ± 1.2% P < .05; I/AaR: 15.2 ± 1.5% vs 21.1 ± 5%, P < .05). CONCLUSIONS The present data disclose Gremlin-1 as an antagonist of TGF-β and presume a role for Gremlin-1/TGF-β interaction in myocardial remodelling following myocardial ischaemia.
Collapse
Affiliation(s)
- Iris I Müller
- Department of Cardiology and Angiology, University Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Martina Schneider
- Department of Cardiology and Angiology, University Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Karin A L Müller
- Department of Cardiology and Angiology, University Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Oleg Lunov
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
21
|
Lázaro SF, Tonhati H, Oliveira HR, Silva AA, Nascimento AV, Santos DJA, Stefani G, Brito LF. Genomic studies of milk-related traits in water buffalo (Bubalus bubalis) based on single-step genomic best linear unbiased prediction and random regression models. J Dairy Sci 2021; 104:5768-5793. [PMID: 33685677 DOI: 10.3168/jds.2020-19534] [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: 08/27/2020] [Accepted: 01/02/2021] [Indexed: 01/14/2023]
Abstract
Genomic selection has been widely implemented in many livestock breeding programs, but it remains incipient in buffalo. Therefore, this study aimed to (1) estimate variance components incorporating genomic information in Murrah buffalo; (2) evaluate the performance of genomic prediction for milk-related traits using single- and multitrait random regression models (RRM) and the single-step genomic best linear unbiased prediction approach; and (3) estimate longitudinal SNP effects and candidate genes potentially associated with time-dependent variation in milk, fat, and protein yields, as well as somatic cell score (SCS) in multiple parities. The data used to estimate the genetic parameters consisted of a total of 323,140 test-day records. The average daily heritability estimates were moderate (0.35 ± 0.02 for milk yield, 0.22 ± 0.03 for fat yield, 0.42 ± 0.03 for protein yield, and 0.16 ± 0.03 for SCS). The highest heritability estimates, considering all traits studied, were observed between 20 and 280 d in milk (DIM). The genetic correlation estimates at different DIM among the evaluated traits ranged from -0.10 (156 to 185 DIM for SCS) to 0.61 (36 to 65 DIM for fat yield). In general, direct selection for any of the traits evaluated is expected to result in indirect genetic gains for milk yield, fat yield, and protein yield but also increase SCS at certain lactation stages, which is undesirable. The predicted RRM coefficients were used to derive the genomic estimated breeding values (GEBV) for each time point (from 5 to 305 DIM). In general, the tuning parameters evaluated when constructing the hybrid genomic relationship matrices had a small effect on the GEBV accuracy and a greater effect on the bias estimates. The SNP solutions were back-solved from the GEBV predicted from the Legendre random regression coefficients, which were then used to estimate the longitudinal SNP effects (from 5 to 305 DIM). The daily SNP effect for 3 different lactation stages were performed considering 3 different lactation stages for each trait and parity: from 5 to 70, from 71 to 150, and from 151 to 305 DIM. Important genomic regions related to the analyzed traits and parities that explain more than 0.50% of the total additive genetic variance were selected for further analyses of candidate genes. In general, similar potential candidate genes were found between traits, but our results suggest evidence of differential sets of candidate genes underlying the phenotypic expression of the traits across parities. These results contribute to a better understanding of the genetic architecture of milk production traits in dairy buffalo and reinforce the relevance of incorporating genomic information to genetically evaluate longitudinal traits in dairy buffalo. Furthermore, the candidate genes identified can be used as target genes in future functional genomics studies.
Collapse
Affiliation(s)
- Sirlene F Lázaro
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907; Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Humberto Tonhati
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Hinayah R Oliveira
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907; Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, N1G 2W1, ON, Canada
| | - Alessandra A Silva
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - André V Nascimento
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Daniel J A Santos
- Department of Animal and Avian Science, University of Maryland, College Park 20742
| | - Gabriela Stefani
- Department of Animal Science, College of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, 14884-900, SP, Brazil
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907.
| |
Collapse
|
22
|
Khatib Shahidi R, M Hoffmann J, Hedjazifar S, Bonnet L, K Baboota R, Heasman S, Church C, Elias I, Bosch F, Boucher J, Hammarstedt A, Smith U. Adult mice are unresponsive to AAV8-Gremlin1 gene therapy targeting the liver. PLoS One 2021; 16:e0247300. [PMID: 33606810 PMCID: PMC7895349 DOI: 10.1371/journal.pone.0247300] [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: 05/29/2020] [Accepted: 02/05/2021] [Indexed: 11/18/2022] Open
Abstract
Objective Gremlin 1 (GREM1) is a secreted BMP2/4 inhibitor which regulates commitment and differentiation of human adipose precursor cells and prevents the browning effect of BMP4. GREM1 is an insulin antagonist and serum levels are high in type 2 diabetes (T2D). We here examined in vivo effects of AAV8 (Adeno-Associated Viral vectors of serotype eight) GREM 1 targeting the liver in mature mice to increase its systemic secretion and also, in a separate study, injected recombinant GREM 1 intraperitoneally. The objective was to characterize systemic effects of GREM 1 on insulin sensitivity, glucose tolerance, body weight, adipose cell browning and other local tissue effects. Methods Adult mice were injected with AAV8 vectors expressing GREM1 in the liver or receiving regular intra-peritoneal injections of recombinant GREM1 protein. The mice were fed with a low fat or high fat diet (HFD) and followed over time. Results Liver-targeted AAV8-GREM1 did not alter body weight, whole-body glucose and insulin tolerance, or adipose tissue gene expression. Although GREM1 protein accumulated in liver cells, GREM1 serum levels were not increased suggesting that it may not have been normally processed for secretion. Hepatic lipid accumulation, inflammation and fibrosis were also not changed. Repeated intraperitoneal rec-GREM1 injections for 5 weeks were also without effects on body weight and insulin sensitivity. UCP1 was slightly but significantly reduced in both white and brown adipose tissue but this was not of sufficient magnitude to alter body weight. We validated that recombinant GREM1 inhibited BMP4-induced pSMAD1/5/9 in murine cells in vitro, but saw no direct inhibitory effect on insulin signalling and pAkt (ser 473 and thr 308) activation. Conclusion GREM1 accumulates intracellularly when overexpressed in the liver cells of mature mice and is apparently not normally processed/secreted. However, also repeated intraperitoneal injections were without effects on body weight and insulin sensitivity and adipose tissue UCP1 levels were only marginally reduced. These results suggest that mature mice do not readily respond to GREMLIN 1 but treatment of murine cells with GREMLIN 1 protein in vitro validated its inhibitory effect on BMP4 signalling while insulin signalling was not altered.
Collapse
Affiliation(s)
- Roxana Khatib Shahidi
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jenny M Hoffmann
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Shahram Hedjazifar
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Laurianne Bonnet
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ritesh K Baboota
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stephanie Heasman
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Christopher Church
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Ivet Elias
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy and Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Jeremie Boucher
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
23
|
Ouahoud S, Hardwick JC, Hawinkels LJ. Extracellular BMP Antagonists, Multifaceted Orchestrators in the Tumor and Its Microenvironment. Int J Mol Sci 2020; 21:ijms21113888. [PMID: 32486027 PMCID: PMC7313454 DOI: 10.3390/ijms21113888] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 02/08/2023] Open
Abstract
The bone morphogenetic proteins (BMPs), a subgroup of the transforming growth factor-β (TGF-β) superfamily, are involved in multiple biological processes such as embryonic development and maintenance of adult tissue homeostasis. The importance of a functional BMP pathway is underlined by various diseases, including cancer, which can arise as a consequence of dysregulated BMP signaling. Mutations in crucial elements of this signaling pathway, such as receptors, have been reported to disrupt BMP signaling. Next to that, aberrant expression of BMP antagonists could also contribute to abrogated signaling. In this review we set out to highlight how BMP antagonists affect not only the cancer cells, but also the other cells present in the microenvironment to influence cancer progression.
Collapse
|
24
|
Tumor-associated macrophage-derived exosomal microRNA-155-5p stimulates intracranial aneurysm formation and macrophage infiltration. Clin Sci (Lond) 2020; 133:2265-2282. [PMID: 31657855 DOI: 10.1042/cs20190680] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 12/22/2022]
Abstract
Tumor-associated macrophages (TAMs) play a regulatory role in inflammation and cancer. Exosomes derived from macrophages carrying microRNAs (miRNAs or miRs) are of great value for cancer therapy. Gremlin 1 (GREM1), a member of the antagonists of secreted bone morphogenetic protein, has been implicated in the pathophysiology of multiple diseases or cancers. Based on the predictions of miRNA-mRNA interaction, GREM1 was found to be a target gene of miR-155-5p. Here, the present study aims to explore the role of TAM-derived exosomal miR-155-5p by regulating GREM1 in intracranial aneurysm (IA). The collected results showed that GREM1 was down-regulated in IA, while miR-155-5p was up-regulated in TAM-derived exosomes. Smooth muscle cells (SMCs) were co-cultured with TAMs or exposed to exosomes derived from TAMs transfected with either miR-155-5p mimic or miR-155-5p inhibitor for exploring their roles in proliferation and migration of SMCs in vitro. Accordingly, in vitro experiments showed that TAM-derived exosomal miR-155-5p could promote proliferation and migration of SMCs by targeting GREM1. The effects of TAM-derived exosomal miR-155-5p on IA formation and TAM activation and infiltration by regulation of GREM1 in vivo were measured in IA rats injected with exosomes or those from TAMs transfected with miR-155-5p inhibitor. In vivo experimental results consistently confirmed that TAM-derived exosomes carrying miR-155-5p promoted IA formation and TAM activation and infiltration. In conclusion, TAM-derived exosomal miR-155-5p promotes IA formation via GREM1, which points to miR-155-5p as a possible therapeutic target for IA.
Collapse
|
25
|
Lavalett L, Ortega H, Barrera LF. Infection of Monocytes From Tuberculosis Patients With Two Virulent Clinical Isolates of Mycobacterium tuberculosis Induces Alterations in Myeloid Effector Functions. Front Cell Infect Microbiol 2020; 10:163. [PMID: 32391286 PMCID: PMC7190864 DOI: 10.3389/fcimb.2020.00163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/26/2020] [Indexed: 12/20/2022] Open
Abstract
Monocytes play a critical role during infection with Mycobacterium tuberculosis (Mtb). They are recruited to the lung, where they participate in the control of infection during active tuberculosis (TB). Alternatively, inflammatory monocytes may participate in inflammation or serve as niches for Mtb infection. Monocytes response to infection may vary depending on the particularities of the clinical isolate of Mtb from which they are infected. In this pilot study, we have examined the baseline mRNA profiles of circulating human monocytes from patients with active TB (MoTB) compared with monocytes from healthy individuals (MoCT). Circulating MoTB displayed a pro-inflammatory transcriptome characterized by increased gene expression of genes associated with cytokines, monocytopoiesis, and down-regulation of MHC class II gene expression. In response to in vitro infection with two clinical isolates of the LAM family of Mtb (UT127 and UT205), MoTB displayed an attenuated inflammatory mRNA profile associated with down-regulation the TREM1 signaling pathway. Furthermore, the gene expression signature induced by Mtb UT205 clinical strain was characterized by the enrichment of genes in pathways and biological processes mainly associated with a signature of IFN-inducible genes and the inhibition of cell death mechanisms compared to MoTB-127, which could favor the establishment and survival of Mtb within the monocytes. These results suggest that circulating MoTB have an altered transcriptome that upon infection with Mtb may help to maintain chronic inflammation and infection. Moreover, this functional abnormality of monocytes may also depend on potential differences in virulence of circulating clinical strains of Mtb.
Collapse
Affiliation(s)
- Lelia Lavalett
- Grupo de Inmunología Celular e Inmunogenética (GICIG), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.,Facultad de Ciencias, Universidad Nacional de Colombia Sede Medellín, Medellín, Colombia
| | - Hector Ortega
- Clínica Cardiovascular Santa María, Medellín, Colombia
| | - Luis F Barrera
- Grupo de Inmunología Celular e Inmunogenética (GICIG), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
26
|
Abstract
Chronic kidney disease (CKD) is a devastating condition that is reaching epidemic levels owing to the increasing prevalence of diabetes mellitus, hypertension and obesity, as well as ageing of the population. Regardless of the underlying aetiology, CKD is slowly progressive and leads to irreversible nephron loss, end-stage renal disease and/or premature death. Factors that contribute to CKD progression include parenchymal cell loss, chronic inflammation, fibrosis and reduced regenerative capacity of the kidney. Current therapies have limited effectiveness and only delay disease progression, underscoring the need to develop novel therapeutic approaches to either stop or reverse progression. Preclinical studies have identified several approaches that reduce fibrosis in experimental models, including targeting cytokines, transcription factors, developmental and signalling pathways and epigenetic modulators, particularly microRNAs. Some of these nephroprotective strategies are now being tested in clinical trials. Lessons learned from the failure of clinical studies of transforming growth factor β1 (TGFβ1) blockade underscore the need for alternative approaches to CKD therapy, as strategies that target a single pathogenic process may result in unexpected negative effects on simultaneously occurring processes. Additional promising avenues include preventing tubular cell injury and anti-fibrotic therapies that target activated myofibroblasts, the main collagen-producing cells.
Collapse
|
27
|
Qin D, Jin X, Jiang Y. Gremlin mediates the TGF-β-induced induction of profibrogenic genes in human retinal pigment epithelial cells. Exp Ther Med 2020; 19:2353-2359. [PMID: 32104303 PMCID: PMC7027231 DOI: 10.3892/etm.2020.8463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/15/2019] [Indexed: 12/26/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is characterised by the contraction and growth of fibrotic membranes on the retina and within the vitreous body. Retinal pigment epithelial (RPE) cells, a major cellular component of the fibrotic membrane, is one of the cell types that have been previously reported to associate with PVR pathogenesis. During PVR, RPE cells undergo increased cell proliferation, migration and the secretion of extracellular matrix molecules, such as fibronectin and type I collagen. A variety of cytokines and growth factors are involved in the formation of the fibrotic membrane. Although gremlin has been reported to serve an important role in the regulation of epithelial-to-mesenchymal transition in PVR, the relationship between gremlin and the expression of profibrogenic factors in human RPE cells remains unclear. In the present study, gremlin promoted RPE cell proliferation and the expression of type I collagen and fibronectin. In addition, knocking down gremlin expression by siRNA significantly suppressed the transforming growth factor (TGF)-β1- and TGF-β2-induced expression of type I collagen and fibronectin in RPE cells. These findings suggest that gremlin may serve an important role in the development of PVR.
Collapse
Affiliation(s)
- Dong Qin
- Henan Eye Institute, Henan Provincial Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Xuemin Jin
- Henan Eye Institute, Henan Provincial Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Yanrong Jiang
- Department of Ophthalmology, People's Hospital, Peking University, Beijing 100044, P.R. China
| |
Collapse
|
28
|
Qin D, Jiang YR, Meng Z. Gremlin in the Vitreous of Patients with Proliferative Diabetic Retinopathy and the Downregulation of Gremlin in Retinal Pigment Epithelial Cells. J Diabetes Res 2020; 2020:9238742. [PMID: 32377526 PMCID: PMC7180400 DOI: 10.1155/2020/9238742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 11/18/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the most common causes of blindness globally. Proliferative DR (PDR), an advanced stage of DR, is characterized by the formation of fibrotic membranes at the vitreoretinal interface. The proliferation, migration, and secretion of extracellular matrix molecules in retinal pigment epithelial (RPE) cells contribute to the formation of fibrotic membranes in PDR. Gremlin has been reported to be upregulated in response to elevated glucose levels in the retina of diabetic rat and bovine pericytes. However, the role of gremlin in PDR remains unclear. In the present study, the vitreous concentrations of gremlin were significantly higher in the PDR (67.79 ± 33.96) group than in the control (45.31 ± 12.31) group, and high glucose levels induced the expression of gremlin in RPE cells. The elevated expression of extracellular matrix molecules, such as fibronectin and collagen IV, was significantly reduced by gremlin siRNA in human RPE cells under high-glucose conditions. Thus, gremlin may play a vital role in the development of PDR.
Collapse
Affiliation(s)
- Dong Qin
- Henan Eye Institute, Henan Provincial Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan-rong Jiang
- Department of Ophthalmology, People's Hospital, Peking University, Beijing, China
| | - Zijun Meng
- Henan Eye Institute, Henan Provincial Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Patsouras A, Farmaki P, Garmpi A, Damaskos C, Garmpis N, Mantas D, Diamantis E. Screening and Risk Assessment of Coronary Artery Disease in Patients With Type 2 Diabetes: An Updated Review. In Vivo 2019; 33:1039-1049. [PMID: 31280191 DOI: 10.21873/invivo.11572] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/25/2022]
Abstract
Current literature indicates that there is a strong correlation between coronary artery disease (CAD) and type 2 diabetes. The arteriosclerotic progression occurs earlier and in a greater extent in the diabetic than in the non-diabetic population. In diabetic subjects, the detection of arterial disease does not always precede the development of an acute arterial incident. Herein, we reviewed studies published within the last 5 years in order to reveal the risk factors for coronary artery disease in patients with type 2 diabetes. In addition, we aimed to discuss how to diagnose in an early stage or even screen the presence of coronary artery disease in asymptomatic diabetic patients. Possible blood markers as predictors of CAD, which are mostly related to the lipidemic profile of subjects, are included in this review. Less invasive imaging methods than conventional coronary angiography, included in the article, are gradually used more in the diagnosis of CAD and show high effectiveness. Data from 23 articles with 22,350 patients having type 2 diabetes were summarized and presented descriptively.
Collapse
Affiliation(s)
| | - Paraskevi Farmaki
- First Department of Pediatrics, Agia Sofia Children's Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Garmpi
- Internal Medicine Department, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Damaskos
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Garmpis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Mantas
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | |
Collapse
|
30
|
Wu Y, Xu W, Hou J, Liu Y, Li R, Liu J, Li C, Tang X, Lin L, Pan Y, Zhang D. Porphyromonas gingivalis-Induced MIF Regulates Intercellular Adhesion Molecule-1 Expression in EA.hy926 Cells and Monocyte-Endothelial Cell Adhesion Through the Receptors CD74 and CXCR4. Inflammation 2019; 42:874-883. [PMID: 30506423 PMCID: PMC6527533 DOI: 10.1007/s10753-018-0942-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Porphyromonas gingivalis (P. gingivalis) is an important pathogen that contributes to periodontal disease and causes infections that promote the progression of atherosclerosis. Our previous studies showed that macrophage migration inhibitory factor (MIF) facilitates monocyte adhesion to endothelial cells by regulating the expression of intercellular adhesion molecule-1 (ICAM-1) in P. gingivalis-infected endothelial cells. However, the detailed pathological role of MIF has yet to be elucidated in this context. To explore the functional receptor(s) of MIF that underlie its participation in the pathogenesis of atherosclerosis, we investigated the expression of the chemokine receptors CD74 and CXCR4 in endothelial cells, both of which were shown to be involved in the adhesion of monocytes to endothelial cells pretreated with P. gingivalis. Furthermore, the formation of a MIF, CD74, and CXCR4 ligand-receptor complex was revealed by our immunofluorescence staining and coimmunoprecipitation results. By interacting with the CD74/CXCR4 receptor complex, MIF may act as a crucial regulator of monocyte-endothelial cell adhesion and promote the atherosclerotic plaque formation induced by P. gingivalis.
Collapse
Affiliation(s)
- Yun Wu
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Wanyue Xu
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Jingya Hou
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Yanqing Liu
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Rong Li
- Department of Periodontics, School of Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Jingbo Liu
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Chen Li
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Xiaolin Tang
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Li Lin
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| | - Dongmei Zhang
- Department of Periodontics and Oral Biology, School of
Stomatology, China Medical University, Heping District, Nanjing North Street No.117, Shenyang, 110002 China
| |
Collapse
|
31
|
Silvério de Barros R, Dias GS, Paula do Rosario A, Paladino FV, Lopes GH, Campos AH. Gremlin-1 potentiates the dedifferentiation of VSMC in early stages of atherosclerosis. Differentiation 2019; 109:28-33. [PMID: 31494396 DOI: 10.1016/j.diff.2019.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/12/2019] [Accepted: 08/27/2019] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMC) are highly specialized, and exhibit a contractile phenotype when mature and fully differentiated, being responsible for vessel homeostasis and blood pressure control. In response to pro-atherogenic stimuli VSMC alter their state of differentiation, increase proliferation and migration, resulting in SMC phenotypes ranging from contractile to synthetic. This variability is observed in cell morphology and expression level of marker genes for differentiation status. There is growing evidence that bone morphogenetic protein (BMP) signaling is involved in vascular diseases, including atherosclerosis. Here, we evaluated in vitro the role of specific agonists/antagonists belonging to the BMP pathway on dedifferentiation of VSMC harvested during early stages of atherosclerosis. RESULTS: Comparing primary VSMC isolated from aortas of susceptible ApoE-/- animals fed 8 weeks of western diet with their littermate controls fed usual diet, we observed that recombinant BMP4 was able to reduce SM22-alpha and alpha actin gene expression indicating dedifferentiation was under way. Unexpectedly, treatment with recombinant Gremlin-1, a known BMP antagonist, also reduced 4-6.5 folds gene expression of SM22-alpha, alpha-actin and, calponin, exclusively in VSMC from ApoE-/- animals, independently on the diet consumed. CONCLUSION: Our data show that BMP4 is capable of modulating of SM22-alpha and alpha actin gene expression, indicative of cell dedifferentiation in VSMC. Additionally, we report for first time that Gremlin-1 acts independently of the BMP pathway and selectively on VSMC from susceptible animals, reducing the expression of all genes evaluated.
Collapse
Affiliation(s)
- Renata Silvério de Barros
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Grazielle Suhett Dias
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Ana Paula do Rosario
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | - Fernanda Vieira Paladino
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil
| | | | - Alexandre Holthausen Campos
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Centro de Pesquisa Experimental, Av Albert Einstein, 627. Morumbi, 2S/Bloco A, São Paulo, SP, CEP 05651-901, Brazil.
| |
Collapse
|
32
|
Mezzano S, Droguett A, Lavoz C, Krall P, Egido J, Ruiz-Ortega M. Gremlin and renal diseases: ready to jump the fence to clinical utility? Nephrol Dial Transplant 2019; 33:735-741. [PMID: 28992340 DOI: 10.1093/ndt/gfx194] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/18/2017] [Indexed: 12/31/2022] Open
Abstract
The current therapeutic strategy for the treatment of chronic kidney diseases only ameliorates disease progression. During renal injury, developmental genes are re-expressed and could be potential therapeutic targets. Among those genes reactivated in the adult damaged kidney, Gremlin is of particular relevance since recent data suggest that it could be a mediator of diabetic nephropathy and other progressive renal diseases. Earlier studies have shown that Gremlin is upregulated in trans-differentiated renal proximal tubular cells and in several chronic kidney diseases associated with fibrosis. However, not much was known about the mechanisms by which Gremlin acts in renal pathophysiology. The role of Gremlin as a bone morphogenetic protein antagonist has clearly been demonstrated in organogenesis and in fibrotic-related disorders. Gremlin binds to vascular endothelial growth factor receptor 2 (VEGFR2) in endothelial and tubular epithelial cells. Activation of the Gremlin-VEGFR2 axis was found in several human nephropathies. We have recently described that Gremlin activates the VEGFR2 signaling pathway in the kidney, eliciting a downstream mechanism linked to renal inflammatory response. Gremlin deletion improves experimental renal damage, diminishing fibrosis. Overall, the available data identify the Gremlin-VEGFR2 axis as a novel therapeutic target for kidney inflammation and fibrosis and provide a rationale for unveiling new concepts to investigate in several clinical conditions.
Collapse
Affiliation(s)
- Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alejandra Droguett
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Lavoz
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Paola Krall
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Jesús Egido
- Division of Nephrology and Hypertension, University Hospital, Fundación Jiménez Díaz-Universidad Autónoma, CIBERDEM, Instituto Renal Reina Sofía, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma Madrid, IIS-Fundación Jiménez Díaz, RedinRen, Madrid, Spain
| |
Collapse
|
33
|
Harris J, VanPatten S, Deen NS, Al-Abed Y, Morand EF. Rediscovering MIF: New Tricks for an Old Cytokine. Trends Immunol 2019; 40:447-462. [DOI: 10.1016/j.it.2019.03.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022]
|
34
|
Li JZ, Cao TH, Han JC, Qu H, Jiang SQ, Xie BD, Yan XL, Wu H, Liu XL, Zhang F, Leng XP, Kang K, Jiang SL. Comparison of adipose‑ and bone marrow‑derived stem cells in protecting against ox‑LDL‑induced inflammation in M1‑macrophage‑derived foam cells. Mol Med Rep 2019; 19:2660-2670. [PMID: 30720126 PMCID: PMC6423631 DOI: 10.3892/mmr.2019.9922] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 12/17/2018] [Indexed: 01/01/2023] Open
Abstract
Adipose‑derived stem cells (ADSCs) and bone marrow‑derived stem cells (BMSCs) are considered to be prospective sources of mesenchymal stromal cells (MSCs), that can be used in cell therapy for atherosclerosis. The present study investigated whether ADSCs co‑cultured with M1 foam macrophages via treatment with oxidized low‑density lipoprotein (ox‑LDL) would lead to similar or improved anti‑inflammatory effects compared with BMSCs. ADSCs, peripheral blood monocytes, BMSCs and ox‑LDL were isolated from ten coronary heart disease (CHD) patients. After three passages, the supernatants of the ADSCs and BMSCs were collected and systematically analysed by liquid chromatography‑quadrupole time‑of‑flight‑mass spectrometry (6530; Agilent Technologies, Inc., Santa Clara, CA, USA). Cis‑9, trans‑11 was deemed to be responsible for the potential differences in the metabolic characteristics of ADSCs and BMSCs. These peripheral blood monocytes were characterized using flow cytometry. Following peripheral blood monocytes differentiation into M1 macrophages, the formation of M1 foam macrophages was achieved through treatment with ox‑LDL. Overall, 2x106 ADSCs, BMSCs or BMSCs+cis‑9, trans‑11 were co‑cultured with M1 foam macrophages. Anti‑inflammatory capability, phagocytic activity, anti‑apoptotic capability and cell viability assays were compared among these groups. It was demonstrated that the accumulation of lipid droplets decreased following ADSCs, BMSCs or BMSCs+cis‑9, trans‑11 treatment in M1 macrophages derived from foam cells. Consistently, ADSCs exhibited great advantageous anti‑inflammatory capabilities, phagocytic activity, anti‑apoptotic capability activity and cell viability over BMSCs or BMSCs+cis‑9, trans‑11. Additionally, BMSCs+cis‑9, trans‑11 also demonstrated marked improvement in anti‑inflammatory capability, phagocytic activity, anti‑apoptotic capability activity and cell viability in comparison with BMSCs. The present results indicated that ADSCs would be more appropriate for transplantation to treat atherosclerosis than BMSCs alone or BMSCs+cis‑9, trans‑11. This may be an important mechanism to regulate macrophage immune function.
Collapse
Affiliation(s)
- Jian-Zhong Li
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Tian-Hui Cao
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Jin-Cheng Han
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Hui Qu
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Shuang-Quan Jiang
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Bao-Dong Xie
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Xiao-Long Yan
- Division of Thoracic Surgery, Tang Du Hospital of Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hua Wu
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Xiang-Lan Liu
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Fan Zhang
- Division of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Xiao-Ping Leng
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Kai Kang
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| | - Shu-Lin Jiang
- Division of Cardiovascular Surgery, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Education Ministry for Myocardial Ischemia, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
35
|
Gawaz M, Borst O. The Role of Platelets in Atherothrombosis. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00026-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
36
|
Rowan SC, Piouceau L, Cornwell J, Li L, McLoughlin P. EXPRESS: Gremlin1 blocks vascular endothelial growth factor signalling in the pulmonary microvascular endothelium. Pulm Circ 2018; 10:2045894018807205. [PMID: 30284507 PMCID: PMC7066471 DOI: 10.1177/2045894018807205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/20/2018] [Indexed: 11/15/2022] Open
Abstract
The bone morphogenetic protein (BMP) antagonist gremlin 1 plays a central role in the pathogenesis of hypoxic pulmonary hypertension (HPH). Recently, non-canonical functions of gremlin 1 have been identified, including specific binding to the vascular endothelial growth factor receptor-2 (VEGFR2). We tested the hypothesis that gremlin 1 modulates VEGFR2 signaling in the pulmonary microvascular endothelium. We examined the effect of gremlin 1 haploinsufficiency on the expression of VEGF responsive genes and proteins in the hypoxic (10% O2) murine lung in vivo. Using human microvascular endothelial cells in vitro we examined the effect of gremlin 1 on VEGF signaling. Gremlin 1 haploinsufficiency (Grem1+/–) attenuated the hypoxia-induced increase in gremlin 1 observed in the wild-type mouse lung. Reduced gremlin 1 expression in hypoxic Grem1+/– mice restored VEGFR2 expression and endothelial nitric oxide synthase (eNOS) expression and activity to normoxic values. Recombinant monomeric gremlin 1 inhibited VEGFA-induced VEGFR2 activation, downstream signaling, and VEGF-induced increases in Bcl-2, cell number, and the anti-apoptotic effect of VEGFA in vitro. These results show that the monomeric form of gremlin 1 acts as an antagonist of VEGFR2 activation in the pulmonary microvascular endothelium. Given the previous demonstration that inhibition of VEGFR2 causes marked worsening of HPH, our results suggest that increased gremlin 1 in the hypoxic lung, in addition to blocking BMP receptor type-2 (BMPR2) signaling, contributes importantly to the development of PH by a non-canonical VEGFR2 blocking activity.
Collapse
Affiliation(s)
- Simon C. Rowan
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Lucie Piouceau
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Joanna Cornwell
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Lili Li
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- UCD School of Medicine and Conway Institute,
University
College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Liu Y, Chen Z, Cheng H, Chen J, Qian J. Gremlin promotes retinal pigmentation epithelial (RPE) cell proliferation, migration and VEGF production via activating VEGFR2-Akt-mTORC2 signaling. Oncotarget 2018; 8:979-987. [PMID: 27894090 PMCID: PMC5352211 DOI: 10.18632/oncotarget.13518] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/29/2016] [Indexed: 11/25/2022] Open
Abstract
Retinopathy of prematurity (ROP) is characterized by late-phase pathologic retinal vasoproliferation. Gremlin is a novel vascular endothelial growth factors (VEGF) receptor 2 (VEGFR2) agonist and promotes angiogenic response. We demonstrated that gremlin expression was significantly increased in retinas of ROP model mice, which was correlated with VEGF upregulation. In retinal pigmentation epithelial (RPE) cells, gremlin activated VEGFR2-Akt-mTORC2 (mammalian target of rapamycin complex 2) signaling, and promoted cell proliferation, migration and VEGF production. VEGFR inhibition (by SU5416) or shRNA knockdown almost abolished gremlin-mediated pleiotropic functions in RPE cells. Further, pharmacological inhibition of Akt-mTOR, or shRNA knockdown of key mTORC2 component (Rictor or Sin1) also attenuated gremlin-exerted activities in RPE cells. We conclude that gremlin promotes RPE cell proliferation, migration and VEGF production possibly via activating VEGFR2-Akt-mTORC2 signaling. Gremlin could be a novel therapeutic target of ROP or other retinal vasoproliferation diseases.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Ophthalmology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zhijun Chen
- Department of Ophthalmology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Haixia Cheng
- Department of Ophthalmology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Chen
- Department of Ophthalmology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Qian
- Department of Ophthalmology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
38
|
Chatterjee M, Behrendt A, Schmid M, Beck S, Schneider M, Mack A, Müller I, Geisler T, Gawaz M. Platelets as a novel source of Gremlin-1: Implications for thromboinflammation. Thromb Haemost 2017; 117:311-324. [DOI: 10.1160/th16-08-0665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/08/2016] [Indexed: 11/05/2022]
Abstract
SummaryPlatelets mediating haemostasis-thrombosis are central players in coronary artery disease (CAD). We characterised platelets as a novel source of Gremlin-1. Platelets express Gremlin-1 like inflammatory and endothelial cells. Gremlin-1 co-localised with P-selectin containing randomly distributed α–granules under resting state, which were peripheralised following platelet activation or adhesion over fibrinogen-coated surface. Gremlin-1 release upon activation with ADP, CRP, and TRAP was detected as enhanced surface expression; also in activated platelet supernatant as detected by Western Blot following CRP activation and by ELISA upon activation with ADP, CRP, PAR-1, and PAR4 agonist. Recombinant (rh)Gremlin-1 synergistically enhanced CRP-triggered intracellular calcium mobilisation, ADP-TRAP induced platelet activation, aggregation, and thrombin-activation triggered apoptosis; also thrombus formation ex vivo. Intracellular localisation of macrophage migration inhibitory factor (MIF) and Gremlin-1 a high-affinity binding partner and functional antagonist of MIF were found in intracoronary thrombus sections from acute coronary syndrome (ACS) patients and showed moderate overlap in α-granules of platelets. Intra-platelet Gremlin-1 levels were significantly decreased in ACS patients as compared to stable CAD (n=235). rhGremlin-1 also counteracted the anti-apoptotic and anti-thrombotic effects of rhMIF on platelets. Platelet-derived-Gremlin-1 prompted monocyte migration, facilitated adhesion under static and dynamic arterial flow conditions to collagen-adherent activated platelets; supported monocyte survival against BH-3-mimetic–induced apoptosis and macrophage differentiation in monocyte-platelet co-culture system, which were counteracted upon Gremlin-1 neutralisation. Thus platelet derived Gremlin-1 might contribute to the elevated circulating levels of Gremlin-1 in ACS and serve as a thrombo-inflammatory mediator in cardiovascular pathophysiologies.
Collapse
|
39
|
Muñoz JC, Martín R, Alonso C, Gutiérrez B, Nieto ML. Relation between serum levels of chemotaxis-related factors and the presence of coronary artery calcification as expression of subclinical atherosclerosis. Clin Biochem 2017; 50:1048-1055. [PMID: 28830786 DOI: 10.1016/j.clinbiochem.2017.08.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/02/2017] [Accepted: 08/19/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Atherosclerotic plaque formation is characterized by recruitment of monocytes/macrophages, which contributes to its calcification by releasing pro-osteogenic cytokines. Chemotaxis-related proteins, including netrin-1, gremlin-1 and macrophage inflammatory protein-1β (MIP-1β), regulate immune cell migration. However, their relation with the presence of subclinical atherosclerosis, assessed by measures of coronary artery calcifications (CAC) in patients without known coronary artery disease (CAD), remains unclear. AIMS To examine whether these chemoattractant-related proteins are associated with the presence of CAC in patients without known CAD. METHODS A retrospective case-control observational study was conducted in 120 outpatients without CAD, undergoing a CAC evaluation by computed tomography with the Agatston Calcium score, categorized as CAC- (none) and CAC+ (≥1). Serum biomarkers were quantified by ELISA. RESULTS Lpa, dyslipidaemia and smoking were significantly higher (p=0.006, p≤0.0001 and p=0.001, respectively) in CAC+ patients. Serum netrin-1 levels were lower in CAC+ than in CAC- patients (196.8±127.8pg/ml versus 748.3±103.2pg/ml, p≤0.0001), and a similar pattern was found for gremlin-1 (1.14±0.39ng/ml versus 4.33±1.20ng/ml, p≤0.0001). However, TNFα and MIP-1β were strongly upregulated in CAC+ patients (447.56±74pg/ml versus 1104±144pg/ml and 402.00±94pg/ml versus 905.0±101.6pg/ml, respectively, p≤0.001). Multivariate analyses revealed that low netrin-1 and gremlin-1 levels and high TNFα and MIP-1β amounts were associated with CAC presence, after adjustment for clinical and biochemical variables. CONCLUSIONS We found a netrin-1 and gremlin-1 deficiency and a TNFα and MIP-1β overproduction in CAC+ patients' serum. These proteins may be used to identify individuals with subclinical atherosclerosis. Further research is warranted in a larger cohort of patients to establish these chemotactic-related proteins as biomarkers that improve CAD risk stratification.
Collapse
Affiliation(s)
- Juan Carlos Muñoz
- Servicio de Cardiología, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - Rubén Martín
- Instituto de Biología y Genética Molecular, CSIC-UVa, Valladolid, Spain
| | - Carmen Alonso
- Servicio de Radiología, Hospital Universitario Rio Hortega, Valladolid, Spain
| | - Beatriz Gutiérrez
- Instituto de Biología y Genética Molecular, CSIC-UVa, Valladolid, Spain
| | - María Luisa Nieto
- Instituto de Biología y Genética Molecular, CSIC-UVa, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV). Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
40
|
Müller KAL, Rath D, Schmid M, Schoenleber H, Gawaz M, Geisler T, Müller II. High Plasma Levels of Gremlin-1 and Macrophage Migration Inhibitory Factor, but Not Their Ratio, Indicate an Increased Risk for Acute Coronary Syndrome in Patients With Type 2 Diabetes Mellitus. Clin Cardiol 2017; 39:201-6. [PMID: 27101443 DOI: 10.1002/clc.22509] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/30/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Chronic inflammation promotes atherosclerosis and is a prognostic factor in coronary artery disease (CAD). Patients with type 2 diabetes mellitus (DM2) are at risk for progressive atherosclerosis. Macrophage migration inhibitory factor (MIF) is a key player in atherosclerosis, mediating pro-inflammatory responses. Its endogenous antagonist Gremlin-1 inhibits foam-cell formation and atheroprogression by binding MIF, neutralizing its proatherosclerotic functions. HYPOTHESIS Plasma levels of MIF and Gremlin-1 correlate with the stability of CAD in patients with DM2. METHOD We assessed plasma levels of Gremlin-1 and MIF in 198 nondiabetic and 88 diabetic patients with symptomatic CAD using enzyme-linked immunosorbent assays. RESULTS Plasma levels of Gremlin-1 were higher DM2 patients (278.8 ± 16.6 vs 224.7 ± 6.7 ng/mL; P = 0.001). MIF levels were elevated but not significantly increased in DM2 (P = 0.098). Interestingly, we found that Gremlin-1 plasma levels were significantly higher in diabetic patients with stable angina pectoris (SAP; n = 53) or acute coronary syndrome (ACS; n = 35) compared with nondiabetic patients with SAP (P = 0.008 and P = 0.011, respectively). MIF levels were significantly higher in diabetic patients with ACS compared with SAP (P < 0.001). Although the single plasma parameters showed an association with DM2 and CAD status, we could not confirm that the Gremlin-1/MIF ratio is significantly different in patients stratified by DM2 and CAD (P = 0.072). Hence, Gremlin-1/MIF ratio was significantly lower in patients with ACS compared with SAP (1.1 ± 0.1 vs 4.4 ± 1.1; P = 0.003). CONCLUSIONS Diabetic patients with ACS show increased levels of Gremlin-1 and MIF, leading to unfavorable Gremlin-1/MIF ratios. However, DM2 alone is not associated with low Gremlin-1/MIF ratios.
Collapse
Affiliation(s)
- Karin A L Müller
- Department of Cardiology and Circulatory Diseases, Internal Medicine Clinic III, Eberhard Karls University Tübingen, Tübingen, Germany.,La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Dominik Rath
- Department of Cardiology and Circulatory Diseases, Internal Medicine Clinic III, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Martina Schmid
- Department of Cardiology and Circulatory Diseases, Internal Medicine Clinic III, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Heiko Schoenleber
- Department of Cardiology and Circulatory Diseases, Internal Medicine Clinic III, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Circulatory Diseases, Internal Medicine Clinic III, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Circulatory Diseases, Internal Medicine Clinic III, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Iris I Müller
- Department of Cardiology and Circulatory Diseases, Internal Medicine Clinic III, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
41
|
Church RH, Ali I, Tate M, Lavin D, Krishnakumar A, Kok HM, Hombrebueno JR, Dunne PD, Bingham V, Goldschmeding R, Martin F, Brazil DP. Gremlin1 plays a key role in kidney development and renal fibrosis. Am J Physiol Renal Physiol 2017; 312:F1141-F1157. [PMID: 28100499 PMCID: PMC5495891 DOI: 10.1152/ajprenal.00344.2016] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 11/30/2016] [Accepted: 01/10/2017] [Indexed: 02/06/2023] Open
Abstract
Gremlin1 (Grem1), an antagonist of bone morphogenetic proteins, plays a key role in embryogenesis. A highly specific temporospatial gradient of Grem1 and bone morphogenetic protein signaling is critical to normal lung, kidney, and limb development. Grem1 levels are increased in renal fibrotic conditions, including acute kidney injury, diabetic nephropathy, chronic allograft nephropathy, and immune glomerulonephritis. We demonstrate that a small number of grem1−/− whole body knockout mice on a mixed genetic background (8%) are viable, with a single, enlarged left kidney and grossly normal histology. The grem1−/− mice displayed mild renal dysfunction at 4 wk, which recovered by 16 wk. Tubular epithelial cell-specific targeted deletion of Grem1 (TEC-grem1-cKO) mice displayed a milder response in the acute injury and recovery phases of the folic acid model. Increases in indexes of kidney damage were smaller in TEC-grem1-cKO than wild-type mice. In the recovery phase of the folic acid model, associated with renal fibrosis, TEC-grem1-cKO mice displayed reduced histological damage and an attenuated fibrotic gene response compared with wild-type controls. Together, these data demonstrate that Grem1 expression in the tubular epithelial compartment plays a significant role in the fibrotic response to renal injury in vivo.
Collapse
Affiliation(s)
- Rachel H Church
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Imran Ali
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Mitchel Tate
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Deborah Lavin
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Arjun Krishnakumar
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Helena M Kok
- Utrecht Medical Centre, Utrecht, The Netherlands
| | - Jose R Hombrebueno
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Philip D Dunne
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland
| | - Victoria Bingham
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland
| | | | - Finian Martin
- Conway Institute, University College Dublin, Dublin, Ireland; and
| | - Derek P Brazil
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom;
| |
Collapse
|
42
|
Ji C, Huang JW, Xu QY, Zhang J, Lin MT, Tu Y, He L, Bi ZG, Cheng B. Gremlin inhibits UV-induced skin cell damages via activating VEGFR2-Nrf2 signaling. Oncotarget 2016; 7:84748-84757. [PMID: 27713170 PMCID: PMC5356696 DOI: 10.18632/oncotarget.12454] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 09/23/2016] [Indexed: 12/14/2022] Open
Abstract
Ultra Violet (UV) radiation induces reactive oxygen species (ROS) production, DNA oxidation and single strand breaks (SSBs), which will eventually lead to skin cell damages or even skin cancer. Here, we tested the potential activity of gremlin, a novel vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) agonist, against UV-induced skin cell damages. We show that gremlin activated VEGFR2 and significantly inhibited UV-induced death and apoptosis of skin keratinocytes and fibroblasts. Pharmacological inhibition or shRNA-mediated knockdown of VEGFR2 almost abolished gremlin-mediated cytoprotection against UV in the skin cells. Further studies showed that gremlin activated VEGFR2 downstream NF-E2-related factor 2 (Nrf2) signaling, which appeared required for subsequent skin cell protection. Nrf2 shRNA knockdown or S40T dominant negative mutation largely inhibited gremlin-mediated skin cell protection against UV. At last, we show that gremlin dramatically inhibited UV-induced ROS production and DNA SSB formation in skin keratinocytes and fibroblasts. We conclude that gremlin protects skin cells from UV damages via activating VEGFR2-Nrf2 signaling. Gremlin could be further tested as a novel anti-UV skin protectant.
Collapse
Affiliation(s)
- Chao Ji
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Jin-wen Huang
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Qiu-yun Xu
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Jing Zhang
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Meng-ting Lin
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| | - Ying Tu
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Yunnan Provincial Institute of Dermatology, Kunming 650032, Yunnan, China
| | - Li He
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Yunnan Provincial Institute of Dermatology, Kunming 650032, Yunnan, China
| | - Zhi-gang Bi
- Department of Dermatology, BenQ Medical Center, Nanjing Medical University, Nanjing 210019, Jiangsu, China
| | - Bo Cheng
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| |
Collapse
|
43
|
Xiang Q, Hong D, Liao Y, Cao Y, Liu M, Pang J, Zhou J, Wang G, Yang R, Wang M, Xiang AP. Overexpression of Gremlin1 in Mesenchymal Stem Cells Improves Hindlimb Ischemia in Mice by Enhancing Cell Survival. J Cell Physiol 2016; 232:996-1007. [PMID: 27579673 DOI: 10.1002/jcp.25578] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 08/29/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Qiuling Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Sun Yat-sen University; Guangzhou Guangdong China
- Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou Guangdong China
| | - Dongxi Hong
- Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou Guangdong China
| | - Yan Liao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Sun Yat-sen University; Guangzhou Guangdong China
| | - Yong Cao
- Cardiovascular Center; Gaozhou People's Hospital; Maoming Guangdong China
| | - Muyun Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Sun Yat-sen University; Guangzhou Guangdong China
| | - Jun Pang
- Guizhou Provincial People's Hospital; Guizhou China
| | - Junjie Zhou
- Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou Guangdong China
| | - Guang Wang
- Division of Histology and Embryology; Medical College, Jinan University; Guangzhou China
| | - Renhao Yang
- Division of Histology and Embryology; Medical College, Jinan University; Guangzhou China
| | - Maosheng Wang
- Cardiovascular Center; Gaozhou People's Hospital; Maoming Guangdong China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Sun Yat-sen University; Guangzhou Guangdong China
- Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou Guangdong China
- Biotherapy Center, The Third Affiliated Hospital; Sun Yat-sen University; Guangzhou Guangdong China
| |
Collapse
|
44
|
Abstract
The discovery of the transforming growth factor β (TGF-β) family ligands and the realization that their bioactivities need to be tightly controlled temporally and spatially led to intensive research that has identified a multitude of extracellular modulators of TGF-β family ligands, uncovered their functions in developmental and pathophysiological processes, defined the mechanisms of their activities, and explored potential modulator-based therapeutic applications in treating human diseases. These studies revealed a diverse repertoire of extracellular and membrane-associated molecules that are capable of modulating TGF-β family signals via control of ligand availability, processing, ligand-receptor interaction, and receptor activation. These molecules include not only soluble ligand-binding proteins that were conventionally considered as agonists and antagonists of TGF-β family of growth factors, but also extracellular matrix (ECM) proteins and proteoglycans that can serve as "sink" and control storage and release of both the TGF-β family ligands and their regulators. This extensive network of soluble and ECM modulators helps to ensure dynamic and cell-specific control of TGF-β family signals. This article reviews our knowledge of extracellular modulation of TGF-β growth factors by diverse proteins and their molecular mechanisms to regulate TGF-β family signaling.
Collapse
Affiliation(s)
- Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
45
|
Koli K, Sutinen E, Rönty M, Rantakari P, Fortino V, Pulkkinen V, Greco D, Sipilä P, Myllärniemi M. Gremlin-1 Overexpression in Mouse Lung Reduces Silica-Induced Lymphocyte Recruitment - A Link to Idiopathic Pulmonary Fibrosis through Negative Correlation with CXCL10 Chemokine. PLoS One 2016; 11:e0159010. [PMID: 27428020 PMCID: PMC4948891 DOI: 10.1371/journal.pone.0159010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/25/2016] [Indexed: 12/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by activation and injury of epithelial cells, the accumulation of connective tissue and changes in the inflammatory microenvironment. The bone morphogenetic protein (BMP) inhibitor protein gremlin-1 is associated with the progression of fibrosis both in human and mouse lung. We generated a transgenic mouse model expressing gremlin-1 in type II lung epithelial cells using the surfactant protein C (SPC) promoter and the Cre-LoxP system. Gremlin-1 protein expression was detected specifically in the lung after birth and did not result in any signs of respiratory insufficiency. Exposure to silicon dioxide resulted in reduced amounts of lymphocyte aggregates in transgenic lungs while no alteration in the fibrotic response was observed. Microarray gene expression profiling and analyses of bronchoalveolar lavage fluid cytokines indicated a reduced lymphocytic response and a downregulation of interferon-induced gene program. Consistent with reduced Th1 response, there was a downregulation of the mRNA and protein expression of the anti-fibrotic chemokine CXCL10, which has been linked to IPF. In human IPF patient samples we also established a strong negative correlation in the mRNA expression levels of gremlin-1 and CXCL10. Our results suggest that in addition to regulation of epithelial-mesenchymal crosstalk during tissue injury, gremlin-1 modulates inflammatory cell recruitment and anti-fibrotic chemokine production in the lung.
Collapse
Affiliation(s)
- Katri Koli
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
- * E-mail:
| | - Eva Sutinen
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center, Department of Pulmonary Medicine, Helsinki, Finland
| | - Mikko Rönty
- Department of Pathology, University of Helsinki and Fimlab laboratories, Pathology, Tampere, Finland
| | - Pia Rantakari
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Vittorio Fortino
- Unit of Systems Toxicology and Nanosafety Centre, Finnish Institute of Occupational Health (FIOH), Helsinki, Finland
| | - Ville Pulkkinen
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center, Department of Pulmonary Medicine, Helsinki, Finland
| | - Dario Greco
- Unit of Systems Toxicology and Nanosafety Centre, Finnish Institute of Occupational Health (FIOH), Helsinki, Finland
| | - Petra Sipilä
- Department of Physiology, Institute of Biomedicine and Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Marjukka Myllärniemi
- Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center, Department of Pulmonary Medicine, Helsinki, Finland
| |
Collapse
|
46
|
Structure of Gremlin-1 and analysis of its interaction with BMP-2. Biochem J 2016; 473:1593-604. [PMID: 27036124 PMCID: PMC4888461 DOI: 10.1042/bcj20160254] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/01/2016] [Indexed: 11/17/2022]
Abstract
We have determined the crystal structure of Gremlin-1 and analysed its interaction with BMP-2. Our results suggest that Gremlin-1 does not inhibit BMP-2 by direct 1:1 binding, but possibly has a novel mechanism of sequestering BMP-2 into a larger oligomeric complex. Bone morphogenetic protein 2 (BMP-2) is a member of the transforming growth factor-β (TGF-β) signalling family and has a very broad biological role in development. Its signalling is regulated by many effectors: transmembrane proteins, membrane-attached proteins and soluble secreted antagonists such as Gremlin-1. Very little is known about the molecular mechanism by which Gremlin-1 and other DAN (differential screening-selected gene aberrative in neuroblastoma) family proteins inhibit BMP signalling. We analysed the interaction of Gremlin-1 with BMP-2 using a range of biophysical techniques, and used mutagenesis to map the binding site on BMP-2. We have also determined the crystal structure of Gremlin-1, revealing a similar conserved dimeric structure to that seen in other DAN family inhibitors. Measurements using biolayer interferometry (BLI) indicate that Gremlin-1 and BMP-2 can form larger complexes, beyond the expected 1:1 stoichiometry of dimers, forming oligomers that assemble in alternating fashion. These results suggest that inhibition of BMP-2 by Gremlin-1 occurs by a mechanism that is distinct from other known inhibitors such as Noggin and Chordin and we propose a novel model of BMP-2–Gremlin-1 interaction yet not seen among any BMP antagonists, and cannot rule out that several different oligomeric states could be found, depending on the concentration of the two proteins.
Collapse
|
47
|
Chatterjee M, von Ungern-Sternberg SNI, Seizer P, Schlegel F, Büttcher M, Sindhu NA, Müller S, Mack A, Gawaz M. Platelet-derived CXCL12 regulates monocyte function, survival, differentiation into macrophages and foam cells through differential involvement of CXCR4-CXCR7. Cell Death Dis 2015; 6:e1989. [PMID: 26583329 PMCID: PMC4670914 DOI: 10.1038/cddis.2015.233] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 06/26/2015] [Accepted: 07/15/2015] [Indexed: 01/18/2023]
Abstract
Platelets store and release CXCL12 (SDF-1), which governs differentiation of hematopoietic progenitors into either endothelial or macrophage-foam cells. CXCL12 ligates CXCR4 and CXCR7 and regulates monocyte/macrophage functions. This study deciphers the relative contribution of CXCR4-CXCR7 in mediating the effects of platelet-derived CXCL12 on monocyte function, survival, and differentiation. CXCL12 and macrophage migration inhibitory factor (MIF) that ligate CXCR4-CXCR7 induced a dynamic bidirectional trafficking of the receptors, causing CXCR4 internalization and CXCR7 externalization during chemotaxis, thereby influencing relative receptor availability, unlike MCP-1. In vivo we found enhanced accumulation of platelets and platelet-macrophage co-aggregates in peritoneal fluid following induction of peritonitis in mice. The relative surface expression of CXCL12, CXCR4, and CXCR7 among infiltrated monocytes was also enhanced as compared with peripheral blood. Platelet-derived CXCL12 from collagen-adherent platelets and recombinant CXCL12 induced monocyte chemotaxis specifically through CXCR4 engagement. Adhesion of monocytes to immobilized CXCL12 and CXCL12-enriched activated platelet surface under static and dynamic arterial flow conditions were mediated primarily through CXCR7 and were counter-regulated by neutralizing platelet-derived CXCL12. Monocytes and culture-derived-M1-M2 macrophages phagocytosed platelets, with the phagocytic potential of culture-derived-M1 macrophages higher than M2 involving CXCR4-CXCR7 participation. CXCR7 was the primary receptor in promoting monocyte survival as exerted by platelet-derived CXCL12 against BH3-mimetic induced apoptosis (phosphatidylserine exposure, caspase-3 activation, loss of mitochondrial transmembrane potential). In co-culture experiments with platelets, monocytes predominantly differentiated into CD163(+) macrophages, which was attenuated upon CXCL12 neutralization and CXCR4/CXCR7 blocking antibodies. Moreover, OxLDL uptake by platelets induced platelet apoptosis, like other platelet agonists TRAP and collagen-related peptide (CRP). CXCL12 facilitated phagocytosis of apoptotic platelets by monocytes and M1-M2 macrophages, also promoted their differentiation into foam cells via CXCR4 and CXCR7. Thus, platelet-derived CXCL12 could regulate monocyte-macrophage functions through differential engagement of CXCR4 and CXCR7, indicating an important role in inflammation at site of platelet accumulation.
Collapse
Affiliation(s)
- M Chatterjee
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| | - S N I von Ungern-Sternberg
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| | - P Seizer
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| | - F Schlegel
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| | - M Büttcher
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| | - N A Sindhu
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| | - S Müller
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| | - A Mack
- Institute of Anatomy, Universität Tübingen, Neuroanatomie, 72074 Tübingen, Germany
| | - M Gawaz
- Medizinische Klinik III, Kardiologie und Kreislauferkrankungen, Universität Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
48
|
Lavoz C, Alique M, Rodrigues-Diez R, Pato J, Keri G, Mezzano S, Egido J, Ruiz-Ortega M. Gremlin regulates renal inflammation via the vascular endothelial growth factor receptor 2 pathway. J Pathol 2015; 236:407-20. [PMID: 25810250 DOI: 10.1002/path.4537] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 01/03/2023]
Abstract
Inflammation is a main feature of progressive kidney disease. Gremlin binds to bone morphogenetic proteins (BMPs), acting as an antagonist and regulating nephrogenesis and fibrosis among other processes. Gremlin also binds to vascular endothelial growth factor receptor-2 (VEGFR2) in endothelial cells to induce angiogenesis. In renal cells, gremlin regulates proliferation and fibrosis, but there are no data about inflammatory-related events. We have investigated the direct effects of gremlin in the kidney, evaluating whether VEGFR2 is a functional gremlin receptor. Administration of recombinant gremlin to murine kidneys induced rapid and sustained activation of VEGFR2 signalling, located in proximal tubular epithelial cells. Gremlin bound to VEGFR2 in these cells in vitro, activating this signalling pathway independently of its action as an antagonist of BMPs. In vivo, gremlin caused early renal damage, characterized by activation of the nuclear factor (NF)-κB pathway linked to up-regulation of pro-inflammatory factors and infiltration of immune inflammatory cells. VEGFR2 blockade diminished gremlin-induced renal inflammatory responses. The link between gremlin/VEGFR2 and NF-κB/inflammation was confirmed in vitro. Gremlin overexpression was associated with VEGFR2 activation in human renal disease and in the unilateral ureteral obstruction experimental model, where VEGFR2 kinase inhibition diminished renal inflammation. Our data show that a gremlin/VEGFR2 axis participates in renal inflammation and could be a novel target for kidney disease.
Collapse
Affiliation(s)
- Carolina Lavoz
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | - Matilde Alique
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | - Raquel Rodrigues-Diez
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| | | | - Gyorgy Keri
- VichemChemie Ltd, Budapest, Hungary.,MTA-SE Pathobiochemistry Research Group, Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Jesús Egido
- Division of Nephrology and Hypertension. IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, CIBERDEM, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, REDINREN, Spain
| |
Collapse
|
49
|
Drechsler M, Duchene J, Soehnlein O. Chemokines Control Mobilization, Recruitment, and Fate of Monocytes in Atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1050-5. [DOI: 10.1161/atvbaha.114.304649] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/05/2015] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of large arteries and, among others, characterized by continuous influx of monocytes into the subendothelial space, subsequent macrophage accumulation, and foam cell formation. Chemokines and their receptors tightly orchestrate monocyte trafficking and fate from birth to death. This brief review summarizes our current understanding of the interplay between monocytes and chemokines entertaining crucial processes in atherosclerosis development, progression, and regression.
Collapse
Affiliation(s)
- Maik Drechsler
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.D., J.D., O.S.); Department of Pathology, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands (M.D., O.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (M.D., O.S.)
| | - Johan Duchene
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.D., J.D., O.S.); Department of Pathology, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands (M.D., O.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (M.D., O.S.)
| | - Oliver Soehnlein
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (M.D., J.D., O.S.); Department of Pathology, Academic Medical Center Amsterdam, University of Amsterdam, Amsterdam, The Netherlands (M.D., O.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (M.D., O.S.)
| |
Collapse
|
50
|
Chatterjee M, Borst O, Walker B, Fotinos A, Vogel S, Seizer P, Mack A, Alampour-Rajabi S, Rath D, Geisler T, Lang F, Langer HF, Bernhagen J, Gawaz M. Macrophage Migration Inhibitory Factor Limits Activation-Induced Apoptosis of Platelets via CXCR7-Dependent Akt Signaling. Circ Res 2014; 115:939-49. [DOI: 10.1161/circresaha.115.305171] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Madhumita Chatterjee
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Oliver Borst
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Britta Walker
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Anna Fotinos
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Sebastian Vogel
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Peter Seizer
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Andreas Mack
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Setareh Alampour-Rajabi
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Dominik Rath
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Tobias Geisler
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Florian Lang
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Harald F. Langer
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Jürgen Bernhagen
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| | - Meinrad Gawaz
- From the Medizinische Klinik III, Kardiologie und Kreislauferkrankungen (M.C., O.B., A.F., S.V., P.S., D.R., T.G., H.F.L., M.G.), Institute of Anatomy (A.M.), and Institute of Physiology (B.W., F.L.), Universität Tübingen, Tübingen, Germany; and Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Uniklinik RWTH Aachen, Aachen, Germany (S.A.-R., J.B.)
| |
Collapse
|