1
|
Huang J, Liao C, Yang J, Zhang L. The role of vascular and lymphatic networks in bone and joint homeostasis and pathology. Front Endocrinol (Lausanne) 2024; 15:1465816. [PMID: 39324127 PMCID: PMC11422228 DOI: 10.3389/fendo.2024.1465816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
The vascular and lymphatic systems are integral to maintaining skeletal homeostasis and responding to pathological conditions in bone and joint tissues. This review explores the interplay between blood vessels and lymphatic vessels in bones and joints, focusing on their roles in homeostasis, regeneration, and disease progression. Type H blood vessels, characterized by high expression of CD31 and endomucin, are crucial for coupling angiogenesis with osteogenesis, thus supporting bone homeostasis and repair. These vessels facilitate nutrient delivery and waste removal, and their dysfunction can lead to conditions such as ischemia and arthritis. Recent discoveries have highlighted the presence and significance of lymphatic vessels within bone tissue, challenging the traditional view that bones are devoid of lymphatics. Lymphatic vessels contribute to interstitial fluid regulation, immune cell trafficking, and tissue repair through lymphangiocrine signaling. The pathological alterations in these networks are closely linked to inflammatory joint diseases, emphasizing the need for further research into their co-regulatory mechanisms. This comprehensive review summarizes the current understanding of the structural and functional aspects of vascular and lymphatic networks in bone and joint tissues, their roles in homeostasis, and the implications of their dysfunction in disease. By elucidating the dynamic interactions between these systems, we aim to enhance the understanding of their contributions to skeletal health and disease, potentially informing the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jingxiong Huang
- Center of Stomatology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
| | - Chengcheng Liao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Guizhou, Zunyi, China
| | - Jian Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Liang Zhang
- Center of Stomatology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Brauer E, Herrera A, Fritsche-Guenther R, Görlitz S, Leemhuis H, Knaus P, Kirwan JA, Duda GN, Petersen A. Mechanical heterogeneity in a soft biomaterial niche controls BMP2 signaling. Biomaterials 2024; 309:122614. [PMID: 38788455 DOI: 10.1016/j.biomaterials.2024.122614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
The extracellular matrix is known to impact cell function during regeneration by modulating growth factor signaling. However, how the mechanical properties and structure of biomaterials can be used to optimize the cellular response to growth factors is widely neglected. Here, we engineered a macroporous biomaterial to study cellular signaling in environments that mimic the mechanical stiffness but also the mechanical heterogeneity of native extracellular matrix. We found that the mechanical interaction of cells with the heterogeneous and non-linear deformation properties of soft matrices (E < 5 kPa) enhances BMP-2 growth factor signaling with high relevance for tissue regeneration. In contrast, this effect is absent in homogeneous hydrogels that are often used to study cell responses to mechanical cues. Live cell imaging and in silico finite element modeling further revealed that a subpopulation of highly active, fast migrating cells is responsible for most of the material deformation, while a second, less active population experiences this deformation as an extrinsic mechanical stimulation. At an overall low cell density, the active cell population dominates the process, suggesting that it plays a particularly important role in early tissue healing scenarios where cells invade tissue defects or implanted biomaterials. Taken together, our findings demonstrate that the mechanical heterogeneity of the natural extracellular matrix environment plays an important role in triggering regeneration by endogenously acting growth factors. This suggests the inclusion of such mechanical complexity as a design parameter in future biomaterials, in addition to established parameters such as mechanical stiffness and stress relaxation.
Collapse
Affiliation(s)
- Erik Brauer
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Berlin School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany; BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Aaron Herrera
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Berlin School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany; BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Raphaela Fritsche-Guenther
- BIH Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Görlitz
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Berlin School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany; BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | | | - Petra Knaus
- Berlin School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany; Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Jennifer A Kirwan
- BIH Metabolomics Platform, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Berlin School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany; BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Ansgar Petersen
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Berlin School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany; BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany.
| |
Collapse
|
3
|
Tian S, Evans PC. BMPER regulates arterial adaptation to flow. NATURE CARDIOVASCULAR RESEARCH 2024; 3:777-779. [PMID: 39196180 DOI: 10.1038/s44161-024-00506-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Siyu Tian
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts & The London Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Paul C Evans
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts & The London Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
4
|
Singh B, Cui K, Eisa-Beygi S, Zhu B, Cowan DB, Shi J, Wang DZ, Liu Z, Bischoff J, Chen H. Elucidating the crosstalk between endothelial-to-mesenchymal transition (EndoMT) and endothelial autophagy in the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 155:107368. [PMID: 38548093 PMCID: PMC11303600 DOI: 10.1016/j.vph.2024.107368] [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: 01/17/2024] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis, a chronic systemic inflammatory condition, is implicated in most cardiovascular ischemic events. The pathophysiology of atherosclerosis involves various cell types and associated processes, including endothelial cell activation, monocyte recruitment, smooth muscle cell migration, involvement of macrophages and foam cells, and instability of the extracellular matrix. The process of endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a pivotal process in mediating vascular inflammation associated with atherosclerosis. This transition occurs gradually, with a significant portion of endothelial cells adopting an intermediate state, characterized by a partial loss of endothelial-specific gene expression and the acquisition of "mesenchymal" traits. Consequently, this shift disrupts endothelial cell junctions, increases vascular permeability, and exacerbates inflammation, creating a self-perpetuating cycle that drives atherosclerotic progression. While endothelial cell dysfunction initiates the development of atherosclerosis, autophagy, a cellular catabolic process designed to safeguard cells by recycling intracellular molecules, is believed to exert a significant role in plaque development. Identifying the pathological mechanisms and molecular mediators of EndoMT underpinning endothelial autophagy, may be of clinical relevance. Here, we offer new insights into the underlying biology of atherosclerosis and present potential molecular mechanisms of atherosclerotic resistance and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Bandana Singh
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Douglas B Cowan
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jinjun Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
5
|
Verdura S, Encinar JA, Gratchev A, Llop-Hernández À, López J, Serrano-Hervás E, Teixidor E, López-Bonet E, Martin-Castillo B, Micol V, Bosch-Barrera J, Cuyàs E, Menendez JA. Silibinin is a suppressor of the metastasis-promoting transcription factor ID3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155493. [PMID: 38484626 DOI: 10.1016/j.phymed.2024.155493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND ID3 (inhibitor of DNA binding/differentiation-3) is a transcription factor that enables metastasis by promoting stem cell-like properties in endothelial and tumor cells. The milk thistle flavonolignan silibinin is a phytochemical with anti-metastatic potential through largely unknown mechanisms. HYPOTHESIS/PURPOSE We have mechanistically investigated the ability of silibinin to inhibit the aberrant activation of ID3 in brain endothelium and non-small cell lung cancer (NSCLC) models. METHODS Bioinformatic analyses were performed to investigate the co-expression correlation between ID3 and bone morphogenic protein (BMP) ligands/BMP receptors (BMPRs) genes in NSCLC patient datasets. ID3 expression was assessed by immunoblotting and qRT-PCR. Luciferase reporter assays were used to evaluate the gene sequences targeted by silibinin to regulate ID3 transcription. In silico computational modeling and LanthaScreen TR-FRET kinase assays were used to characterize and validate the BMPR inhibitory activity of silibinin. Tumor tissues from NSCLC xenograft models treated with oral silibinin were used to evaluate the in vivo anti-ID3 effects of silibinin. RESULTS Analysis of lung cancer patient datasets revealed a top-ranked positive association of ID3 with the BMP9 endothelial receptor ACVRL1/ALK1 and the BMP ligand BMP6. Silibinin treatment blocked the BMP9-induced activation of the ALK1-phospho-SMAD1/5-ID3 axis in brain endothelial cells. Constitutive, acquired, and adaptive expression of ID3 in NSCLC cells were all significantly downregulated in response to silibinin. Silibinin blocked ID3 transcription via BMP-responsive elements in ID3 gene enhancers. Silibinin inhibited the kinase activities of BMPRs in the micromolar range, with the lower IC50 values occurring against ACVRL1/ALK1 and BMPR2. In an in vivo NSCLC xenograft model, tumoral overexpression of ID3 was completely suppressed by systematically achievable oral doses of silibinin. CONCLUSIONS ID3 is a largely undruggable metastasis-promoting transcription factor. Silibinin is a novel suppressor of ID3 that may be explored as a novel therapeutic approach to interfere with the metastatic dissemination capacity of NSCLC.
Collapse
Affiliation(s)
- Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - José Antonio Encinar
- Institute of Research, Development and Innovation in Health Biotechnology of Elche (IDiBE), Universitas Miguel Hernández (UMH), Elche 03202, Spain
| | - Alexei Gratchev
- Laboratory for Tumor Stromal Cell Biology, Institute of Carcinogenesis, Nikolaj Nikolajevich (N.N.) Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia
| | - Àngela Llop-Hernández
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Júlia López
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Eila Serrano-Hervás
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Eduard Teixidor
- Precision Oncology Group (OncoGir-Pro), Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Medical Oncology, Catalan Institute of Oncology, Girona, 17007, Spain
| | - Eugeni López-Bonet
- Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona 17007, Spain
| | - Begoña Martin-Castillo
- Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Unit of Clinical Research, Catalan Institute of Oncology, Girona, 17007, Spain
| | - Vicente Micol
- Institute of Research, Development and Innovation in Health Biotechnology of Elche (IDiBE), Universitas Miguel Hernández (UMH), Elche 03202, Spain; CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
| | - Joaquim Bosch-Barrera
- Precision Oncology Group (OncoGir-Pro), Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Medical Oncology, Catalan Institute of Oncology, Girona, 17007, Spain; Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain.
| |
Collapse
|
6
|
Lang A, Benn A, Collins JM, Wolter A, Balcaen T, Kerckhofs G, Zwijsen A, Boerckel JD. Endothelial SMAD1/5 signaling couples angiogenesis to osteogenesis in juvenile bone. Commun Biol 2024; 7:315. [PMID: 38480819 PMCID: PMC10937971 DOI: 10.1038/s42003-024-05915-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Skeletal development depends on coordinated angiogenesis and osteogenesis. Bone morphogenetic proteins direct bone formation in part by activating SMAD1/5 signaling in osteoblasts. However, the role of SMAD1/5 in skeletal endothelium is unknown. Here, we found that endothelial cell-conditional SMAD1/5 depletion in juvenile mice caused metaphyseal and diaphyseal hypervascularity, resulting in altered trabecular and cortical bone formation. SMAD1/5 depletion induced excessive sprouting and disrupting the morphology of the metaphyseal vessels, with impaired anastomotic loop formation at the chondro-osseous junction. Endothelial SMAD1/5 depletion impaired growth plate resorption and, upon long-term depletion, abrogated osteoprogenitor recruitment to the primary spongiosa. Finally, in the diaphysis, endothelial SMAD1/5 activity was necessary to maintain the sinusoidal phenotype, with SMAD1/5 depletion inducing formation of large vascular loops and elevated vascular permeability. Together, endothelial SMAD1/5 activity sustains skeletal vascular morphogenesis and function and coordinates growth plate remodeling and osteoprogenitor recruitment dynamics in juvenile mouse bone.
Collapse
Affiliation(s)
- Annemarie Lang
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany.
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden (TUD), Fetscherstrasse 74, Dresden, 01307, Germany.
| | - Andreas Benn
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, 3000, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Leuven, 3000, Belgium
| | - Joseph M Collins
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Angelique Wolter
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, 10117, Germany
- Department of Veterinary Medicine, Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Freie Universität Berlin, Berlin, 14163, Germany
| | - Tim Balcaen
- Institute of Mechanics, Materials and Civil Engineering, Biomechanics lab, UCLouvain, Louvain-la-Neuve, 1348, Belgium
- Institute of Experimental and Clinical Research, Pole of Morphology, UCLouvain, Brussels, 1200, Belgium
- KU Leuven, Department of Chemistry, Sustainable Chemistry for Metals and Molecules, Leuven, 3000, Belgium
| | - Greet Kerckhofs
- Institute of Mechanics, Materials and Civil Engineering, Biomechanics lab, UCLouvain, Louvain-la-Neuve, 1348, Belgium
- Institute of Experimental and Clinical Research, Pole of Morphology, UCLouvain, Brussels, 1200, Belgium
- Department of Materials Engineering, KU Leuven, Heverlee, 3001, Belgium
- Division for Skeletal Tissue Engineering, Prometheus, KU Leuven, Leuven, 3000, Belgium
| | - An Zwijsen
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, 3000, Belgium
| | - Joel D Boerckel
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Zhang W, Hu M, Yang L, Zhou Z, Chen R, Liu Z. Expression pattern of the bone morphogenetic protein-4 and its relationship with inflammation, vascular injury in patients suffered the arterial occlusive diseases. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:279-285. [PMID: 38755724 PMCID: PMC11103063 DOI: 10.11817/j.issn.1672-7347.2024.230264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Indexed: 05/18/2024]
Abstract
OBJECTIVES Bone morphogenetic protein-4 (BMP4) has been proved to be an important regulatory factor for the pathological process of atherosclerosis (AS). However, there are few related clinical studies. This study aims to investigate the levels of plasma BMP4 in patients suffering from the arterial occlusive diseases (ACD) characterized by AS, and further to test the relationship between BMP4 and inflammation and vascular injury. METHODS A total of 38 ACD patients (the ACD group) and 38 healthy people for the physical examination (the control group) were enrolled. The plasma in each subject from both groups was obtained to test the levels of BMP4, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-10, and vascular endothelial cadherin (VE-cadherin), and the relationship between BMP4 and the detected indicators above were further analyzed. RESULTS Compared with the control group, the patients in the ACD group displayed significant elevations in the neutrophil to lymphocyte ratio [NLR, 1.63 (1.26, 1.91) vs 3.43 (2.16, 6.61)] and platelet to lymphocyte ratio [PLR, 6.37 (5.26, 7.74) vs 15.79 (7.97, 20.53)], while decrease in the lymphocyte to monocyte ratio [LMR, 5.67 (4.41, 7.14) vs 3.43 (2.07, 3.74)] (all P<0.05). Besides, the ACD patients displayed significant elevations in plasma BMP4 [581.26 (389.85, 735.64) pg/mL vs 653.97(510.95, 890.43) pg/mL], TNF-α [254.16 (182.96, 340.70) pg/mL vs 293.29(238.90, 383.44) pg/mL], and VE-cadherin [1.54 (1.08, 2.13) ng/mL vs 1.85 (1.30, 2.54) ng/mL], and decrease in IL-10 [175.89 (118.39, 219.25) pg/mL vs 135.92 (95.80, 178.04) pg/mL] (all P<0.05). While the levels of IL-1β remained statistically comparable between the 2 groups (P=0.09). Furthermore, the plasma BMP4 levels were further revealed to be positively correlated with the levels of IL-1β (r=0.35), TNF-α (r=0.31) and VE-cadherin (r=0.47), while they were negatively correlated with the levels of IL-10 (r=-0.37; all P<0.01). CONCLUSIONS After ACD occurrence, the patients' plasma concentrations of BMP4 would be upregulated, which may serve as a candidate to indicate the levels of inflammation and vascular injury.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Department of Laboratory, Second Xiangya Hospital, Central South University, Changsha 410011.
| | - Min Hu
- Department of Laboratory, Second Xiangya Hospital, Central South University, Changsha 410011
| | - Lin Yang
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha 410011
| | - Zhifang Zhou
- Department of Laboratory, Second Xiangya Hospital, Central South University, Changsha 410011
| | - Ruohong Chen
- Department of Laboratory, Second Xiangya Hospital, Central South University, Changsha 410011.
| | - Zhaoyang Liu
- Department of Laboratory, Second Xiangya Hospital, Central South University, Changsha 410011.
- Office of the CPC Committee, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
8
|
Jatzlau J, Mendez PL, Altay A, Raaz L, Zhang Y, Mähr S, Sesver A, Reichenbach M, Mundlos S, Vingron M, Knaus P. Fluid shear stress-modulated chromatin accessibility reveals the mechano-dependency of endothelial SMAD1/5-mediated gene transcription. iScience 2023; 26:107405. [PMID: 37680470 PMCID: PMC10481294 DOI: 10.1016/j.isci.2023.107405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 09/09/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling and fluid shear stress (FSS) mediate complementary functions in vascular homeostasis and disease development. It remains to be shown whether altered chromatin accessibility downstream of BMP and FSS offers a crosstalk level to explain changes in SMAD-dependent transcription. Here, we employed ATAC-seq to analyze arterial endothelial cells stimulated with BMP9 and/or FSS. We found that BMP9-sensitive regions harbor non-palindromic GC-rich SMAD-binding elements (GGCTCC) and 69.7% of these regions become BMP-insensitive in the presence of FSS. While GATA and KLF transcription factor (TF) motifs are unique to BMP9- and FSS-sensitive regions, respectively, SOX motifs are common to both. Finally, we show that both SOX(13/18) and GATA(2/3/6) family members are directly upregulated by SMAD1/5. These findings highlight the mechano-dependency of SMAD-signaling by a sequential mechanism of first elevated pioneer TF expression, allowing subsequent chromatin opening to eventually providing accessibility to novel SMAD binding sites.
Collapse
Affiliation(s)
- Jerome Jatzlau
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Paul-Lennard Mendez
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
| | - Aybuge Altay
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Lion Raaz
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
- Institute of Medical and Human Genetics, Charité Universitätsmedizin, 13353 Berlin, Germany
| | - Yufei Zhang
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Sophia Mähr
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Akin Sesver
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Maria Reichenbach
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
- Institute of Medical and Human Genetics, Charité Universitätsmedizin, 13353 Berlin, Germany
| | - Martin Vingron
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
| | - Petra Knaus
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), 13353 Berlin, Germany
- International Max-Planck Research School for Biology AND Computation (IMPRS-BAC), 14195 Berlin, Germany
| |
Collapse
|
9
|
Sánchez-Duffhues G, Hiepen C. Human iPSCs as Model Systems for BMP-Related Rare Diseases. Cells 2023; 12:2200. [PMID: 37681932 PMCID: PMC10487005 DOI: 10.3390/cells12172200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Disturbances in bone morphogenetic protein (BMP) signalling contribute to onset and development of a number of rare genetic diseases, including Fibrodysplasia ossificans progressiva (FOP), Pulmonary arterial hypertension (PAH), and Hereditary haemorrhagic telangiectasia (HHT). After decades of animal research to build a solid foundation in understanding the underlying molecular mechanisms, the progressive implementation of iPSC-based patient-derived models will improve drug development by addressing drug efficacy, specificity, and toxicity in a complex humanized environment. We will review the current state of literature on iPSC-derived model systems in this field, with special emphasis on the access to patient source material and the complications that may come with it. Given the essential role of BMPs during embryonic development and stem cell differentiation, gain- or loss-of-function mutations in the BMP signalling pathway may compromise iPSC generation, maintenance, and differentiation procedures. This review highlights the need for careful optimization of the protocols used. Finally, we will discuss recent developments towards complex in vitro culture models aiming to resemble specific tissue microenvironments with multi-faceted cellular inputs, such as cell mechanics and ECM together with organoids, organ-on-chip, and microfluidic technologies.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), ISPA-HUCA, Avda. de Roma, s/n, 33011 Oviedo, Spain
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Christian Hiepen
- Department of Engineering and Natural Sciences, Westphalian University of Applied Sciences, August-Schmidt-Ring 10, 45665 Recklinghausen, Germany
| |
Collapse
|
10
|
Dragoni S, Turowski P. Vascular Signalling. Cells 2023; 12:2038. [PMID: 37626847 PMCID: PMC10453014 DOI: 10.3390/cells12162038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
In all vertebrates, closed blood and open lymph circulatory systems are essential for the delivery of nutrients and oxygen to tissues, waste clearance, and immune function [...].
Collapse
Affiliation(s)
- Silvia Dragoni
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Patric Turowski
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
11
|
Baccouche B, Lietuvninkas L, Kazlauskas A. Activin A Limits VEGF-Induced Permeability via VE-PTP. Int J Mol Sci 2023; 24:8698. [PMID: 37240047 PMCID: PMC10218593 DOI: 10.3390/ijms24108698] [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: 04/19/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical success of neutralizing vascular endothelial growth factor (VEGF) has unequivocally identified VEGF as a driver of retinal edema that underlies a variety of blinding conditions. VEGF is not the only input that is received and integrated by the endothelium. For instance, the permeability of blood vessels is also regulated by the large and ubiquitously expressed transforming growth factor beta (TGF-β) family. In this project, we tested the hypothesis that members of the TGF-β family influence the VEGF-mediated control of the endothelial cell barrier. To this end, we compared the effect of bone morphogenetic protein-9 (BMP-9), TGF-β1, and activin A on the VEGF-driven permeability of primary human retinal endothelial cells. While BMP-9 and TGF-β1 had no effect on VEGF-induced permeability, activin A limited the extent to which VEGF relaxed the barrier. This activin A effect was associated with the reduced activation of VEGFR2 and its downstream effectors and an increased expression of vascular endothelial tyrosine phosphatase (VE-PTP). Attenuating the expression or activity of VE-PTP overcame the effect of activin A. Taken together, these observations indicate that the TGF-β superfamily governed VEGF-mediated responsiveness in a ligand-specific manner. Furthermore, activin A suppressed the responsiveness of cells to VEGF, and the underlying mechanism involved the VE-PTP-mediated dephosphorylation of VEGFR2.
Collapse
Affiliation(s)
- Basma Baccouche
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lina Lietuvninkas
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Andrius Kazlauskas
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Piekarska K, Bonowicz K, Grzanka A, Jaworski ŁM, Reiter RJ, Slominski AT, Steinbrink K, Kleszczyński K, Gagat M. Melatonin and TGF-β-Mediated Release of Extracellular Vesicles. Metabolites 2023; 13:metabo13040575. [PMID: 37110233 PMCID: PMC10142249 DOI: 10.3390/metabo13040575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The immune system, unlike other systems, must be flexible and able to "adapt" to fully cope with lurking dangers. The transition from intracorporeal balance to homeostasis disruption is associated with activation of inflammatory signaling pathways, which causes modulation of the immunology response. Chemotactic cytokines, signaling molecules, and extracellular vesicles act as critical mediators of inflammation and participate in intercellular communication, conditioning the immune system's proper response. Among the well-known cytokines allowing for the development and proper functioning of the immune system by mediating cell survival and cell-death-inducing signaling, the tumor necrosis factor α (TNF-α) and transforming growth factor β (TGF-β) are noteworthy. The high bloodstream concentration of those pleiotropic cytokines can be characterized by anti- and pro-inflammatory activity, considering the powerful anti-inflammatory and anti-oxidative stress capabilities of TGF-β known from the literature. Together with the chemokines, the immune system response is also influenced by biologically active chemicals, such as melatonin. The enhanced cellular communication shows the relationship between the TGF-β signaling pathway and the extracellular vesicles (EVs) secreted under the influence of melatonin. This review outlines the findings on melatonin activity on TGF-β-dependent inflammatory response regulation in cell-to-cell communication leading to secretion of the different EV populations.
Collapse
Affiliation(s)
- Klaudia Piekarska
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Klaudia Bonowicz
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Alina Grzanka
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Łukasz M Jaworski
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA
| | - Andrzej T Slominski
- Department of Dermatology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Pathology and Laboratory Medicine Service, VA Medical Center, Birmingham, AL 35294, USA
| | - Kerstin Steinbrink
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Konrad Kleszczyński
- Department of Dermatology, University of Münster, Von-Esmarch-Str. 58, 48149 Münster, Germany
| | - Maciej Gagat
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| |
Collapse
|
13
|
Tuning between Nuclear Organization and Functionality in Health and Disease. Cells 2023; 12:cells12050706. [PMID: 36899842 PMCID: PMC10000962 DOI: 10.3390/cells12050706] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/08/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
The organization of eukaryotic genome in the nucleus, a double-membraned organelle separated from the cytoplasm, is highly complex and dynamic. The functional architecture of the nucleus is confined by the layers of internal and cytoplasmic elements, including chromatin organization, nuclear envelope associated proteome and transport, nuclear-cytoskeletal contacts, and the mechano-regulatory signaling cascades. The size and morphology of the nucleus could impose a significant impact on nuclear mechanics, chromatin organization, gene expression, cell functionality and disease development. The maintenance of nuclear organization during genetic or physical perturbation is crucial for the viability and lifespan of the cell. Abnormal nuclear envelope morphologies, such as invagination and blebbing, have functional implications in several human disorders, including cancer, accelerated aging, thyroid disorders, and different types of neuro-muscular diseases. Despite the evident interplay between nuclear structure and nuclear function, our knowledge about the underlying molecular mechanisms for regulation of nuclear morphology and cell functionality during health and illness is rather poor. This review highlights the essential nuclear, cellular, and extracellular components that govern the organization of nuclei and functional consequences associated with nuclear morphometric aberrations. Finally, we discuss the recent developments with diagnostic and therapeutic implications targeting nuclear morphology in health and disease.
Collapse
|
14
|
Han B, Cao C, Wang A, Zhao Y, Jin M, Wang Y, Chen S, Yu M, Yang Z, Qu X, Wang X. Injectable Double-Network Hydrogel-Based Three-Dimensional Cell Culture Systems for Regenerating Dental Pulp. ACS APPLIED MATERIALS & INTERFACES 2023; 15:7821-7832. [PMID: 36734883 DOI: 10.1021/acsami.2c20848] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The regeneration of dental pulp tissue is very important, but difficult, in dentistry. The biocompatibility, water content, and viscoelastic properties of pulp-like tissue must be optimized to achieve the efficient transfer of metabolites and nutrients, a suitable degradation rate, distribution of encapsulated cells, injectability, and gelation in situ under physiological conditions. As promising materials for pulp regeneration, hydrogel scaffolds have been produced to simulate the extracellular matrix and transmit signaling molecules. It is imperative to develop hydrogels to effectively regenerate pulp tissue for clinical application. Here, two injectable double-network (DN) hydrogel-based three-dimensional (3D) cell culture systems were developed for regenerating dental pulp. The microstructure, mechanical property, rheology property, and degradation behavior of the injectable DN glycol chitosan-based hydrogels in a simulated root canal model were characterized and compared to a single-network (SN) glycol chitosan-based hydrogel. Human dental pulp stem cells (hDPSCs) were then encapsulated into the GC-based hydrogels for the regeneration of pulp tissue, and the biological performance was investigated both in vitro and in vivo. The results showed that the DN hydrogels had ideal injectability under physiological conditions due to the dynamic nature of the crosslinks. Besides, the DN hydrogels exhibited better mechanical properties and longer degradation duration than the corresponding SN hydrogel. As a 3D cell culture system, the characteristics of the DN hydrogel facilitated odontogenic differentiation and mineralization of hDPSCs in vitro. Further in vivo analysis confirmed that the chemical composition, matrix stiffness, and degradation rate of the DN hydrogel matched those of pulp-like fibrous connective tissue, which might be related to Smad3 activation. These findings demonstrate that DN glycol chitosan-based hydrogels are suitable for the regeneration of pulp tissue.
Collapse
Affiliation(s)
- Bing Han
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Chunling Cao
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Aijing Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yanran Zhao
- College of Materials Science and Opto-electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Moran Jin
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Yuhan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Shuqin Chen
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Min Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| | - Zhenzhong Yang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaozhong Qu
- College of Materials Science and Opto-electronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing 100081, China
| |
Collapse
|
15
|
Lang A, Benn A, Wolter A, Balcaen T, Collins J, Kerckhofs G, Zwijsen A, Boerckel JD. Endothelial SMAD1/5 signaling couples angiogenesis to osteogenesis during long bone growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.522994. [PMID: 36712097 PMCID: PMC9881901 DOI: 10.1101/2023.01.07.522994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Skeletal development depends on coordinated angiogenesis and osteogenesis. Bone morphogenetic proteins direct bone development by activating SMAD1/5 signaling in osteoblasts. However, the role of SMAD1/5 in skeletal endothelium is unknown. Here, we found that endothelial cell-conditional SMAD1/5 depletion in juvenile mice caused metaphyseal and diaphyseal hypervascularity, resulting in altered cancellous and cortical bone formation. SMAD1/5 depletion induced excessive sprouting, disrupting the columnar structure of the metaphyseal vessels and impaired anastomotic loop morphogenesis at the chondro-osseous junction. Endothelial SMAD1/5 depletion impaired growth plate resorption and, upon long term depletion, abrogated osteoprogenitor recruitment to the primary spongiosa. Finally, in the diaphysis, endothelial SMAD1/5 activity was necessary to maintain the sinusoidal phenotype, with SMAD1/5 depletion inducing formation of large vascular loops, featuring elevated endomucin expression, ectopic tip cell formation, and hyperpermeability. Together, endothelial SMAD1/5 activity sustains skeletal vascular morphogenesis and function and coordinates growth plate remodeling and osteoprogenitor recruitment dynamics during bone growth.
Collapse
Affiliation(s)
- Annemarie Lang
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - Andreas Benn
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, 3000 Leuven, Belgium
| | - Angelique Wolter
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Tim Balcaen
- Biomechanics lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Molecular Design and Synthesis, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Joseph Collins
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Pole of Morphology, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Materials Engineering, KU Leuven, Heverlee, Belgium
- Prometheus, Division for Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - An Zwijsen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Belgium
| | - Joel D. Boerckel
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
16
|
Heydarian M, Oak P, Zhang X, Kamgari N, Kindt A, Koschlig M, Pritzke T, Gonzalez-Rodriguez E, Förster K, Morty RE, Häfner F, Hübener C, Flemmer AW, Yildirim AO, Sudheendra D, Tian X, Petrera A, Kirsten H, Ahnert P, Morrell N, Desai TJ, Sucre J, Spiekerkoetter E, Hilgendorff A. Relationship between impaired BMP signalling and clinical risk factors at early-stage vascular injury in the preterm infant. Thorax 2022; 77:1176-1186. [PMID: 35580897 PMCID: PMC9685723 DOI: 10.1136/thoraxjnl-2021-218083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 04/11/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic lung disease, that is, bronchopulmonary dysplasia (BPD) is the most common complication in preterm infants and develops as a consequence of the misguided formation of the gas-exchange area undergoing prenatal and postnatal injury. Subsequent vascular disease and its progression into pulmonary arterial hypertension critically determines long-term outcome in the BPD infant but lacks identification of early, disease-defining changes. METHODS We link impaired bone morphogenetic protein (BMP) signalling to the earliest onset of vascular pathology in the human preterm lung and delineate the specific effects of the most prevalent prenatal and postnatal clinical risk factors for lung injury mimicking clinically relevant conditions in a multilayered animal model using wild-type and transgenic neonatal mice. RESULTS We demonstrate (1) the significant reduction in BMP receptor 2 (BMPR2) expression at the onset of vascular pathology in the lung of preterm infants, later mirrored by reduced plasma BMP protein levels in infants with developing BPD, (2) the rapid impairment (and persistent change) of BMPR2 signalling on postnatal exposure to hyperoxia and mechanical ventilation, aggravated by prenatal cigarette smoke in a preclinical mouse model and (3) a link to defective alveolar septation and matrix remodelling through platelet derived growth factor-receptor alpha deficiency. In a treatment approach, we partially reversed vascular pathology by BMPR2-targeted treatment with FK506 in vitro and in vivo. CONCLUSION We identified impaired BMP signalling as a hallmark of early vascular disease in the injured neonatal lung while outlining its promising potential as a future biomarker or therapeutic target in this growing, high-risk patient population.
Collapse
Affiliation(s)
- Motaharehsadat Heydarian
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Prajakta Oak
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Xin Zhang
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Nona Kamgari
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alida Kindt
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Markus Koschlig
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Tina Pritzke
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Erika Gonzalez-Rodriguez
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kai Förster
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Rory E Morty
- Department of Translational Pulmonology, University Hospital Heidelberg, Translational Lung Research Center campus of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Friederike Häfner
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christoph Hübener
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Andreas W Flemmer
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Ali Oender Yildirim
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Deepti Sudheendra
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Xuefei Tian
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Nick Morrell
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Tushar J Desai
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Jennifer Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee, USA
| | - Edda Spiekerkoetter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Anne Hilgendorff
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Center for Comprehensive Developmental Care (CDeCLMU), Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| |
Collapse
|
17
|
Greiffer L, Liebau E, Herrmann FC, Spiegler V. Condensed tannins act as anthelmintics by increasing the rigidity of the nematode cuticle. Sci Rep 2022; 12:18850. [PMID: 36344622 PMCID: PMC9640668 DOI: 10.1038/s41598-022-23566-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Tannins and tanniferous plant extracts have been discussed as sustainable means for helminth control in the past two decades in response to a dramatic increase of resistances towards standard anthelmintics. While their bioactivities have been broadly investigated in vitro and in vivo, less is known about their mode of action in nematodes, apart from their protein binding properties. In the current study we therefore investigated the impact of a phytochemically well characterized plant extract from Combretum mucronatum, known to contain procyanidins as the active compounds, on the model organism Caenorhabditis elegans. By different microscopic techniques, the cuticle was identified as the main binding site for tannins, whereas underlying tissues did not seem to be affected. In addition to disruptions of the cuticle structure, molting defects occurred at all larval stages. Finally, an increased rigidity of the nematodes' cuticle due to binding of tannins was confirmed by force spectroscopic measurements. This could be a key finding to explain several anthelmintic activities reported for tannins, especially impairment of molting or exsheathment as well as locomotion.
Collapse
Affiliation(s)
- Luise Greiffer
- Institute for Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Eva Liebau
- Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Fabian C Herrmann
- Institute for Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Verena Spiegler
- Institute for Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany.
| |
Collapse
|
18
|
Bautch VL, Mukouyama YS. The Beauty and Complexity of Blood Vessel Patterning. Cold Spring Harb Perspect Med 2022; 12:a041167. [PMID: 35379659 PMCID: PMC9619359 DOI: 10.1101/cshperspect.a041167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
This review highlights new concepts in vascular patterning in the last 10 years, with emphasis on its beauty and complexity. Endothelial cell signaling pathways that respond to molecular or mechanical signals are described, and examples of vascular patterning that use these pathways in brain, skin, heart, and kidney are highlighted. The pathological consequences of patterning loss are discussed in the context of arteriovenous malformations (AVMs), and prospects for the next 10 years presented.
Collapse
Affiliation(s)
- Victoria L Bautch
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
19
|
Wang C, Xing Y, Zhang J, He M, Dong J, Chen S, Wu H, Huang HY, Chou CH, Bai L, He F, She J, Su A, Wang Y, Thistlethwaite PA, Huang HD, Yuan JXJ, Yuan ZY, Shyy JYJ. MED1 Regulates BMP/TGF-β in Endothelium: Implication for Pulmonary Hypertension. Circ Res 2022; 131:828-841. [PMID: 36252121 DOI: 10.1161/circresaha.122.321532] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Dysregulated BMP (bone morphogenetic protein) or TGF-β (transforming growth factor beta) signaling pathways are imperative in idiopathic and familial pulmonary arterial hypertension (PAH) as well as experimental pulmonary hypertension (PH) in rodent models. MED1 (mediator complex subunit 1) is a key transcriptional co-activator and KLF4 (Krüppel-like factor 4) is a master transcription factor in endothelium. However, MED1 and KLF4 epigenetic and transcriptional regulations of the BMP/TGF-β axes in pulmonary endothelium and their dysregulations leading to PAH remain elusive. We investigate the MED1/KLF4 co-regulation of the BMP/TGF-β axes in endothelium by studying the epigenetic regulation of BMPR2 (BMP receptor type II), ETS-related gene (ERG), and TGFBR2 (TGF-β receptor 2) and their involvement in the PH. METHODS High-throughput screening involving data from RNA-seq, MED1 ChIP-seq, H3K27ac ChIP-seq, ATAC-seq, and high-throughput chromosome conformation capture together with in silico computations were used to explore the epigenetic and transcriptional regulation of BMPR2, ERG, and TGFBR2 by MED1 and KLF4. In vitro experiments with cultured pulmonary arterial endothelial cells (ECs) and bulk assays were used to validate results from these in silico analyses. Lung tissue from patients with idiopathic PAH, animals with experimental PH, and mice with endothelial ablation of MED1 (EC-MED1-/-) were used to study the PH-protective effect of MED1. RESULTS Levels of MED1 were decreased in lung tissue or pulmonary arterial endothelial cells from idiopathic PAH patients and rodent PH models. Mechanistically, MED1 acted synergistically with KLF4 to transactivate BMPR2, ERG, and TGFBR2 via chromatin remodeling and enhancer-promoter interactions. EC-MED1-/- mice showed PH susceptibility. In contrast, MED1 overexpression mitigated the PH phenotype in rodents. CONCLUSIONS A homeostatic regulation of BMPR2, ERG, and TGFBR2 in ECs by MED1 synergistic with KLF4 is essential for the normal function of the pulmonary endothelium. Dysregulation of MED1 and the resulting impairment of the BMP/TGF-β signaling is implicated in the disease progression of PAH in humans and PH in rodent models.
Collapse
Affiliation(s)
- Chen Wang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.).,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Yuanming Xing
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.).,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Jiao Zhang
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.).,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.).,Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| | - Ming He
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| | - Jianjie Dong
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.).,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.).,Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| | - Shanshan Chen
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Haoyu Wu
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - Hsi-Yuan Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.).,School of Life and Health Sciences, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.)
| | - Chih-Hung Chou
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan (C.-H.C.)
| | - Liang Bai
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - Fangzhou He
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Jianqing She
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - Ailing Su
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, China (C.W., Y.X., J.Z., J.D., S.C., L.B., F.H., A.S.)
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China (Y.W.)
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, CA (P.A.T.)
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.).,School of Life and Health Sciences, The Chinese University of Hong Kong-Shenzhen, Shenzhen, China (H.-Y.H., H.-D.H.)
| | - Jason X-J Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA (J.X.-J.Y.)
| | - Zu-Yi Yuan
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, China (C.W., Y.X., J.Z., J.D., H.W., L.B., J.S., Z.-Y.)
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA (J.Z., M.H., J.D., J.Y.-J.)
| |
Collapse
|
20
|
Mendez PL, Obendorf L, Jatzlau J, Burdzinski W, Reichenbach M, Nageswaran V, Haghikia A, Stangl V, Hiepen C, Knaus P. Atheroprone fluid shear stress-regulated ALK1-Endoglin-SMAD signaling originates from early endosomes. BMC Biol 2022; 20:210. [PMID: 36171573 PMCID: PMC9520843 DOI: 10.1186/s12915-022-01396-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fluid shear stress enhances endothelial SMAD1/5 signaling via the BMP9-bound ALK1 receptor complex supported by the co-receptor Endoglin. While moderate SMAD1/5 activation is required to maintain endothelial quiescence, excessive SMAD1/5 signaling promotes endothelial dysfunction. Increased BMP signaling participates in endothelial-to-mesenchymal transition and inflammation culminating in vascular diseases such as atherosclerosis. While the function of Endoglin has so far been described under picomolar concentrations of BMP9 and short-term shear application, we investigated Endoglin under physiological BMP9 and long-term pathophysiological shear conditions. RESULTS We report here that knock-down of Endoglin leads to exacerbated SMAD1/5 phosphorylation and atheroprone gene expression profile in HUVECs sheared for 24 h. Making use of the ligand-trap ALK1-Fc, we furthermore show that this increase is dependent on BMP9/10. Mechanistically, we reveal that long-term exposure of ECs to low laminar shear stress leads to enhanced Endoglin expression and endocytosis of Endoglin in Caveolin-1-positive early endosomes. In these endosomes, we could localize the ALK1-Endoglin complex, labeled BMP9 as well as SMAD1, highlighting Caveolin-1 vesicles as a SMAD signaling compartment in cells exposed to low atheroprone laminar shear stress. CONCLUSIONS We identified Endoglin to be essential in preventing excessive activation of SMAD1/5 under physiological flow conditions and Caveolin-1-positive early endosomes as a new flow-regulated signaling compartment for BMP9-ALK1-Endoglin signaling axis in atheroprone flow conditions.
Collapse
Affiliation(s)
- Paul-Lennard Mendez
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- International Max-Planck Research School for Biology and Computation, Berlin, Germany
| | - Leon Obendorf
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Wiktor Burdzinski
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin School for Regenerative Therapies, Berlin, Germany
| | - Maria Reichenbach
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Vanasa Nageswaran
- Charité-Universitätsmedizin Berlin, Klinik für Kardiologie, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Arash Haghikia
- Charité-Universitätsmedizin Berlin, Klinik für Kardiologie, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany
| | - Verena Stangl
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Faculty of Engineering and Natural Sciences, Westphalian University of Applied Sciences, Recklinghausen, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany.
- International Max-Planck Research School for Biology and Computation, Berlin, Germany.
- Berlin School for Regenerative Therapies, Berlin, Germany.
| |
Collapse
|
21
|
Integrin-Linked Kinase Expression in Human Valve Endothelial Cells Plays a Protective Role in Calcific Aortic Valve Disease. Antioxidants (Basel) 2022; 11:antiox11091736. [PMID: 36139812 PMCID: PMC9495882 DOI: 10.3390/antiox11091736] [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: 07/31/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is highly prevalent during aging. CAVD initiates with endothelial dysfunction, leading to lipid accumulation, inflammation, and osteogenic transformation. Integrin-linked kinase (ILK) participates in the progression of cardiovascular diseases, such as endothelial dysfunction and atherosclerosis. However, ILK role in CAVD is unknown. First, we determined that ILK expression is downregulated in aortic valves from patients with CAVD compared to non-CAVD, especially at the valve endothelium, and negatively correlated with calcification markers. Silencing ILK expression in human valve endothelial cells (siILK-hVECs) induced endothelial-to-mesenchymal transition (EndMT) and promoted a switch to an osteoblastic phenotype; SiILK-hVECs expressed increased RUNX2 and developed calcified nodules. siILK-hVECs exhibited decreased NO production and increased nitrosative stress, suggesting valvular endothelial dysfunction. NO treatment of siILK-hVECs prevented VEC transdifferentiation, while treatment with an eNOS inhibitor mimicked ILK-silencing induction of EndMT. Accordingly, NO treatment inhibited VEC calcification. Mechanistically, siILK-hVECs showed increased Smad2 phosphorylation, suggesting a TGF-β-dependent mechanism, and NO treatment decreased Smad2 activation and RUNX2. Experiments performed in eNOS KO mice confirmed the involvement of the ILK-eNOS signaling pathway in valve calcification, since aortic valves from these animals showed decreased ILK expression, increased RUNX2, and calcification. Our study demonstrated that ILK endothelial expression participates in human CAVD development by preventing endothelial osteogenic transformation.
Collapse
|
22
|
Närvä E, Taskinen ME, Lilla S, Isomursu A, Pietilä M, Weltner J, Isola J, Sihto H, Joensuu H, Zanivan S, Norman J, Ivaska J. MASTL is enriched in cancerous and pluripotent stem cells and influences OCT1/OCT4 levels. iScience 2022; 25:104459. [PMID: 35677646 PMCID: PMC9167974 DOI: 10.1016/j.isci.2022.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/04/2022] [Accepted: 05/13/2022] [Indexed: 11/01/2022] Open
Abstract
MASTL is a mitotic accelerator with an emerging role in breast cancer progression. However, the mechanisms behind its oncogenicity remain largely unknown. Here, we identify a previously unknown role and eminent expression of MASTL in stem cells. MASTL staining from a large breast cancer patient cohort indicated a significant association with β3 integrin, an established mediator of breast cancer stemness. MASTL silencing reduced OCT4 levels in human pluripotent stem cells and OCT1 in breast cancer cells. Analysis of the cell-surface proteome indicated a strong link between MASTL and the regulation of TGF-β receptor II (TGFBR2), a key modulator of TGF-β signaling. Overexpression of wild-type and kinase-dead MASTL in normal mammary epithelial cells elevated TGFBR2 levels. Conversely, MASTL depletion in breast cancer cells attenuated TGFBR2 levels and downstream signaling through SMAD3 and AKT pathways. Taken together, these results indicate that MASTL supports stemness regulators in pluripotent and cancerous stem cells.
Collapse
Affiliation(s)
- Elisa Närvä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Maria E. Taskinen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | | | - Aleksi Isomursu
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Mika Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Jere Weltner
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Jorma Isola
- Laboratory of Cancer Biology, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland
| | - Harri Sihto
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | - Heikki Joensuu
- University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Sara Zanivan
- CRUK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Jim Norman
- CRUK Beatson Institute, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
- Department of Life Technologies, University of Turku, 20520 Turku, Finland
- Western Finnish Cancer Center (FICAN West), University of Turku, 20520 Turku, Finland
- Foundation for the Finnish Cancer Institute, Tukholmankatu 8, Helsinki, Finland
| |
Collapse
|
23
|
Joshi SR, Liu J, Bloom T, Karaca Atabay E, Kuo TH, Lee M, Belcheva E, Spaits M, Grenha R, Maguire MC, Frost JL, Wang K, Briscoe SD, Alexander MJ, Herrin BR, Castonguay R, Pearsall RS, Andre P, Yu PB, Kumar R, Li G. Sotatercept analog suppresses inflammation to reverse experimental pulmonary arterial hypertension. Sci Rep 2022; 12:7803. [PMID: 35551212 PMCID: PMC9098455 DOI: 10.1038/s41598-022-11435-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/14/2022] [Indexed: 11/22/2022] Open
Abstract
Sotatercept is an activin receptor type IIA-Fc (ActRIIA-Fc) fusion protein that improves cardiopulmonary function in patients with pulmonary arterial hypertension (PAH) by selectively trapping activins and growth differentiation factors. However, the cellular and molecular mechanisms of ActRIIA-Fc action are incompletely understood. Here, we determined through genome-wide expression profiling that inflammatory and immune responses are prominently upregulated in the lungs of a Sugen-hypoxia rat model of severe angio-obliterative PAH, concordant with profiles observed in PAH patients. Therapeutic treatment with ActRIIA-Fc-but not with a vasodilator-strikingly reversed proinflammatory and proliferative gene expression profiles and normalized macrophage infiltration in diseased rodent lungs. Furthermore, ActRIIA-Fc normalized pulmonary macrophage infiltration and corrected cardiopulmonary structure and function in Bmpr2 haploinsufficient mice subjected to hypoxia, a model of heritable PAH. Three high-affinity ligands of ActRIIA-Fc each induced macrophage activation in vitro, and their combined immunoneutralization in PAH rats produced cardiopulmonary benefits comparable to those elicited by ActRIIA-Fc. Our results in complementary experimental and genetic models of PAH reveal therapeutic anti-inflammatory activities of ActRIIA-Fc that, together with its known anti-proliferative effects on vascular cell types, could underlie clinical activity of sotatercept as either monotherapy or add-on to current PAH therapies.
Collapse
Affiliation(s)
- Sachindra R Joshi
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jun Liu
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Troy Bloom
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
- Ultivue, Cambridge, MA, USA
| | - Elif Karaca Atabay
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Tzu-Hsing Kuo
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Michael Lee
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Elitza Belcheva
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Matthew Spaits
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Rosa Grenha
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Michelle C Maguire
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jeffrey L Frost
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Kathryn Wang
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Steven D Briscoe
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Mark J Alexander
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Brantley R Herrin
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Roselyne Castonguay
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - R Scott Pearsall
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
- Cellarity, Cambridge, MA, USA
| | - Patrick Andre
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ravindra Kumar
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA
| | - Gang Li
- Discovery Group, Acceleron Pharma Inc., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
24
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
25
|
Andre P, Joshi SR, Briscoe SD, Alexander MJ, Li G, Kumar R. Therapeutic Approaches for Treating Pulmonary Arterial Hypertension by Correcting Imbalanced TGF-β Superfamily Signaling. Front Med (Lausanne) 2022; 8:814222. [PMID: 35141256 PMCID: PMC8818880 DOI: 10.3389/fmed.2021.814222] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterized by high blood pressure in the pulmonary circulation driven by pathological remodeling of distal pulmonary arteries, leading typically to death by right ventricular failure. Available treatments improve physical activity and slow disease progression, but they act primarily as vasodilators and have limited effects on the biological cause of the disease—the uncontrolled proliferation of vascular endothelial and smooth muscle cells. Imbalanced signaling by the transforming growth factor-β (TGF-β) superfamily contributes extensively to dysregulated vascular cell proliferation in PAH, with overactive pro-proliferative SMAD2/3 signaling occurring alongside deficient anti-proliferative SMAD1/5/8 signaling. We review the TGF-β superfamily mechanisms underlying PAH pathogenesis, superfamily interactions with inflammation and mechanobiological forces, and therapeutic strategies under development that aim to restore SMAD signaling balance in the diseased pulmonary arterial vessels. These strategies could potentially reverse pulmonary arterial remodeling in PAH by targeting causative mechanisms and therefore hold significant promise for the PAH patient population.
Collapse
|
26
|
Goodman MB, Savage-Dunn C. Reciprocal interactions between transforming growth factor beta signaling and collagens: Insights from Caenorhabditis elegans. Dev Dyn 2022; 251:47-60. [PMID: 34537996 PMCID: PMC8982858 DOI: 10.1002/dvdy.423] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023] Open
Abstract
Studies in genetically tractable organisms such as the nematode Caenorhabditis elegans have led to pioneering insights into conserved developmental regulatory mechanisms. For example, Smad signal transducers for the transforming growth factor beta (TGF-β) superfamily were first identified in C. elegans and in the fruit fly Drosophila. Recent studies of TGF-β signaling and the extracellular matrix (ECM) in C. elegans have forged unexpected links between signaling and the ECM, yielding novel insights into the reciprocal interactions that occur across tissues and spatial scales, and potentially providing new opportunities for the study of biomechanical regulation of gene expression.
Collapse
Affiliation(s)
- Miriam B. Goodman
- Department of Molecular and Cellular Physiology, Stanford University, CA 94304
| | - Cathy Savage-Dunn
- Department of Biology, Queens College at the City University of New York, 11367,Correspondence to: >
| |
Collapse
|
27
|
Stuelten CH, Zhang YE. Transforming Growth Factor-β: An Agent of Change in the Tumor Microenvironment. Front Cell Dev Biol 2021; 9:764727. [PMID: 34712672 PMCID: PMC8545984 DOI: 10.3389/fcell.2021.764727] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Transforming Growth Factor-β (TGF-β) is a key regulator of embryonic development, adult tissue homeostasis, and lesion repair. In tumors, TGF-β is a potent inhibitor of early stage tumorigenesis and promotes late stage tumor progression and metastasis. Here, we review the roles of TGF-β as well as components of its signaling pathways in tumorigenesis. We will discuss how a core property of TGF-β, namely its ability to change cell differentiation, leads to the transition of epithelial cells, endothelial cells and fibroblasts to a myofibroblastoid phenotype, changes differentiation and polarization of immune cells, and induces metabolic reprogramming of cells, all of which contribute to the progression of epithelial tumors.
Collapse
Affiliation(s)
- Christina H. Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Ying E. Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
28
|
Huang Q, Gan Y, Yu Z, Wu H, Zhong Z. Endothelial to Mesenchymal Transition: An Insight in Atherosclerosis. Front Cardiovasc Med 2021; 8:734550. [PMID: 34604359 PMCID: PMC8484517 DOI: 10.3389/fcvm.2021.734550] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis is a fundamental disease of the cardiovascular system that leads to high morbidity and mortality worldwide. The endothelium is the first protective barrier in atherosclerosis. Endothelial cells have the potential to be transformed into mesenchymal cells, in a process termed endothelial to mesenchymal transition (EndMT). On the one hand, EndMT is known to contribute to atherosclerosis by inducing a number of phenotypes ranging from endothelial cell dysfunction to plaque formation. On the other hand, risk factors for atherosclerosis can lead to EndMT. A substantial body of evidence has suggested that EndMT induces the development of atherosclerosis; therefore, a deeper understanding of the molecular mechanisms underlying EndMT in atherosclerosis might provide insights to reverse this condition.
Collapse
Affiliation(s)
- Qingyan Huang
- Center for Precision Medicine, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Yuhong Gan
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Zhikang Yu
- Center for Precision Medicine, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Heming Wu
- Center for Precision Medicine, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| | - Zhixiong Zhong
- Center for Precision Medicine, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Clinical Molecular Diagnostics and Antibody Therapeutics, Meizhou People's Hospital, Meizhou Academy of Medical Sciences, Meizhou, China
| |
Collapse
|
29
|
Han O, Pak B, Jin SW. The Role of BMP Signaling in Endothelial Heterogeneity. Front Cell Dev Biol 2021; 9:673396. [PMID: 34235147 PMCID: PMC8255612 DOI: 10.3389/fcell.2021.673396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023] Open
Abstract
Bone morphogenetic proteins (BMPs), which compose the largest group of the transforming growth factor-β (TGF-ß) superfamily, have been implied to play a crucial role in diverse physiological processes. The most intriguing feature of BMP signaling is that it elicits heterogeneous responses from cells with equivalent identity, thus permitting highly context-dependent signaling outcomes. In endothelial cells (ECs), which are increasingly perceived as a highly heterogeneous population of cells with respect to their morphology, function, as well as molecular characteristics, BMP signaling has shown to elicit diverse and often opposite effects, illustrating the innate complexity of signaling responses. In this review, we provide a concise yet comprehensive overview of how outcomes of BMP signaling are modulated in a context-dependent manner with an emphasis on the underlying molecular mechanisms and summarize how these regulations of the BMP signaling promote endothelial heterogeneity.
Collapse
Affiliation(s)
- Orjin Han
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Boryeong Pak
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| | - Suk-Won Jin
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, South Korea
| |
Collapse
|
30
|
The BMP Pathway in Blood Vessel and Lymphatic Vessel Biology. Int J Mol Sci 2021; 22:ijms22126364. [PMID: 34198654 PMCID: PMC8232321 DOI: 10.3390/ijms22126364] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) were originally identified as the active components in bone extracts that can induce ectopic bone formation. In recent decades, their key role has broadly expanded beyond bone physiology and pathology. Nowadays, the BMP pathway is considered an important player in vascular signaling. Indeed, mutations in genes encoding different components of the BMP pathway cause various severe vascular diseases. Their signaling contributes to the morphological, functional and molecular heterogeneity among endothelial cells in different vessel types such as arteries, veins, lymphatic vessels and capillaries within different organs. The BMP pathway is a remarkably fine-tuned pathway. As a result, its signaling output in the vessel wall critically depends on the cellular context, which includes flow hemodynamics, interplay with other vascular signaling cascades and the interaction of endothelial cells with peri-endothelial cells and the surrounding matrix. In this review, the emerging role of BMP signaling in lymphatic vessel biology will be highlighted within the framework of BMP signaling in the circulatory vasculature.
Collapse
|
31
|
Rödel CJ, Abdelilah-Seyfried S. A zebrafish toolbox for biomechanical signaling in cardiovascular development and disease. Curr Opin Hematol 2021; 28:198-207. [PMID: 33714969 DOI: 10.1097/moh.0000000000000648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW The zebrafish embryo has emerged as a powerful model organism to investigate the mechanisms by which biophysical forces regulate vascular and cardiac cell biology during development and disease. A versatile arsenal of methods and tools is available to manipulate and analyze biomechanical signaling. This review aims to provide an overview of the experimental strategies and tools that have been utilized to study biomechanical signaling in cardiovascular developmental processes and different vascular disease models in the zebrafish embryo. Within the scope of this review, we focus on work published during the last two years. RECENT FINDINGS Genetic and pharmacological tools for the manipulation of cardiac function allow alterations of hemodynamic flow patterns in the zebrafish embryo and various types of transgenic lines are available to report endothelial cell responses to biophysical forces. These tools have not only revealed the impact of biophysical forces on cardiovascular development but also helped to establish more accurate models for cardiovascular diseases including cerebral cavernous malformations, hereditary hemorrhagic telangiectasias, arteriovenous malformations, and lymphangiopathies. SUMMARY The zebrafish embryo is a valuable vertebrate model in which in-vivo manipulations of biophysical forces due to cardiac contractility and blood flow can be performed. These analyses give important insights into biomechanical signaling pathways that control endothelial and endocardial cell behaviors. The technical advances using this vertebrate model will advance our understanding of the impact of biophysical forces in cardiovascular pathologies.
Collapse
Affiliation(s)
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
32
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
33
|
Rybtsova N, Berezina T, Kagansky A, Rybtsov S. Can Blood-Circulating Factors Unveil and Delay Your Biological Aging? Biomedicines 2020; 8:E615. [PMID: 33333870 PMCID: PMC7765271 DOI: 10.3390/biomedicines8120615] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022] Open
Abstract
According to the World Health Organization, the population of over 60 will double in the next 30 years in the developed countries, which will enforce a further raise of the retirement age and increase the burden on the healthcare system. Therefore, there is an acute issue of maintaining health and prolonging active working longevity, as well as implementation of early monitoring and prevention of premature aging and age-related disorders to avoid early disability. Traditional indicators of biological age are not always informative and often require extensive and expensive analysis. The study of blood factors is a simple and easily accessible way to assess individual health and supplement the traditional indicators of a person's biological age with new objective criteria. With age, the processes of growth and development, tissue regeneration and repair decline; they are gradually replaced by enhanced catabolism, inflammatory cell activity, and insulin resistance. The number of senescent cells supporting the inflammatory loop rises; cellular clearance by autophagy and mitophagy slows down, resulting in mitochondrial and cellular damage and dysfunction. Monitoring of circulated blood factors not only reflects these processes, but also allows suggesting medical intervention to prevent or decelerate the development of age-related diseases. We review the age-related blood factors discussed in recent publications, as well as approaches to slowing aging for healthy and active longevity.
Collapse
Affiliation(s)
- Natalia Rybtsova
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Tatiana Berezina
- Department of Scientific Basis of Extreme Psychology, Moscow State University of Psychology and Education, 127051 Moscow, Russia;
| | - Alexander Kagansky
- Centre for Genomic and Regenerative Medicine, School of Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Stanislav Rybtsov
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK;
| |
Collapse
|
34
|
Potential Second-Hits in Hereditary Hemorrhagic Telangiectasia. J Clin Med 2020; 9:jcm9113571. [PMID: 33167572 PMCID: PMC7694477 DOI: 10.3390/jcm9113571] [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: 10/04/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant genetic disorder that presents with telangiectases in skin and mucosae, and arteriovenous malformations (AVMs) in internal organs such as lungs, liver, and brain. Mutations in ENG (endoglin), ACVRL1 (ALK1), and MADH4 (Smad4) genes account for over 95% of HHT. Localized telangiectases and AVMs are present in different organs, with frequencies which differ among affected individuals. By itself, HHT gene heterozygosity does not account for the focal nature and varying presentation of the vascular lesions leading to the hypothesis of a “second-hit” that triggers the lesions. Accumulating research has identified a variety of triggers that may synergize with HHT gene heterozygosity to generate the vascular lesions. Among the postulated second-hits are: mechanical trauma, light, inflammation, vascular injury, angiogenic stimuli, shear stress, modifier genes, and somatic mutations in the wildtype HHT gene allele. The aim of this review is to summarize these triggers, as well as the functional mechanisms involved.
Collapse
|