1
|
Gao Y, Meng X, Zhu H, Zao X, Wu D, Guo Q, Li X, Dong H, Zhang D. Exosomes Derived from the Serum of Mice That Received a Huoxue Yiqi Recipe Promote Angiogenesis Following Myocardial Infarction. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40358553 DOI: 10.1021/acsami.5c02784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Proangiogenic therapy offers a promising strategy for treating and preventing heart failure and cardiac remodeling following a myocardial infarction (MI). Although exosome-based proangiogenic therapy has significant potential in regenerative medicine and MI treatment, its application remains limited by suboptimal therapeutic efficacy. Here, we present exosomes (HXYQR-Exo) derived from the serum of mice treated with the Huoxue Yiqi Recipe (HXYQR) to promote angiogenesis and repair cardiac tissue post-MI, with a systematic elucidation of the underlying mechanisms. Our findings show that HXYQR-Exo incorporates pharmaceutically active components of HXYQR, enhancing the proliferation, invasion, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) under hypoxic conditions. In vivo studies demonstrate significant improvements in cardiac function and angiogenesis. Mechanistic investigations reveal that these effects are mediated through the activation of the HIF-1α/VEGF, Focal Adhesion Kinase (FAK), and p38/Mitogen-Activated Protein Kinase-Activated Protein Kinase (MAPKAPK)/Heat Shock Protein 27 (HSP27) pathways. This study introduces an exosome-based approach for MI treatment and cardiac repair, offering an effective strategy to enhance exosome biological activities and functions via traditional Chinese medicine preconditioning.
Collapse
Affiliation(s)
- Yijie Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
- Beijing Key Laboratory for Bioengineering and Sensing Technology Research Centre for Bioengineering and Sensing Technology School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 10083, P. R. China
- National Center for Integrative Medicine; Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xiangdan Meng
- Beijing Key Laboratory for Bioengineering and Sensing Technology Research Centre for Bioengineering and Sensing Technology School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 10083, P. R. China
| | - Haiyan Zhu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Xiaobin Zao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Dandan Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Qianqian Guo
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| | - Xianlun Li
- National Center for Integrative Medicine; Department of Integrative Medicine Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Haifeng Dong
- Beijing Key Laboratory for Bioengineering and Sensing Technology Research Centre for Bioengineering and Sensing Technology School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 10083, P. R. China
- Marshall Laboratory of Biomedical Engineering, Shenzhen Key Laboratory for Nano-Biosensing Technology, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, P. R. China
| | - Dongmei Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, No. 5 Haiyuncang, Beijing 100700, China
| |
Collapse
|
2
|
Zhang R, Wang Y, Jiang H, Aheniyazi A, Tao J, Li J, Yang Y. Therapeutic Angiogenesis Mediated by Traditional Chinese Medicine: Advances in Cardiovascular Disease Treatment. JOURNAL OF ETHNOPHARMACOLOGY 2025:119871. [PMID: 40345269 DOI: 10.1016/j.jep.2025.119871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/26/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese Medicine (TCM) shows growing potential as an adjunct or alternative therapy for vascular occlusion diseases (e.g., stroke, peripheral artery disease) by promoting therapeutic angiogenesis to restore blood flow in ischemic regions while minimizing side effects. AIMS OF THE STUDY This review examines TCM-mediated angiogenesis mechanisms and therapeutic advances in vascular occlusion management, establishing a theoretical foundation for clinical translation and precision medicine development. MATERIALS AND METHODS We systematically analyzed PubMed articles on TCM-induced angiogenesis in vascular occlusion diseases, focusing on herbal formulations, single herbs, bioactive compounds, and their associated signaling pathways. Search PubMed for studies investigating the role of Chinese herbal medicine (TCM), natural compounds, and herbal medicine in angiogenesis, while excluding research related to cancer, tumor, or oncological contexts. RESULTS TCM formulas, individual herbs, and monomeric compounds enhance endothelial cell proliferation, migration, and tube formation via pathways such as HIF/VEGF, PI3K/AKT, NOTCH, BMP/ALK, and Apelin/APJ, improving ischemic blood flow. CONCLUSION This review highlights angiogenesis as a novel strategy for vascular occlusive diseases and underscores TCM's efficacy through multi-target angiogenic regulation mechanism.However, further research using modern medical technologies is needed to optimize clinical application and advance precision medicine.
Collapse
Affiliation(s)
- Rong Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Yunze Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Haoyan Jiang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Aliyanmu Aheniyazi
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Jin Tao
- Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China.
| | - Yining Yang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830000, China; Department of Cardiology, People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Urumqi, China.
| |
Collapse
|
3
|
Hiepen C, Benamar M, Barrasa-Fano J, Condor M, Ilhan M, Münch J, Hastar N, Kerkhoff Y, Harms GS, Mielke T, Koenig B, Block S, Rocks O, Abdelilah-Seyfried S, Van Oosterwyck H, Knaus P. Endothelial tip-cell position, filopodia formation and biomechanics require BMPR2 expression and signaling. Commun Biol 2025; 8:21. [PMID: 39779836 PMCID: PMC11711618 DOI: 10.1038/s42003-024-07431-8] [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: 01/02/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Blood vessel formation relies on biochemical and mechanical signals, particularly during sprouting angiogenesis when endothelial tip cells (TCs) guide sprouting through filopodia formation. The contribution of BMP receptors in defining tip-cell characteristics is poorly understood. Our study combines genetic, biochemical, and molecular methods together with 3D traction force microscopy, which reveals an essential role of BMPR2 for actin-driven filopodia formation and mechanical properties of endothelial cells (ECs). Targeting of Bmpr2 reduced sprouting angiogenesis in zebrafish and BMPR2-deficient human ECs formed fewer filopodia, affecting cell migration and actomyosin localization. Spheroid assays revealed a reduced sprouting of BMPR2-deficient ECs in fibrin gels. Even more strikingly, in mosaic spheroids, BMPR2-deficient ECs failed to acquire tip-cell positions. Yet, 3D traction force microscopy revealed that these distinct cell behaviors of BMPR2-deficient tip cells cannot be explained by differences in force-induced matrix deformations, even though these cells adopted distinct cone-shaped morphologies. Notably, BMPR2 positively regulates local CDC42 activity at the plasma membrane to promote filopodia formation. Our findings reveal that BMPR2 functions as a nexus integrating biochemical and biomechanical processes crucial for TCs during angiogenesis.
Collapse
Affiliation(s)
- Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany.
- Westphalian University of Applied Sciences, August-Schmidt-Ring 10, 45665, Recklinghausen, Germany.
| | - Mounir Benamar
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Jorge Barrasa-Fano
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
| | - Mar Condor
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
| | - Mustafa Ilhan
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
- Berlin School of Integrative Oncology, Augustenburger Platz 1, D-13353, Berlin, Germany
| | - Juliane Münch
- Universität Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht Strasse 24-25, 14476, Potsdam-Golm, Germany
| | - Nurcan Hastar
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Yannic Kerkhoff
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Gregory S Harms
- Universitätsmedizin, Johannes Gutenberg-Universität Mainz, Cell Biology Unit, Imaging Core Facility and the Research Center for Immune Intervention, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Thorsten Mielke
- Max-Planck-Institute for Molecular Genetics, Microscopy & Cryo-Electron Microscopy, Ihnestraße 63-73, 14195, Berlin, Germany
| | - Benjamin Koenig
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
- Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Stephan Block
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Oliver Rocks
- Charité - Universitätsmedizin Berlin, Systemic Cell Dynamics, Charitéplatz 1, 10117, Berlin, Germany
| | - Salim Abdelilah-Seyfried
- Universität Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht Strasse 24-25, 14476, Potsdam-Golm, Germany
| | - Hans Van Oosterwyck
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
- KU Leuven, Prometheus Division of Skeletal Tissue Engineering, Leuven, Belgium
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany.
| |
Collapse
|
4
|
Hong Y, Wang H, Xie H, Zhong X, Chen X, Yu L, Zhang Y, Zhang J, Wang Q, Tang B, Lu L, Guo D. Qishen Granule protects against myocardial ischemia by promoting angiogenesis through BMP2-Dll4-Notch1 pathway. CHINESE HERBAL MEDICINES 2025; 17:139-147. [PMID: 39949802 PMCID: PMC11814263 DOI: 10.1016/j.chmed.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/12/2023] [Accepted: 12/23/2023] [Indexed: 02/16/2025] Open
Abstract
Objective Therapeutic angiogenesis has become a promising approach for treating ischemic heart disease (IHD). The present study aims to investigate the effects of Qishen Granule (QSG) on angiogenesis in myocardial ischemia (MI) and the potential mechanism. Methods In vivo study was conducted on rat model of myocardial infarction. QSG was performed daily at a dose of 2.352 g/kg for four weeks. Cardiac function was assessed by echocardiogram and pro-angiogenic effects were evaluated by Laser Doppler and CD31 expression. Oxygen-glucose deprivation (OGD) was applied in cultured human umbilical vein endothelial cells (HUVECs). Cell viability, wound healing and tube formation assay were used to test functions of HUVECs. ELISA and Western blots were used to assess protein expressions of bone morphogenetic protein 2-delta-like 4-notch homolog 1 (BMP2-Dll4-Notch1) signaling pathway. Results The results showed that QSG improved heart function, cardiac blood flow and microvessel density in myocardial ischemic rats. In vitro, QSG protected HUVECs by promoting the cell viability and tube formation. QSG upregulated bone morphogenetic protein-2 (BMP2) and downregulated delta-like 4 (Dll4) and notch homolog 1 (Notch1) expressions both in rats and HUVECs. Conclusion QSG protected against MI by promoting angiogenesis through BMP2-Dll4-Notch1 pathway. BMP2 might be a promising therapeutic target for IHD.
Collapse
Affiliation(s)
- Yiqin Hong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hui Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hanyan Xie
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinyi Zhong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lishuang Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yawen Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingmei Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qiyan Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Binghua Tang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Linghui Lu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dongqing Guo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
5
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [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: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
6
|
Pak B, Kim M, Han O, Lee HW, Dubrac A, Choi W, Yang JM, Boyé K, Cho H, Citrin KM, Kim I, Eichmann A, Bautch VL, Jin SW. ACVR1/ALK2-p21 signaling axis modulates proliferation of the venous endothelium in the retinal vasculature. Angiogenesis 2024; 27:765-777. [PMID: 38955953 DOI: 10.1007/s10456-024-09936-6] [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: 03/04/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The proliferation of the endothelium is a highly coordinated process to ensure the emergence, expansion, and homeostasis of the vasculature. While Bone Morphogenetic Protein (BMP) signaling fine-tunes the behaviors of endothelium in health and disease, how BMP signaling influences the proliferation of endothelium and therefore, modulates angiogenesis remains largely unknown. Here, we evaluated the role of Activin A Type I Receptor (ACVR1/ALK2), a key BMP receptor in the endothelium, in modulating the proliferation of endothelial cells. We show that ACVR1/ALK2 is a key modulator for the proliferation of endothelium in the retinal vessels. Loss of endothelial ALK2 leads to a significant reduction in endothelial proliferation and results in fewer branches/endothelial cells in the retinal vessels. Interestingly, venous endothelium appears to be more susceptible to ALK2 deletion. Mechanistically, ACVR1/ALK2 inhibits the expression of CDKN1A/p21, a critical negative regulator of cell cycle progression, in a SMAD1/5-dependent manner, thereby enabling the venous endothelium to undergo active proliferation by suppressing CDKN1A/p21. Taken together, our findings show that BMP signaling mediated by ACVR1/ALK2 provides a critical yet previously underappreciated input to modulate the proliferation of venous endothelium, thereby fine-tuning the context of angiogenesis in health and disease.
Collapse
Affiliation(s)
- Boryeong Pak
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Minjung Kim
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Orjin Han
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Heon-Woo Lee
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Pharmacy, Chosun University, Gwangju, Korea
| | - Alexandre Dubrac
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- CHU Sainte-Justine Research Center, and Department of Pathology and Cellular Biology, Université de Montréal, Montréal, QC, Canada
| | - Woosoung Choi
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Jee Myung Yang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Kevin Boyé
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Heewon Cho
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Kathryn M Citrin
- Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Injune Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Victoria L Bautch
- Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Suk-Won Jin
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea.
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Danes D, Raval P, Singh A, Pillai L, Balakrishnan S. Exposure to a sublethal dose of technical grade flubendiamide hampers angiogenesis in the chicken chorioallantoic membrane. Toxicol Res 2024; 40:627-638. [PMID: 39345746 PMCID: PMC11436694 DOI: 10.1007/s43188-024-00254-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 10/01/2024] Open
Abstract
UNLABELLED Pesticides are commonly employed to enhance agricultural productivity to meet the demands of the expanding global populace. Their harmful impact on non-target organisms is a severe cause of concern, and hence, new, presumably safer variants are developed. Flubendiamide is one such insecticide that targets caterpillars of insect pests. To evaluate its safety, we exposed early chicken embryos to technical-grade flubendiamide. Based on a dose range analysis, a lowest observed effect concentration (LOEC) of 25 µg/50µL (500 ppm) was selected for further experiments. LC-MS/MS analysis confirmed the presence of flubendiamide in the treated embryos. Gross morphology of embryos on days 2, 3 and 4 revealed reduced vascular area in the chorioallantoic membrane (CAM). The CAM vessel analysis showed reduced vascular networks in treated group. Hence, we hypothesized that flubendiamide, at LOEC, alters the expression patterns of the essential signaling molecules involved in angiogenesis, leading to compromised blood vessel development in CAM. An initial in silico study of flubendiamide was conducted with proteins involved in the CAM angiogenesis pathway. The docking scores revealed flubendiamide's direct influence on the functionality of angiogenic proteins. Hence, the expression patterns of key regulators of angiogenesis were studied on days 2, 3 and 4 at the transcript and protein levels. The results revealed a significant reduction in VEGFα, AKT, KDR, PCNA, PI3K, BMP2, BMP6, SHH and WNT7A expression in treated embryos, while expression of CASPASE-3 and RHOB were upregulated. Immunolocalization of Cl. Caspase-3 reaffirmed heightened apoptosis in the CAM of day 2 embryos. The study thus confirms that flubendiamide at a sublethal dose can hamper CAM angiogenesis and reduce the vascular networks in developing chick embryos by targeting the VEGF signaling cascade. This finding points to the teratogenic potential of flubendiamide and prompts throughput screening for safety. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s43188-024-00254-z.
Collapse
Affiliation(s)
- Dhanush Danes
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002 India
| | - Pooja Raval
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002 India
| | - Anjali Singh
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002 India
| | - Lakshmi Pillai
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002 India
| | - Suresh Balakrishnan
- Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002 India
| |
Collapse
|
8
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
9
|
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
|
10
|
Al Tarrass M, Belmudes L, Koça D, Azemard V, Liu H, Al Tabosh T, Ciais D, Desroches-Castan A, Battail C, Couté Y, Bouvard C, Bailly S. Large-scale phosphoproteomics reveals activation of the MAPK/GADD45β/P38 axis and cell cycle inhibition in response to BMP9 and BMP10 stimulation in endothelial cells. Cell Commun Signal 2024; 22:158. [PMID: 38439036 PMCID: PMC10910747 DOI: 10.1186/s12964-024-01486-0] [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: 11/24/2023] [Accepted: 01/11/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND BMP9 and BMP10 are two major regulators of vascular homeostasis. These two ligands bind with high affinity to the endothelial type I kinase receptor ALK1, together with a type II receptor, leading to the direct phosphorylation of the SMAD transcription factors. Apart from this canonical pathway, little is known. Interestingly, mutations in this signaling pathway have been identified in two rare cardiovascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. METHODS To get an overview of the signaling pathways modulated by BMP9 and BMP10 stimulation in endothelial cells, we employed an unbiased phosphoproteomic-based strategy. Identified phosphosites were validated by western blot analysis and regulated targets by RT-qPCR. Cell cycle analysis was analyzed by flow cytometry. RESULTS Large-scale phosphoproteomics revealed that BMP9 and BMP10 treatment induced a very similar phosphoproteomic profile. These BMPs activated a non-canonical transcriptional SMAD-dependent MAPK pathway (MEKK4/P38). We were able to validate this signaling pathway and demonstrated that this activation required the expression of the protein GADD45β. In turn, activated P38 phosphorylated the heat shock protein HSP27 and the endocytosis protein Eps15 (EGF receptor pathway substrate), and regulated the expression of specific genes (E-selectin, hyaluronan synthase 2 and cyclooxygenase 2). This study also highlighted the modulation in phosphorylation of proteins involved in transcriptional regulation (phosphorylation of the endothelial transcription factor ERG) and cell cycle inhibition (CDK4/6 pathway). Accordingly, we found that BMP10 induced a G1 cell cycle arrest and inhibited the mRNA expression of E2F2, cyclinD1 and cyclinA1. CONCLUSIONS Overall, our phosphoproteomic screen identified numerous proteins whose phosphorylation state is impacted by BMP9 and BMP10 treatment, paving the way for a better understanding of the molecular mechanisms regulated by BMP signaling in vascular diseases.
Collapse
Affiliation(s)
- Mohammad Al Tarrass
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
| | - Lucid Belmudes
- Grenoble Alpes University, CEA, INSERM, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Dzenis Koça
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
| | - Valentin Azemard
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
| | - Hequn Liu
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
| | - Tala Al Tabosh
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
| | - Delphine Ciais
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
- Present address: Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | | | - Christophe Battail
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
- Grenoble Alpes University, CEA, INSERM, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Yohann Couté
- Grenoble Alpes University, CEA, INSERM, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Claire Bouvard
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France
| | - Sabine Bailly
- Biosanté Unit U1292, Grenoble Alpes University, CEA, Grenoble, 38000, France.
| |
Collapse
|
11
|
Nan W, He Y, Shen S, Wu M, Wang S, Zhang Y. BMP4 inhibits corneal neovascularization by interfering with tip cells in angiogenesis. Exp Eye Res 2023; 237:109680. [PMID: 37858608 DOI: 10.1016/j.exer.2023.109680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Corneal neovascularization (CNV) can lead to impaired corneal transparency, resulting in vision loss or blindness. The primary pathological mechanism underlying CNV is an imbalance between pro-angiogenic and anti-angiogenic factors, with inflammation playing a crucial role. Notably, a vascular endothelial growth factor(VEGF)-A gradient triggers the selection of single endothelial cells(ECs) into primary tip cells that guide sprouting, while a dynamic balance between tip and stalk cells maintains a specific ratio to promote CNV. Despite the central importance of tip-stalk cell selection and shuffling, the underlying mechanisms remain poorly understood. In this study, we examined the effects of bone morphogenetic protein 4 (BMP4) on VEGF-A-induced lumen formation in human umbilical vein endothelial cells (HUVECs) and CD34-stained tip cell formation. In vivo, BMP4 inhibited CNV caused by corneal sutures. This process was achieved by BMP4 decreasing the protein expression of VEGF-A and VEGFR2 in corneal tissue after corneal suture injury. By observing the ultrastructure of the cornea, BMP4 inhibited the sprouting of tip cells and brought forward the appearance of intussusception. Meanwhile, BMP4 attenuated the inflammatory response by inhibiting neutrophil extracellular traps (NETs)formation through the NADPH oxidase-2(NOX-2)pathway. Our results indicate that BMP4 inhibits the formation of tip cells by reducing the generation of NETs, disrupting the dynamic balance of tip and stalk cells and thereby inhibiting CNV, suggesting that BMP4 may be a potential therapeutic target for CNV.
Collapse
Affiliation(s)
- Weijin Nan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Yuxi He
- Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Sitong Shen
- Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Meiliang Wu
- Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Shurong Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Yan Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China; Corneal Refraction Department, The Second Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
12
|
Gupta S, Sharma A, Petrovski G, Verma RS. Vascular reconstruction of the decellularized biomatrix for whole-organ engineering-a critical perspective and future strategies. Front Bioeng Biotechnol 2023; 11:1221159. [PMID: 38026872 PMCID: PMC10680456 DOI: 10.3389/fbioe.2023.1221159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Whole-organ re-engineering is the most challenging goal yet to be achieved in tissue engineering and regenerative medicine. One essential factor in any transplantable and functional tissue engineering is fabricating a perfusable vascular network with macro- and micro-sized blood vessels. Whole-organ development has become more practical with the use of the decellularized organ biomatrix (DOB) as it provides a native biochemical and structural framework for a particular organ. However, reconstructing vasculature and re-endothelialization in the DOB is a highly challenging task and has not been achieved for constructing a clinically transplantable vascularized organ with an efficient perfusable capability. Here, we critically and articulately emphasized factors that have been studied for the vascular reconstruction in the DOB. Furthermore, we highlighted the factors used for vasculature development studies in general and their application in whole-organ vascular reconstruction. We also analyzed in detail the strategies explored so far for vascular reconstruction and angiogenesis in the DOB for functional and perfusable vasculature development. Finally, we discussed some of the crucial factors that have been largely ignored in the vascular reconstruction of the DOB and the future directions that should be addressed systematically.
Collapse
Affiliation(s)
- Santosh Gupta
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Akriti Sharma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
- Department of Ophthalmology, University of Split School of Medicine and University Hospital Centre, Split, Croatia
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
13
|
Pulkkinen HH, Kivistö-Rahnasto A, Korpela H, Heikkilä M, Järveläinen N, Siimes S, Kilpeläinen L, Laham-Karam N, Ylä-Herttuala S, Laakkonen JP. BMP2 gene transfer induces pericardial effusion and inflammatory response in the ischemic porcine myocardium. Front Cardiovasc Med 2023; 10:1279613. [PMID: 38028463 PMCID: PMC10655027 DOI: 10.3389/fcvm.2023.1279613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Pro-angiogenic gene therapy is being developed to treat coronary artery disease (CAD). We recently showed that bone morphogenetic protein 2 (BMP2) and vascular endothelial growth factor-A synergistically regulate endothelial cell sprouting in vitro. BMP2 was also shown to induce endocardial angiogenesis in neonatal mice post-myocardial infarction. In this study, we investigated the potential of BMP2 gene transfer to improve cardiomyocyte function and neovessel formation in a pig chronic myocardial infarction model. Ischemia was induced in domestic pigs by placing a bottleneck stent in the proximal part of the left anterior descending artery 14 days before gene transfer. Intramyocardial gene transfers with adenovirus vectors (1 × 1012 viral particles/pig) containing either human BMP2 (AdBMP2) or beta-galactosidase (AdLacZ) control gene were performed using a needle injection catheter. BMP2 transgene expression in the myocardium was detected with immunofluorescence staining in the gene transfer area 6 days after AdBMP2 administration. BMP2 gene transfer did not induce angiogenesis or cardiomyocyte proliferation in the ischemic pig myocardium as determined by the quantitations of CD31 or Ki-67 stainings, respectively. Accordingly, no changes in heart contractility were detected in left ventricular ejection fraction and strain measurements. However, BMP2 gene transfer induced pericardial effusion (AdBMP2: 9.41 ± 3.17 mm; AdLacZ: 3.07 ± 1.33 mm) that was measured by echocardiography. Furthermore, an increase in the number of immune cells and CD3+ T cells was found in the BMP2 gene transfer area. No changes were detected in the clinical chemistry analysis of pig serum or histology of the major organs, implicating that the gene transfer did not induce general toxicity, myocardial injury, or off-target effects. Finally, the levels of fibrosis and cardiomyocyte apoptosis detected by Sirius red or caspase 3 stainings, respectively, remained unaltered between the groups. Our results demonstrate that BMP2 gene transfer causes inflammatory changes and pericardial effusion in the adult ischemic myocardium, which thus does not support its therapeutic use in chronic CAD.
Collapse
Affiliation(s)
- H. H. Pulkkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - A. Kivistö-Rahnasto
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - H. Korpela
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M. Heikkilä
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - N. Järveläinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S. Siimes
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - L. Kilpeläinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - N. Laham-Karam
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S. Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Heart Center, Kuopio University Hospital, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - J. P. Laakkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
14
|
Hu J, Xu Z, Liao D, Jiang Y, Pu H, Wu Z, Xu X, Zhao Z, Liu J, Lu X, Liu X, Li B. An H 2 S-BMP6 Dual-Loading System with Regulating Yap/Taz and Jun Pathway for Synergistic Critical Limb Ischemia Salvaging Therapy. Adv Healthc Mater 2023; 12:e2301316. [PMID: 37531238 DOI: 10.1002/adhm.202301316] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/12/2023] [Indexed: 08/04/2023]
Abstract
Critical limb ischemia, the final course of peripheral artery disease, is characterized by an insufficient supply of blood flow and excessive oxidative stress. H2 S molecular therapy possesses huge potential for accelerating revascularization and scavenging intracellular reactive oxygen species (ROS). Moreover, it is found that BMP6 is the most significantly up-expressed secreted protein-related gene in HUVECs treated with GYY4137, a H2 S donor, based on the transcriptome analysis. Herein, a UIO-66-NH2 @GYY4137@BMP6 co-delivery nanoplatform to strengthen the therapeutic effects of limb ischemia is developed. The established UIO-66-NH2 @GYY4137@BMP6 nanoplatform exerts its proangiogenic and anti-oxidation functions by regulating key pathways. The underlying molecular mechanisms of UIO-66-NH2 @GYY4137@BMP6 dual-loading system lie in the upregulation of phosphorylated YAP/TAZ and Jun to promote HUVECs proliferation and downregulation of phosphorylated p53/p21 to scavenge excessive ROS. Meanwhile, laser-doppler perfusion imaging (LDPI), injury severity evaluation, and histological analysis confirm the excellent therapeutic effects of UIO-66-NH2 @GYY4137@BMP6 in vivo. This work may shed light on the treatment of critical limb ischemia by regulating YAP, Jun, and p53 signaling pathways based on gas-protein synergistic therapy.
Collapse
Affiliation(s)
- Jiateng Hu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhijue Xu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Donghui Liao
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, 523700, China
| | - Yihong Jiang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Hongji Pu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xintong Xu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhen Zhao
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Jianqiang Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, 523700, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xiaobing Liu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| |
Collapse
|
15
|
Kulikauskas MR, Oatley M, Yu T, Liu Z, Matsumura L, Kidder E, Ruter D, Bautch VL. Endothelial cell SMAD6 balances Alk1 function to regulate adherens junctions and hepatic vascular development. Development 2023; 150:dev201811. [PMID: 37787089 PMCID: PMC10629679 DOI: 10.1242/dev.201811] [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: 03/24/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
BMP signaling is crucial to blood vessel formation and function, but how pathway components regulate vascular development is not well-understood. Here, we find that inhibitory SMAD6 functions in endothelial cells to negatively regulate ALK1-mediated responses, and it is required to prevent vessel dysmorphogenesis and hemorrhage in the embryonic liver vasculature. Reduced Alk1 gene dosage rescued embryonic hepatic hemorrhage and microvascular capillarization induced by Smad6 deletion in endothelial cells in vivo. At the cellular level, co-depletion of Smad6 and Alk1 rescued the destabilized junctions and impaired barrier function of endothelial cells depleted for SMAD6 alone. Mechanistically, blockade of actomyosin contractility or increased PI3K signaling rescued endothelial junction defects induced by SMAD6 loss. Thus, SMAD6 normally modulates ALK1 function in endothelial cells to regulate PI3K signaling and contractility, and SMAD6 loss increases signaling through ALK1 that disrupts endothelial cell junctions. ALK1 loss-of-function also disrupts vascular development and function, indicating that balanced ALK1 signaling is crucial for proper vascular development and identifying ALK1 as a 'Goldilocks' pathway in vascular biology that requires a certain signaling amplitude, regulated by SMAD6, to function properly.
Collapse
Affiliation(s)
- Molly R. Kulikauskas
- Cell Biology and Physiology Curriculum, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Morgan Oatley
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Tianji Yu
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ziqing Liu
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Lauren Matsumura
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elise Kidder
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Dana Ruter
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Victoria L. Bautch
- Cell Biology and Physiology Curriculum, The University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biology, The University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, The University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
16
|
Tielemans B, Wagenaar A, Belge C, Delcroix M, Quarck R. Pulmonary arterial hypertension drugs can partially restore altered angiogenic capacities in bmpr2-silenced human lung microvascular endothelial cells. Pulm Circ 2023; 13:e12293. [PMID: 37790139 PMCID: PMC10543474 DOI: 10.1002/pul2.12293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/31/2023] [Accepted: 09/17/2023] [Indexed: 10/05/2023] Open
Abstract
Mutations in the bone morphogenetic protein receptor type 2 (bmpr2) gene and signaling pathway impairment are observed in heritable and idiopathic pulmonary arterial hypertension (PAH). In PAH, endothelial dysfunction is currently handled by drugs targeting the endothelin-1 (ET-1), nitric oxide (NO), and prostacyclin (PGI2) pathways. The role of angiogenesis in the disease process and the effect of PAH therapies on dysregulated angiogenesis remain inconclusive. We aim to investigate in vitro whether (i) bmpr2 silencing can impair angiogenic capacity of human lung microvascular endothelial cells (HLMVECs) and (ii) PAH therapies can restore them. The effects of macitentan (ET-1), tadalafil (NO), and selexipag (PGI2), on BMPRII pathway activation, endothelial barrier function, and angiogenesis were investigated in bmpr2-silenced HLMVECs. Stable bmpr2 silencing resulted in impaired migration and tube formation in vitro capacity. Inhibition of ET-1 pathway was able to partially restore tube formation in bmpr2-silenced HLMVECs, whereas none of the therapies was able to restore endothelial barrier function, no deleterious effects were observed. Our findings highlight the potential role of BMPRII signaling pathway in driving pulmonary endothelial cell angiogenesis. In addition, PAH drugs display limited effects on endothelial function when BMPRII is impaired, suggesting that innovative therapeutic strategies targeting BMPRII signaling are needed to better rescue endothelial dysfunction in PAH.
Collapse
Affiliation(s)
- Birger Tielemans
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Biomedical MRI, Department of Imaging and PathologyUniversity of LeuvenLeuvenBelgium
| | - Allard Wagenaar
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA)University of LeuvenLeuvenBelgium
| | - Catharina Belge
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Clinical Department of Respiratory Diseases, University HospitalsUniversity of LeuvenLeuvenBelgium
| | - Marion Delcroix
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Clinical Department of Respiratory Diseases, University HospitalsUniversity of LeuvenLeuvenBelgium
| | - Rozenn Quarck
- Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases & Metabolism (CHROMETA) & Clinical Department of Respiratory Diseases, University HospitalsUniversity of LeuvenLeuvenBelgium
| |
Collapse
|
17
|
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: 0.5] [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
|
18
|
Rao S, Liu M, Iosef C, Knutsen C, Alvira CM. Endothelial-specific loss of IKKβ disrupts pulmonary endothelial angiogenesis and impairs postnatal lung growth. Am J Physiol Lung Cell Mol Physiol 2023; 325:L299-L313. [PMID: 37310763 PMCID: PMC10625829 DOI: 10.1152/ajplung.00034.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023] Open
Abstract
Pulmonary angiogenesis drives alveolarization, but the transcriptional regulators directing pulmonary angiogenesis remain poorly defined. Global, pharmacological inhibition of nuclear factor-kappa B (NF-κB) impairs pulmonary angiogenesis and alveolarization. However, establishing a definitive role for NF-κB in pulmonary vascular development has been hindered by embryonic lethality induced by constitutive deletion of NF-κB family members. We created a mouse model allowing inducible deletion of the NF-κB activator, IKKβ, in endothelial cells (ECs) and assessed the effect on lung structure, endothelial angiogenic function, and the lung transcriptome. Embryonic deletion of IKKβ permitted lung vascular development but resulted in a disorganized vascular plexus, while postnatal deletion significantly decreased radial alveolar counts, vascular density, and proliferation of both endothelial and nonendothelial lung cells. Loss of IKKβ impaired survival, proliferation, migration, and angiogenesis in primary lung ECs in vitro, in association with decreased expression of VEGFR2 and activation of downstream effectors. Loss of endothelial IKKβ in vivo induced broad changes in the lung transcriptome with downregulation of genes related to mitotic cell cycle, extracellular matrix (ECM)-receptor interaction, and vascular development, and the upregulation of genes related to inflammation. Computational deconvolution suggested that loss of endothelial IKKβ decreased general capillary, aerocyte capillary, and alveolar type I cell abundance. Taken together, these data definitively establish an essential role for endogenous endothelial IKKβ signaling during alveolarization. A deeper understanding of the mechanisms directing this developmental, physiological activation of IKKβ in the lung vasculature may provide novel targets for the development of strategies to enhance beneficial proangiogenic signaling in lung development and disease.NEW & NOTEWORTHY This study highlights the cell-specific complexity of nuclear factor kappa B signaling in the developing lung by demonstrating that inducible loss of IKKβ in endothelial cells impairs alveolarization, disrupts EC angiogenic function, and broadly represses genes important for vascular development.
Collapse
Affiliation(s)
- Shailaja Rao
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Min Liu
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristiana Iosef
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Carsten Knutsen
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| |
Collapse
|
19
|
Darwish NHE, Hussein KA, Elmasry K, Ibrahim AS, Humble J, Moustafa M, Awadalla F, Al-Shabrawey M. Bone Morphogenetic Protein-4 Impairs Retinal Endothelial Cell Barrier, a Potential Role in Diabetic Retinopathy. Cells 2023; 12:1279. [PMID: 37174679 PMCID: PMC10177364 DOI: 10.3390/cells12091279] [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: 03/14/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Bone Morphogenetic Protein 4 (BMP4) is a secreted growth factor of the Transforming Growth Factor beta (TGFβ) superfamily. The goal of this study was to test whether BMP4 contributes to the pathogenesis of diabetic retinopathy (DR). Immunofluorescence of BMP4 and the vascular marker isolectin-B4 was conducted on retinal sections of diabetic and non-diabetic human and experimental mice. We used Akita mice as a model for type-1 diabetes. Proteins were extracted from the retina of postmortem human eyes and 6-month diabetic Akita mice and age-matched control. BMP4 levels were measured by Western blot (WB). Human retinal endothelial cells (HRECs) were used as an in vitro model. HRECs were treated with BMP4 (50 ng/mL) for 48 h. The levels of phospho-smad 1/5/9 and phospho-p38 were measured by WB. BMP4-treated and control HRECs were also immunostained with anti-Zo-1. We also used electric cell-substrate impedance sensing (ECIS) to calculate the transcellular electrical resistance (TER) under BMP4 treatment in the presence and absence of noggin (200 ng/mL), LDN193189 (200 nM), LDN212854 (200 nM) or inhibitors of vascular endothelial growth factor receptor 2 (VEGFR2; SU5416, 10 μM), p38 (SB202190, 10 μM), ERK (U0126, 10 μM) and ER stress (Phenylbutyric acid or PBA, 30 μmol/L). The impact of BMP4 on matrix metalloproteinases (MMP2 and MMP9) was also evaluated using specific ELISA kits. Immunofluorescence of human and mouse eyes showed increased BMP4 immunoreactivity, mainly localized in the retinal vessels of diabetic humans and mice compared to the control. Western blots of retinal proteins showed a significant increase in BMP4 expression in diabetic humans and mice compared to the control groups (p < 0.05). HRECs treated with BMP4 showed a marked increase in phospho-smad 1/5/9 (p = 0.039) and phospho-p38 (p = 0.013). Immunofluorescence of Zo-1 showed that BMP4-treated cells exhibited significant barrier disruption. ECIS also showed a marked decrease in TER of HRECs by BMP4 treatment compared to vehicle-treated HRECs (p < 0.001). Noggin, LDN193189, LDN212854, and inhibitors of p38 and VEGFR2 significantly mitigated the effects of BMP4 on the TER of HRECs. Our finding provides important insights regarding the role of BMP4 as a potential player in retinal endothelial cell dysfunction in diabetic retinopathy and could be a novel target to preserve the blood-retinal barrier during diabetes.
Collapse
Affiliation(s)
- Noureldien H. E. Darwish
- Eye Research Center, Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35111, Egypt
| | - Khaled A. Hussein
- Oral and Dental Research Insitute, Department of Oral Medicine and Surgery, National Research Center, Cairo 11553, Egypt
| | - Khaled Elmasry
- Department of Oral Biology and Diagnostic Science, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Mansoura 35111, Egypt
| | - Ahmed S. Ibrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35111, Egypt
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Julia Humble
- Eye Research Center, Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
| | - Mohamed Moustafa
- Eye Research Center, Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
| | - Fatma Awadalla
- Eye Research Center, Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35111, Egypt
| | - Mohamed Al-Shabrawey
- Eye Research Center, Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Mansoura 35111, Egypt
| |
Collapse
|
20
|
Brown M, Zhu S, Taylor L, Tabrizian M, Li-Jessen NY. Unraveling the Relevance of Tissue-Specific Decellularized Extracellular Matrix Hydrogels for Vocal Fold Regenerative Biomaterials: A Comprehensive Proteomic and In Vitro Study. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200095. [PMID: 37547672 PMCID: PMC10398787 DOI: 10.1002/anbr.202200095] [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] [Indexed: 02/04/2023] Open
Abstract
Decellularized extracellular matrix (dECM) is a promising material for tissue engineering applications. Tissue-specific dECM is often seen as a favorable material that recapitulates a native-like microenvironment for cellular remodeling. However, the minute quantity of dECM derivable from small organs like the vocal fold (VF) hampers manufacturing scalability. Small intestinal submucosa (SIS), a commercial product with proven regenerative capacity, may be a viable option for VF applications. This study aims to compare dECM hydrogels derived from SIS or VF tissue with respect to protein content and functionality using mass spectrometry-based proteomics and in vitro studies. Proteomic analysis reveals that VF and SIS dECM share 75% of core matrisome proteins. Although VF dECM proteins have greater overlap with native VF, SIS dECM shows less cross-sample variability. Following decellularization, significant reductions of soluble collagen (61%), elastin (81%), and hyaluronan (44%) are noted in VF dECM. SIS dECM contains comparable elastin and hyaluronan but 67% greater soluble collagen than VF dECM. Cells deposit more neo-collagen on SIS than VF-dECM hydrogels, whereas neo-elastin (~50 μg/scaffold) and neo-hyaluronan (~ 6 μg/scaffold) are comparable between the two hydrogels. Overall, SIS dECM possesses reasonably similar proteomic profile and regenerative capacity to VF dECM. SIS dECM is considered a promising alternative for dECM-derived biomaterials for VF regeneration.
Collapse
Affiliation(s)
- Mika Brown
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
| | - Shirley Zhu
- Department of Microbiology and Immunology 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
| | - Lorne Taylor
- The Proteomics Platform, McGill University Health Center 1001 Decarie Boulevard Montreal Suite E01.5056 Montreal, Quebec, H4A 3J1, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
- Department of Bioengineering, McGill University 740 Avenue Dr. Penfield, Room 4300, Montreal, QC H3A 0G1, Canada
- Faculty of Dentistry, McGill University 740 Avenue Dr. Penfield, Room 4300, Montreal, QC H3A 0G1, Canada
| | - Nicole Y.K. Li-Jessen
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
- School of Communication Sciences and Disorders, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
- Department of Otolaryngology - Head and Neck Surgery, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
- Research Institute of McGill University Health Center, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
| |
Collapse
|
21
|
Kulikauskas MR, Oatley M, Yu T, Liu Z, Matsumura L, Kidder E, Ruter D, Bautch VL. Endothelial Cell SMAD6 Balances ACVRL1/Alk1 Function to Regulate Adherens Junctions and Hepatic Vascular Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.534007. [PMID: 36993438 PMCID: PMC10055411 DOI: 10.1101/2023.03.23.534007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
BMP signaling is critical to blood vessel formation and function, but how pathway components regulate vascular development is not well-understood. Here we find that inhibitory SMAD6 functions in endothelial cells to negatively regulate ALK1/ACVRL1-mediated responses, and it is required to prevent vessel dysmorphogenesis and hemorrhage in the embryonic liver vasculature. Reduced Alk1 gene dosage rescued embryonic hepatic hemorrhage and microvascular capillarization induced by Smad6 deletion in endothelial cells in vivo . At the cellular level, co-depletion of Smad6 and Alk1 rescued the destabilized junctions and impaired barrier function of endothelial cells depleted for SMAD6 alone. At the mechanistic level, blockade of actomyosin contractility or increased PI3K signaling rescued endothelial junction defects induced by SMAD6 loss. Thus, SMAD6 normally modulates ALK1 function in endothelial cells to regulate PI3K signaling and contractility, and SMAD6 loss increases signaling through ALK1 that disrupts endothelial junctions. ALK1 loss-of-function also disrupts vascular development and function, indicating that balanced ALK1 signaling is crucial for proper vascular development and identifying ALK1 as a "Goldilocks" pathway in vascular biology regulated by SMAD6.
Collapse
Affiliation(s)
- Molly R Kulikauskas
- Cell Biology and Physiology Curriculum, The University of North Carolina, Chapel Hill, NC USA
| | - Morgan Oatley
- Department of Biology, The University of North Carolina, Chapel Hill, NC USA
| | - Tianji Yu
- Department of Biology, The University of North Carolina, Chapel Hill, NC USA
| | - Ziqing Liu
- Department of Biology, The University of North Carolina, Chapel Hill, NC USA
| | - Lauren Matsumura
- Department of Biology, The University of North Carolina, Chapel Hill, NC USA
| | - Elise Kidder
- Department of Biology, The University of North Carolina, Chapel Hill, NC USA
| | - Dana Ruter
- Department of Biology, The University of North Carolina, Chapel Hill, NC USA
| | - Victoria L Bautch
- Cell Biology and Physiology Curriculum, The University of North Carolina, Chapel Hill, NC USA
- Department of Biology, The University of North Carolina, Chapel Hill, NC USA
- McAllister Heart Institute, The University of North Carolina, Chapel Hill, NC USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC USA
| |
Collapse
|
22
|
Zhang H, Liesveld JL, Calvi LM, Lipe BC, Xing L, Becker MW, Schwarz EM, Yeh SCA. The roles of bone remodeling in normal hematopoiesis and age-related hematological malignancies. Bone Res 2023; 11:15. [PMID: 36918531 PMCID: PMC10014945 DOI: 10.1038/s41413-023-00249-w] [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: 08/01/2022] [Revised: 12/24/2022] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Prior research establishing that bone interacts in coordination with the bone marrow microenvironment (BMME) to regulate hematopoietic homeostasis was largely based on analyses of individual bone-associated cell populations. Recent advances in intravital imaging has suggested that the expansion of hematopoietic stem cells (HSCs) and acute myeloid leukemia cells is restricted to bone marrow microdomains during a distinct stage of bone remodeling. These findings indicate that dynamic bone remodeling likely imposes additional heterogeneity within the BMME to yield differential clonal responses. A holistic understanding of the role of bone remodeling in regulating the stem cell niche and how these interactions are altered in age-related hematological malignancies will be critical to the development of novel interventions. To advance this understanding, herein, we provide a synopsis of the cellular and molecular constituents that participate in bone turnover and their known connections to the hematopoietic compartment. Specifically, we elaborate on the coupling between bone remodeling and the BMME in homeostasis and age-related hematological malignancies and after treatment with bone-targeting approaches. We then discuss unresolved questions and ambiguities that remain in the field.
Collapse
Affiliation(s)
- Hengwei Zhang
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Jane L Liesveld
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura M Calvi
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Endocrinology/Metabolism, University of Rochester Medical Center, Rochester, NY, USA
| | - Brea C Lipe
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Allergy/Immunology/Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Shu-Chi A Yeh
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Physiology/Pharmacology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
23
|
Vliora M, Ravelli C, Grillo E, Corsini M, Flouris AD, Mitola S. The impact of adipokines on vascular networks in adipose tissue. Cytokine Growth Factor Rev 2023; 69:61-72. [PMID: 35953434 DOI: 10.1016/j.cytogfr.2022.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023]
Abstract
Adipose tissue (AT) is a highly active and plastic endocrine organ. It secretes numerous soluble molecules known as adipokines, which act locally to AT control the remodel and homeostasis or exert pleiotropic functions in different peripheral organs. Aberrant production or loss of certain adipokines contributes to AT dysfunction associated with metabolic disorders, including obesity. The AT plasticity is strictly related to tissue vascularization. Angiogenesis supports the AT expansion, while regression of blood vessels is associated with AT hypoxia, which in turn mediates tissue inflammation, fibrosis and metabolic dysfunction. Several adipokines can regulate endothelial cell functions and are endowed with either pro- or anti-angiogenic properties. Here we address the role of adipokines in the regulation of angiogenesis. A better understanding of the link between adipokines and angiogenesis will open the way for novel therapeutic approaches to treat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Vliora
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece; Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Michela Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy
| | - Andreas D Flouris
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Via Branze 39, Brescia, Italy.
| |
Collapse
|
24
|
Grillo E, Ravelli C, Colleluori G, D'Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev 2023; 69:51-60. [PMID: 36155165 DOI: 10.1016/j.cytogfr.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Francesco D'Agostino
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Domenichini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
25
|
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
|
26
|
Fang J, Liu H, Qiao W, Xu T, Yang Y, Xie H, Lam CH, Yeung KWK, Zhao X. Biomimicking Leaf-Vein Engraved Soft and Elastic Membrane Promotes Vascular Reconstruction. Adv Healthc Mater 2023; 12:e2201220. [PMID: 36330558 DOI: 10.1002/adhm.202201220] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Hierarchical vasculature reconstruction is fundamental for tissue regeneration. The regeneration of functional vascular network requires a proper directional guidance, especially in case of large-size defects. To provide the "running track" for vasculature, a leaf-vein mimetic membrane using soft and elastic poly(lactide-co-propylene glycol-co-lactide) dimethacrylate is developed. Engraved with an interconnected and perfusable leaf-vein micropattern, the membrane can guide human umbilical vein endothelial cells (HUVECs) to form vasculature in vitro. In particular, the "running track" upregulates the angiogenesis-related gene expression and promotes the HUVECs to differentiate into tip cells and stalk cells via tuning vascular endothelial growth factor receptor 2 signaling transduction. As a proof of concept, its revascularization capability using a rat calvarial defect model in vivo is evaluated. The in vivo results demonstrate that the leaf-vein engraved membrane accelerates the formation and maturation of vasculature, leading to a hierarchical blood vessel network. With the superior pro-vasculature property, it is believed that the leaf-vein engraved membrane is not only an ideal candidate for the reconstruction of calvarial vasculature but also a promising solution for more complicated vasculature reconstruction, such as muscle, skin, and heart.
Collapse
Affiliation(s)
- Jinghan Fang
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Huaqian Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Wei Qiao
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Tianpeng Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Yuhe Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Huizhi Xie
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Chun-Hei Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Kelvin W K Yeung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| |
Collapse
|
27
|
Sulfated galactoglucan impedes xenografted lung cancer cell growth by blocking angiogenesis via binding BMPRs. Carbohydr Polym 2022; 289:119412. [DOI: 10.1016/j.carbpol.2022.119412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/28/2022] [Accepted: 03/22/2022] [Indexed: 11/20/2022]
|
28
|
Feng R, Lu M, Yang Y, Luo P, Liu L, Xu K, Xu P. Genome- and transcriptome-wide association studies show that pulmonary embolism is associated with bone-forming proteins. Expert Rev Hematol 2022; 15:951-958. [PMID: 35848930 DOI: 10.1080/17474086.2022.2103534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Pulmonary embolism (PE) is a leading cause of death in stroke patients and a severe health burden worldwide. There is a pressing need to understand the mechanisms by which it occurs and to identify at-risk patients efficiently and accurately. OBJECTIVES The aim of this paper was to analyze the genetic correlation between PE and human plasma proteins through genome-wide association study (GWAS) with transcriptome-wide association study (TWAS), in combination with mRNA expression profiling at three levels: DNA, RNA, and protein. METHODS First, based on data from GWAS in European populations, we performed a linkage disequilibrium score regression (LDSC) analysis of plasma proteins and PE in 3,283 individuals and additionally analyzed the genetic association between PE and fracture. Then, we performed a TWAS on PE GWAS data using skeletal muscle and blood for gene expression references. Finally, we validated the genetic correlation between PE and human plasma proteins by co-matching the genes encoding the identified proteins and those identified using TWAS with the differentially expressed genes obtained from mRNA expression profiling of PE (Figure1). RESULTS We identified five plasma proteins associated with PE, including hydroxycarboxylic acid receptor 2, defensin 118, and bone morphogenetic protein (BMP) 7, as well as a relationship between PE and fracture. Comparison of genes encoding these proteins with genes obtained from TWAS and then with differentially expressed genes obtained from PE mRNA expression profiling revealed that PE was highly correlated with the BMP family of genes.
Collapse
Affiliation(s)
- Ruoyang Feng
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Mengnan Lu
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yanni Yang
- Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Pan Luo
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Lin Liu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi, 710054, China
| |
Collapse
|
29
|
Kaspiris A, Hadjimichael AC, Vasiliadis ES, Papachristou DJ, Giannoudis PV, Panagiotopoulos EC. Therapeutic Efficacy and Safety of Osteoinductive Factors and Cellular Therapies for Long Bone Fractures and Non-Unions: A Meta-Analysis and Systematic Review. J Clin Med 2022; 11:3901. [PMID: 35807186 PMCID: PMC9267779 DOI: 10.3390/jcm11133901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Long bone fractures display significant non-union rates, but the exact biological mechanisms implicated in this devastating complication remain unclear. The combination of osteogenetic and angiogenetic factors at the fracture site is an essential prerequisite for successful bone regeneration. The aim of this study is to investigate the results of the clinical implantation of growth factors for intraoperative enhancement of osteogenesis for the treatment of long bone fractures and non-unions. METHODS A systematic literature review search was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines in the PubMed and Web of Science databases from the date of inception of each database through to 10 January 2022. Specific inclusion and exclusion criteria were applied in order to identify relevant studies reporting on the treatment of upper and lower limb long bone non-unions treated with osteoinductive or cellular factors. RESULTS Overall, 18 studies met the inclusion criteria and examined the effectiveness of the application of Bone Morphogenetic Proteins-2 and -7 (BMPs), platelet rich plasma (PRP) and mesenchymal stem cells (MSCs). Despite the existence of limitations in the studies analysed (containing mixed groups of open and close fractures, different types of fractures, variability of treatment protocols, different selection criteria and follow-up periods amongst others), their overall effectiveness was found significantly increased in patients who received them compared with the controls (I2 = 60%, 95% CI = 1.59 [0.99-2.54], Z =1.93, p = 0.05). CONCLUSION Administration of BMP-2 and -7, PRP and MSCs were considered effective and safe methods in fracture treatment, increasing bone consolidation, reducing time to repair and being linked to satisfactory postoperative functional scores.
Collapse
Affiliation(s)
- Angelos Kaspiris
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Argyris C. Hadjimichael
- Department of Orthopaedics, St. Mary’s Hospital, Imperial College Healthcare NHS Trust, Praed Street, London W2 1NY, UK;
| | - Elias S. Vasiliadis
- Third Department of Orthopaedic Surgery, School of Medicine, “KAT” General Hospital, National and Kapodistrian University of Athens, 2 Nikis Street, 14561 Athens, Greece;
| | - Dionysios J. Papachristou
- Laboratory of Bone and Soft Tissue Studies, Department of Anatomy-Histology-Embryology, University Patras Medical School, 26504 Patras, Greece;
| | - Peter V. Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds LS7 4SA, UK;
- NIHR Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds LS7 4SA, UK
| | - Elias C. Panagiotopoulos
- Department of Trauma and Orthopaedics, Patras University Hospital and Medical School, 26504 Patras, Greece;
| |
Collapse
|
30
|
Long X, Qiu Z, Li C, Wang Y, Li J, Zhao R, Rong J, Gu N, Yuan J, Ge J, Shi B. CircERBB2IP promotes post-infarction revascularization via the miR-145a-5p/Smad5 axis. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:573-586. [PMID: 35592503 PMCID: PMC9096260 DOI: 10.1016/j.omtn.2022.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 04/15/2022] [Indexed: 10/27/2022]
|
31
|
Kulikauskas MR, X S, Bautch VL. The versatility and paradox of BMP signaling in endothelial cell behaviors and blood vessel function. Cell Mol Life Sci 2022; 79:77. [PMID: 35044529 PMCID: PMC8770421 DOI: 10.1007/s00018-021-04033-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022]
Abstract
Blood vessels expand via sprouting angiogenesis, and this process involves numerous endothelial cell behaviors, such as collective migration, proliferation, cell–cell junction rearrangements, and anastomosis and lumen formation. Subsequently, blood vessels remodel to form a hierarchical network that circulates blood and delivers oxygen and nutrients to tissue. During this time, endothelial cells become quiescent and form a barrier between blood and tissues that regulates transport of liquids and solutes. Bone morphogenetic protein (BMP) signaling regulates both proangiogenic and homeostatic endothelial cell behaviors as blood vessels form and mature. Almost 30 years ago, human pedigrees linked BMP signaling to diseases associated with blood vessel hemorrhage and shunts, and recent work greatly expanded our knowledge of the players and the effects of vascular BMP signaling. Despite these gains, there remain paradoxes and questions, especially with respect to how and where the different and opposing BMP signaling outputs are regulated. This review examines endothelial cell BMP signaling in vitro and in vivo and discusses the paradox of BMP signals that both destabilize and stabilize endothelial cell behaviors.
Collapse
Affiliation(s)
- Molly R Kulikauskas
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shaka X
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
32
|
Vakhrushev IV, Nezhurina EK, Karalkin PA, Tsvetkova AV, Sergeeva NS, Majouga AG, Yarygin KN. Heterotypic Multicellular Spheroids as Experimental and Preclinical Models of Sprouting Angiogenesis. BIOLOGY 2021; 11:18. [PMID: 35053016 PMCID: PMC8772844 DOI: 10.3390/biology11010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
Sprouting angiogenesis is the common response of live tissues to physiological and pathological angiogenic stimuli. Its accurate evaluation is of utmost importance for basic research and practical medicine and pharmacology and requires adequate experimental models. A variety of assays for angiogenesis were developed, none of them perfect. In vitro approaches are generally less physiologically relevant due to the omission of essential components regulating the process. However, only in vitro models can be entirely non-xenogeneic. The limitations of the in vitro angiogenesis assays can be partially overcome using 3D models mimicking tissue O2 and nutrient gradients, the influence of the extracellular matrix (ECM), and enabling cell-cell interactions. Here we present a review of the existing models of sprouting angiogenesis that are based on the use of endothelial cells (ECs) co-cultured with perivascular or other stromal cells. This approach provides an excellent in vitro platform for further decoding of the cellular and molecular mechanisms of sprouting angiogenesis under conditions close to the in vivo conditions, as well as for preclinical drug testing and preclinical research in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Igor V. Vakhrushev
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | - Elizaveta K. Nezhurina
- P.A. Hertsen Moscow Oncology Research Center, National Medical Research Radiological Center, 125284 Moscow, Russia;
| | - Pavel A. Karalkin
- Institute for Cluster Oncology, Sechenov University, 119435 Moscow, Russia;
| | | | - Nataliya S. Sergeeva
- Department of Biology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Alexander G. Majouga
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| |
Collapse
|
33
|
Comparative Transcriptomic Analysis of Regenerated Skins Provides Insights into Cutaneous Air-Breathing Formation in Fish. BIOLOGY 2021; 10:biology10121294. [PMID: 34943209 PMCID: PMC8698756 DOI: 10.3390/biology10121294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022]
Abstract
Cutaneous air-breathing is one of the air-breathing patterns in bimodal respiration fishes, while little is known about its underlying formation mechanisms. Here, we first investigated the skin regeneration of loach (Misgurnus anguillicaudatus, a cutaneous air-breathing fish) and yellow catfish (Pelteobagrus fulvidraco, a water-breathing fish) through morphological and histological observations. Then, the original skins (OS: MOS, POS) and regenerated skins (RS: MRS, PRS) when their capillaries were the most abundant (the structural foundation of air-breathing in fish) during healing, of the two fish species were collected for high-throughput RNA-seq. A total of 56,054 unigenes and 53,731 unigenes were assembled in loach and yellow catfish, respectively. A total of 640 (460 up- and 180 down-regulated) and 4446 (2340 up- and 2106 down-regulated) differentially expressed genes (DEGs) were respectively observed in RS/OS of loach and yellow catfish. Subsequently, the two DEG datasets were clustered in GO, KOG and KEGG databases, and further analyzed by comparison and screening. Consequently, tens of genes and thirteen key pathways were targeted, indicating that these genes and pathways had strong ties to cutaneous skin air-breathing in loach. This study provides new insights into the formation mechanism of cutaneous air-breathing and also offers a substantial contribution to the gene expression profiles of skin regeneration in fish.
Collapse
|
34
|
Liu W, Luo H, Wei Q, Liu J, Wu J, Zhang Y, Chen L, Ren W, Shao L. Electrochemically derived nanographene oxide activates endothelial tip cells and promotes angiogenesis by binding endogenous lysophosphatidic acid. Bioact Mater 2021; 9:92-104. [PMID: 34820558 PMCID: PMC8586026 DOI: 10.1016/j.bioactmat.2021.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/08/2023] Open
Abstract
Graphene oxide (GO) exhibits good mechanical and physicochemical characteristics and has extensive application prospects in bone tissue engineering. However, its effect on angiogenesis is unclear, and its potential toxic effects are heavily disputed. Herein, we found that nanographene oxide (NGO) synthesized by one-step water electrolytic oxidation is smaller and shows superior biocompatibility. Moreover, NGO significantly enhanced angiogenesis in calvarial bone defect areas in vivo, providing a good microenvironment for bone regeneration. Endothelial tip cell differentiation is an important step in the initiation of angiogenesis. We verified that NGO activates endothelial tip cells by coupling with lysophosphatidic acid (LPA) in serum via strong hydrogen bonding interactions, which has not been reported. In addition, the mechanism by which NGO promotes angiogenesis was systematically studied. NGO-coupled LPA activates LPAR6 and facilitates the formation of migratory tip cells via Hippo/Yes-associated protein (YAP) independent of reactive oxygen species (ROS) stimulation or additional complex modifications. These results provide an effective strategy for the application of electrochemically derived NGO and more insight into NGO-mediated angiogenesis. Electrochemically derived nanographene oxide (NGO) has good cytocompatibility without upregulating reactive oxygen species. NGO exhibits better dispersibility and couples with endogenous lysophosphatidic acid (LPA) in body fluid. NGO enhances the angiogenesis by recruiting endogenous LPA and promoting endothelial tip cell formation.
Collapse
Affiliation(s)
- Wenjing Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Haiyun Luo
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qinwei Wei
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Junrong Wu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Lili Chen
- Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wencai Ren
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
- Corresponding author. Stomatological Hospital, Southern Medical University, Guangzhou 510280, China Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China.
| |
Collapse
|
35
|
Boog H, Medda R, Cavalcanti-Adam EA. Single Cell Center of Mass for the Analysis of BMP Receptor Heterodimers Distributions. J Imaging 2021; 7:jimaging7110219. [PMID: 34821850 PMCID: PMC8620704 DOI: 10.3390/jimaging7110219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 11/16/2022] Open
Abstract
At the plasma membrane, transmembrane receptors are at the interface between cells and their environment. They allow sensing and transduction of chemical and mechanical extracellular signals. The spatial distribution of receptors and the specific recruitment of receptor subunits to the cell membrane is crucial for the regulation of signaling and cell behavior. However, it is challenging to define what regulates such spatial patterns for receptor localization, as cell shapes are extremely diverse when cells are maintained in standard culture conditions. Bone morphogenetic protein receptors (BMPRs) are serine-threonine kinases, which build heteromeric complexes of BMPRI and II. These are especially interesting targets for receptor distribution studies, since the signaling pathways triggered by BMPR-complexes depends on their dimerization mode. They might exist as preformed complexes, or assemble upon binding of BMP, triggering cell signaling which leads to differentiation or migration. In this work we analyzed BMPR receptor distributions in single cells grown on micropatterns, which allow not only to control cell shape, but also the distribution of intracellular organelles and protein assemblies. We developed a script called ComRed (Center Of Mass Receptor Distribution), which uses center of mass calculations to analyze the shift and spread of receptor distributions according to the different cell shapes. ComRed was tested by simulating changes in experimental data showing that shift and spread of distributions can be reliably detected. Our ComRed-based analysis of BMPR-complexes indicates that receptor distribution depends on cell polarization. The absence of a coordinated internalization after addition of BMP suggests that a rapid and continual recycling of BMPRs might occur. Receptor complexes formation and localization in cells induced by BMP might yield insights into the local regulation of different signaling pathways.
Collapse
Affiliation(s)
- Hendrik Boog
- Department of Cellular Biophysics-Growth Factor Mechanobiology, Max-Planck-Institute for Medical Research, 69120 Heidelberg, Germany; (H.B.); (R.M.)
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht-Karls-Universitaet Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Rebecca Medda
- Department of Cellular Biophysics-Growth Factor Mechanobiology, Max-Planck-Institute for Medical Research, 69120 Heidelberg, Germany; (H.B.); (R.M.)
| | - Elisabetta Ada Cavalcanti-Adam
- Department of Cellular Biophysics-Growth Factor Mechanobiology, Max-Planck-Institute for Medical Research, 69120 Heidelberg, Germany; (H.B.); (R.M.)
- Correspondence:
| |
Collapse
|
36
|
Sun H, Dong J, Wang Y, Shen S, Shi Y, Zhang L, Zhao J, Sun X, Jiang Q. Polydopamine-Coated Poly(l-lactide) Nanofibers with Controlled Release of VEGF and BMP-2 as a Regenerative Periosteum. ACS Biomater Sci Eng 2021; 7:4883-4897. [PMID: 34472855 DOI: 10.1021/acsbiomaterials.1c00246] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The periosteum plays an important role in vascularization and ossification during bone repair. However, in most studies, an artificial periosteum cannot restore both functions of the periosteum concurrently. In this study, a novel nanofiber that can sustain the release of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-2 (BMP-2) was fabricated to enhance the durability of angiogenesis and osteogenesis during bone regeneration. A cell-free tissue engineered periosteum based on an electrospinning poly-l-lactic acid (PLLA) nanofiber was fabricated, on which VEGF and BMP-2 were immobilized through a polydopamine (PDA) coating conveniently and safely (BVP@PLLA membrane). The results indicated a significantly improved loading rate as well as a slow and sustained release of VEGF and BMP-2 with the help of the PDA coating. BMP-2 immobilized on nanofibers successfully induced the osteogenic differentiation of human bone marrow mesenchymal stem cells (BMSCs) in vitro with high expression of runt-related transcription factor 2 (Runx2), osteopontin (OPN), and alkaline phosphatase (ALP). Similarly, angiogenic differentiation of BMSCs with the expression of fetal liver kinase-1 (Flk-1) and vascular endothelial cadherin (VE-cadherin) was observed under the environment of VEGF sustained release. Moreover, an in vivo study revealed that the BVP@PLLA membrane could enhance vascular formation and new bone formation, which accelerates bone regeneration in rat femoral defects along with a massive periosteum defect. Therefore, our study suggests that the novel artificial periosteum with dual growth factor controlled release is a promising system to improve bone regeneration in bone defects along with a massive periosteum defect.
Collapse
Affiliation(s)
- Han Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, Jiangsu 213003, P.R. China
| | - Jian Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China
| | - Yangyufan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China
| | - Siyu Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China
| | - Yong Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China
| | - Lei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China
| | - Jie Zhao
- Department of Orthopedics, The Affiliated Wujin Hospital of Jiangsu University, 2 Yongning Road, Changzhou, Jiangsu 213003, P.R. China
| | - Xiaoliang Sun
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, Jiangsu 213003, P.R. China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
37
|
Hu H, Wang S, He Y, Shen S, Yao B, Xu D, Liu X, Zhang Y. The role of bone morphogenetic protein 4 in corneal injury repair. Exp Eye Res 2021; 212:108769. [PMID: 34537186 DOI: 10.1016/j.exer.2021.108769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 08/30/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE Corneal injury may cause neovascularization and lymphangiogenesis in cornea which have a detrimental effect to vision and even lead to blindness. Bone morphogenetic protein 4 (BMP4) regulates a variety of biological processes, which is closely relevant to the regulation of corneal epithelium and angiogenesis. Herein, we aimed to evaluate the effect of BMP4 on corneal neovascularization (CNV), corneal lymphangiogenesis (CL), corneal epithelial repair, and the role of BMP4/Smad pathway in these processes. METHODS We used MTT assay to determine the optimal concentration of BMP4. The suture method was performed to induce rat CNV and CL. We used ink perfusion and HE staining to visualize the morphological change of CNV, and utilized RT-qPCR and ELISA to investigate the expression of angiogenic factors and lymphangiogenic factors. The effects of BMP4 and anti-VEGF antibody on migration, proliferation and adhesion of corneal epithelium were determined by scratch test, MTT assay and cell adhesion test. RESULTS BMP4 significantly inhibited CNV and possibly CL. Topical BMP4 resulted in increased expression of endogenous BMP4, and decreased expression of angiogenic factors and lymphangiogenic factors. Compared with anti-VEGF antibody, BMP4 enhanced corneal epithelium migration, proliferation and adhesion, which facilitated corneal epithelial injury repair. Simultaneously, these processes could be regulated by BMP4/Smad pathway. CONCLUSIONS Our results demonstrated unreported effects of BMP4 on CNV, CL, and corneal epithelial repair, suggesting that BMP4 may represent a potential therapeutic target in corneal injury repair.
Collapse
Affiliation(s)
- Huicong Hu
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Shurong Wang
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Yuxi He
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Sitong Shen
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Boyuan Yao
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Duo Xu
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Xin Liu
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Yan Zhang
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
38
|
Endothelial Heterogeneity in Development and Wound Healing. Cells 2021; 10:cells10092338. [PMID: 34571987 PMCID: PMC8469713 DOI: 10.3390/cells10092338] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
The vasculature is comprised of endothelial cells that are heterogeneous in nature. From tissue resident progenitors to mature differentiated endothelial cells, the diversity of these populations allows for the formation, maintenance, and regeneration of the vascular system in development and disease, particularly during situations of wound healing. Additionally, the de-differentiation and plasticity of different endothelial cells, especially their capacity to undergo endothelial to mesenchymal transition, has also garnered significant interest due to its implication in disease progression, with emphasis on scarring and fibrosis. In this review, we will pinpoint the seminal discoveries defining the phenotype and mechanisms of endothelial heterogeneity in development and disease, with a specific focus only on wound healing.
Collapse
|
39
|
Guo Y, Mei F, Huang Y, Ma S, Wei Y, Zhang X, Xu M, He Y, Heng BC, Chen L, Deng X. Matrix stiffness modulates tip cell formation through the p-PXN-Rac1-YAP signaling axis. Bioact Mater 2021; 7:364-376. [PMID: 34466738 PMCID: PMC8379356 DOI: 10.1016/j.bioactmat.2021.05.033] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/05/2021] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Endothelial tip cell outgrowth of blood-vessel sprouts marks the initiation of angiogenesis which is critical in physiological and pathophysiological procedures. However, how mechanical characteristics of extracellular matrix (ECM) modulates tip cell formation has been largely neglected. In this study, we found enhanced CD31 expression in the stiffening outer layer of hepatocellular carcinoma than in surrounding soft tissues. Stiffened matrix promoted sprouting from endothelial cell (EC) spheroids and upregulated expressions of tip cell-enriched genes in vitro. Moreover, tip cells showed increased cellular stiffness, more actin cytoskeleton organization and enhanced YAP nuclear transfer than stalk and phalanx ECs. We further uncovered that substrate stiffness regulates FAK and Paxillin phosphorylation in focal adhesion of ECs promoting Rac1 transition from inactive to active state. YAP is subsequently activated and translocated into nucleus, leading to increased tip cell specification. p-Paxillin can also loosen the intercellular connection which also facilitates tip cell specification. Collectively our present study shows that matrix stiffness modulates tip cell formation through p-PXN-Rac1-YAP signaling axis, shedding light on the role of mechanotransduction in tip cell formation. This is of special significance in biomaterial design and treatment of some pathological situations. Mechanotransduction is implicated in angiogenesis and tip cell formation. Tip cells showed different mechanical properties from stalk and phalanx ECs. Paxillin, Rac1 and YAP might be novel treatment targets for some diseases. Material stiffness affects tip cell specification.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Feng Mei
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ying Huang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Siqin Ma
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Xuehui Zhang
- Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, PR China
- Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
- Corresponding author. Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China
- Department of Dental Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, 100081, PR China
- Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Corresponding author. Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| |
Collapse
|
40
|
Tang H, Zhang X, Xue G, Xu F, Wang Q, Yang P, Hong B, Xu Y, Huang Q, Liu J, Zuo Q. The biology of bone morphogenetic protein signaling pathway in cerebrovascular system. Chin Neurosurg J 2021; 7:36. [PMID: 34465399 PMCID: PMC8408949 DOI: 10.1186/s41016-021-00254-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/08/2021] [Indexed: 11/30/2022] Open
Abstract
Bone morphogenetic protein belongs to transcription growth factor superfamily β; bone morphogenetic protein signal pathway regulates cell proliferation, differentiation, and apoptosis among different tissues. Cerebrovascular system supplies sufficient oxygen and blood into brain to maintain its normal function. The disorder of cerebrovascular system will result into serious cerebrovascular diseases, which is gradually becoming a major threat to human health in modern society. In recent decades, many studies have revealed the underlying biology and mechanism of bone morphogenetic protein signal pathway played in cerebrovascular system. This review will discuss the relationship between the two aspects, aiming to provide new perspective for non-invasive treatment and basic research of cerebrovascular diseases.
Collapse
Affiliation(s)
- Haishuang Tang
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China.,Naval Medical Center of PLA, Naval Military Medical University, Shanghai, 200050, People's Republic of China
| | - Xiaoxi Zhang
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Gaici Xue
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Fengfeng Xu
- Naval Medical Center of PLA, Naval Military Medical University, Shanghai, 200050, People's Republic of China
| | - Qingsong Wang
- Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, People's Republic of China
| | - Pengfei Yang
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Bo Hong
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Yi Xu
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Qinghai Huang
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China
| | - Jianmin Liu
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China.
| | - Qiao Zuo
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, 168 Changhai Road, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
41
|
Zeng A, Wang SR, He YX, Yan Y, Zhang Y. Progress in understanding of the stalk and tip cells formation involvement in angiogenesis mechanisms. Tissue Cell 2021; 73:101626. [PMID: 34479073 DOI: 10.1016/j.tice.2021.101626] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/28/2022]
Abstract
Vascular sprouting is a key process of angiogenesis and mainly related to the formation of stalk and tip cells. Many studies have found that angiogenesis has a great clinical significance in promoting the functional repair of impaired tissues and anti-angiogenesis is a key to treatment of many tumors. Therefore, how the pathways regulate angiogenesis by regulating the formation of stalk and tip cells is an urgent problem for researchers. This review mainly summarizes the research progress of pathways affecting the formation of stalk and tip cells during angiogenesis in recent years, including the main signaling pathways (such as VEGF-VEGFR-Dll4-Notch signaling pathway, ALK-Smad signaling pathway,CCN1-YAP/YAZ signaling pathway and other signaling pathways) and cellular actions (such as cellular metabolisms, intercellular tension and other actions), aiming to further give the readers an insight into the mechanism of regulating the formation of stalk and tip cells during angiogenesis and provide more targets for anti-angiogenic drugs.
Collapse
Affiliation(s)
- Ao Zeng
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Shu-Rong Wang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Yu-Xi He
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Yu Yan
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Yan Zhang
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China.
| |
Collapse
|
42
|
Cornelissen A, Guo L, Fernandez R, Kelly MC, Janifer C, Kuntz S, Sakamoto A, Jinnouchi H, Sato Y, Paek KH, Kolodgie FD, Romero ME, Surve D, Virmani R, Finn AV. Endothelial Recovery in Bare Metal Stents and Drug-Eluting Stents on a Single-Cell Level. Arterioscler Thromb Vasc Biol 2021; 41:2277-2292. [PMID: 34162228 DOI: 10.1161/atvbaha.121.316472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anne Cornelissen
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,Department of Cardiology, University Hospital RWTH Aachen, Germany (A.C.)
| | - Liang Guo
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Raquel Fernandez
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Michael C Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD (M.C.K.)
| | - Christine Janifer
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Salome Kuntz
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Atsushi Sakamoto
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Hiroyuki Jinnouchi
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Yu Sato
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Ka Hyun Paek
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Frank D Kolodgie
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Maria E Romero
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Dipti Surve
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Renu Virmani
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Aloke V Finn
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,University of Maryland, School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
43
|
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: 1.8] [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
|
44
|
Inhibiting Endothelial Cell Function in Normal and Tumor Angiogenesis Using BMP Type I Receptor Macrocyclic Kinase Inhibitors. Cancers (Basel) 2021; 13:cancers13122951. [PMID: 34204675 PMCID: PMC8231556 DOI: 10.3390/cancers13122951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 01/05/2023] Open
Abstract
Angiogenesis, i.e., the formation of new blood vessels from pre-existing endothelial cell (EC)-lined vessels, is critical for tissue development and also contributes to neovascularization-related diseases, such as cancer. Vascular endothelial growth factor (VEGF) and bone morphogenetic proteins (BMPs) are among many secreted cytokines that regulate EC function. While several pharmacological anti-angiogenic agents have reached the clinic, further improvement is needed to increase clinical efficacy and to overcome acquired therapy resistance. More insights into the functional consequences of targeting specific pathways that modulate blood vessel formation may lead to new therapeutic approaches. Here, we synthesized and identified two macrocyclic small molecular compounds termed OD16 and OD29 that inhibit BMP type I receptor (BMPRI)-induced SMAD1/5 phosphorylation and downstream gene expression in ECs. Of note, OD16 and OD29 demonstrated higher specificity against BMPRI activin receptor-like kinase 1/2 (ALK1/2) than the commonly used small molecule BMPRI kinase inhibitor LDN-193189. OD29, but not OD16, also potently inhibited VEGF-induced extracellular regulated kinase MAP kinase phosphorylation in ECs. In vitro, OD16 and OD29 exerted strong inhibition of BMP9 and VEGF-induced ECs migration, invasion and cord formation. Using Tg (fli:EGFP) zebrafish embryos, we found that OD16 and OD29 potently antagonized dorsal longitudinal anastomotic vessel (DLAV), intra segmental vessel (ISV), and subintestinal vessel (SIV) formation during embryonic development. Moreover, the MDA-MB-231 breast cancer cell-induced tumor angiogenesis in zebrafish embryos was significantly decreased by OD16 and OD29. Both macrocyclic compounds might provide a steppingstone for the development of novel anti-angiogenesis therapeutic agents.
Collapse
|
45
|
Bhatti FUR, Dadwal UC, Valuch CR, Tewari NP, Awosanya OD, de Andrade Staut C, Sun S, Mendenhall SK, Perugini AJ, Nagaraj RU, Battina HL, Nazzal MK, Blosser RJ, Maupin KA, Childress PJ, Li J, Kacena MA. The effects of high fat diet, bone healing, and BMP-2 treatment on endothelial cell growth and function. Bone 2021; 146:115883. [PMID: 33581374 PMCID: PMC8009863 DOI: 10.1016/j.bone.2021.115883] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023]
Abstract
Angiogenesis is a vital process during the regeneration of bone tissue. The aim of this study was to investigate angiogenesis at the fracture site as well as at distal locations from obesity-induced type 2 diabetic mice that were treated with bone morphogenetic protein-2 (BMP-2, local administration at the time of surgery) to heal a femoral critical sized defect (CSD) or saline as a control. Mice were fed a high fat diet (HFD) to induce a type 2 diabetic-like phenotype while low fat diet (LFD) animals served as controls. Endothelial cells (ECs) were isolated from the lungs (LECs) and bone marrow (BMECs) 3 weeks post-surgery, and the fractured femurs were also examined. Our studies demonstrate that local administration of BMP-2 at the fracture site in a CSD model results in complete bone healing within 3 weeks for all HFD mice and 66.7% of LFD mice, whereas those treated with saline remain unhealed. At the fracture site, vessel parameters and adipocyte numbers were significantly increased in BMP-2 treated femurs, irrespective of diet. At distal sites, LEC and BMEC proliferation was not altered by diet or BMP-2 treatment. HFD increased the tube formation ability of both LECs and BMECs. Interestingly, BMP-2 treatment at the time of surgery reduced tube formation in LECs and humeri BMECs. However, migration of BMECs from HFD mice treated with BMP-2 was increased compared to BMECs from HFD mice treated with saline. BMP-2 treatment significantly increased the expression of CD31, FLT-1, and ANGPT2 in LECs and BMECs in LFD mice, but reduced the expression of these same genes in HFD mice. To date, this is the first study that depicts the systemic influence of fracture surgery and local BMP-2 treatment on the proliferation and angiogenic potential of ECs derived from the bone marrow and lungs.
Collapse
Affiliation(s)
- Fazal Ur Rehman Bhatti
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Ushashi C Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Conner R Valuch
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Nikhil P Tewari
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Olatundun D Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | | | - Seungyup Sun
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Stephen K Mendenhall
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Anthony J Perugini
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rohit U Nagaraj
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Hanisha L Battina
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rachel J Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Kevin A Maupin
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Paul J Childress
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA.
| |
Collapse
|
46
|
Elmasry K, Habib S, Moustafa M, Al-Shabrawey M. Bone Morphogenetic Proteins and Diabetic Retinopathy. Biomolecules 2021; 11:biom11040593. [PMID: 33919531 PMCID: PMC8073699 DOI: 10.3390/biom11040593] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) play an important role in bone formation and repair. Recent studies underscored their essential role in the normal development of several organs and vascular homeostasis in health and diseases. Elevated levels of BMPs have been linked to the development of cardiovascular complications of diabetes mellitus. However, their particular role in the pathogenesis of microvascular dysfunction associated with diabetic retinopathy (DR) is still under-investigated. Accumulated evidence from our and others’ studies suggests the involvement of BMP signaling in retinal inflammation, hyperpermeability and pathological neovascularization in DR and age-related macular degeneration (AMD). Therefore, targeting BMP signaling in diabetes is proposed as a potential therapeutic strategy to halt the development of microvascular dysfunction in retinal diseases, particularly in DR. The goal of this review article is to discuss the biological functions of BMPs, their underlying mechanisms and their potential role in the pathogenesis of DR in particular.
Collapse
Affiliation(s)
- Khaled Elmasry
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Culver Vision discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Dakahlia Governorate 35516, Egypt
| | - Samar Habib
- Department of Medical Parasitology, Mansoura Faculty of Medicine, Mansoura University, Dakahlia Governorate 35516, Egypt;
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mohamed Moustafa
- Culver Vision discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mohamed Al-Shabrawey
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Culver Vision discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-(706)721-4278 or +1-(706)721-4279
| |
Collapse
|
47
|
Yao LL, Hu JX, Li Q, Lee D, Ren X, Zhang JS, Sun D, Zhang HS, Wang YG, Mei L, Xiong WC. Astrocytic neogenin/netrin-1 pathway promotes blood vessel homeostasis and function in mouse cortex. J Clin Invest 2021; 130:6490-6509. [PMID: 32853179 DOI: 10.1172/jci132372] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Astrocytes have multiple functions in the brain, including affecting blood vessel (BV) homeostasis and function. However, the underlying mechanisms remain elusive. Here, we provide evidence that astrocytic neogenin (NEO1), a member of deleted in colorectal cancer (DCC) family netrin receptors, is involved in blood vessel homeostasis and function. Mice with Neo1 depletion in astrocytes exhibited clustered astrocyte distribution and increased BVs in their cortices. These BVs were leaky, with reduced blood flow, disrupted vascular basement membranes (vBMs), decreased pericytes, impaired endothelial cell (EC) barrier, and elevated tip EC proliferation. Increased proliferation was also detected in cultured ECs exposed to the conditioned medium (CM) of NEO1-depleted astrocytes. Further screening for angiogenetic factors in the CM identified netrin-1 (NTN1), whose expression was decreased in NEO1-depleted cortical astrocytes. Adding NTN1 into the CM of NEO1-depleted astrocytes attenuated EC proliferation. Expressing NTN1 in NEO1 mutant cortical astrocytes ameliorated phenotypes in blood-brain barrier (BBB), EC, and astrocyte distribution. NTN1 depletion in astrocytes resulted in BV/BBB deficits in the cortex similar to those in Neo1 mutant mice. In aggregate, these results uncovered an unrecognized pathway, astrocytic NEO1 to NTN1, not only regulating astrocyte distribution, but also promoting cortical BV homeostasis and function.
Collapse
Affiliation(s)
- Ling-Ling Yao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jin-Xia Hu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Institute of Stroke Center and Department of Neurology, Xuzhou Medical University, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
| | - Qiang Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xiao Ren
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun-Shi Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neurology, Huaihe Hospital, Henan University, Kaifeng, Henan, China
| | - Dong Sun
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hong-Sheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yong-Gang Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
48
|
Tang YC, Gottlieb A. Explainable drug sensitivity prediction through cancer pathway enrichment. Sci Rep 2021; 11:3128. [PMID: 33542382 PMCID: PMC7862690 DOI: 10.1038/s41598-021-82612-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Computational approaches to predict drug sensitivity can promote precision anticancer therapeutics. Generalizable and explainable models are of critical importance for translation to guide personalized treatment and are often overlooked in favor of prediction performance. Here, we propose PathDSP: a pathway-based model for drug sensitivity prediction that integrates chemical structure information with enrichment of cancer signaling pathways across drug-associated genes, gene expression, mutation and copy number variation data to predict drug response on the Genomics of Drug Sensitivity in Cancer dataset. Using a deep neural network, we outperform state-of-the-art deep learning models, while demonstrating good generalizability a separate dataset of the Cancer Cell Line Encyclopedia as well as provide explainable results, demonstrated through case studies that are in line with current knowledge. Additionally, our pathway-based model achieved a good performance when predicting unseen drugs and cells, with potential utility for drug development and for guiding individualized medicine.
Collapse
Affiliation(s)
- Yi-Ching Tang
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Assaf Gottlieb
- Center for Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
49
|
Seeherman HJ, Li XJ, Wozney JM. Activation of Bone Remodeling Compartments in BMP-2-Injected Knees Supports a Local Vascular Mechanism for Arthritis-Related Bone Changes. J Bone Joint Surg Am 2021; 103:e8. [PMID: 33315697 DOI: 10.2106/jbjs.20.00883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Synovial membrane-derived factors are implicated in arthritis-related bone changes. The route that synovial factors use to access subchondral bone and the mechanisms responsible for these bone changes remain unclear. A safety study involving intra-articular injection of bone morphogenetic protein-2 (BMP-2)/calcium phosphate matrix (CPM) or CPM addresses these issues. METHODS Knee joints in 21 monkeys were injected with CPM or 1.5 or 4.5 mg/mL BMP-2/CPM and were evaluated at 1 and 8 weeks. Contralateral joints were injected with saline solution. Knee joints in 4 animals each were injected with 1.5 or 4.5 mg/mL BMP-2/CPM. Contralateral joints were injected with corresponding treatments at 8 weeks. Both joints were evaluated at 16 weeks. Harvested joints were evaluated grossly and with histomorphometry. Knee joints in 3 animals were injected with 125I-labeled BMP-2/CPM and evaluated with scintigraphy and autoradiography at 2 weeks to determine BMP-2 distribution. RESULTS All treatments induced transient synovitis and increased capsular vascularization, observed to anastomose with metaphyseal venous sinusoids, but did not damage articular cartilage. Both treatments induced unanticipated activation of vascular-associated trabecular bone remodeling compartments (BRCs) restricted to injected knees. Bone volume increased in BMP-2/CPM-injected knees at 8 and 16 weeks. Scintigraphy demonstrated metaphyseal 125I-labeled BMP-2 localization restricted to injected knees, confirming local rather than systemic BMP-2 release. Autoradiography demonstrated that BMP-2 diffusion through articular cartilage into the metaphysis was blocked by the tidemark. The lack of marrow activation or de novo bone formation, previously reported following metaphyseal BMP-2/CPM administration, confirmed BMP-2 and synovial-derived factors were not free in the marrow. The 125I-labeled BMP-2/CPM, observed within venous sinusoids of injected knees, confirmed the potential for capsular and metaphyseal venous portal communication. CONCLUSIONS This study identifies a synovitis-induced venous portal circulation between the joint capsule and the metaphysis as an alternative to systemic circulation and local diffusion for synovial membrane-derived factors to reach subchondral bone. This study also identifies vascular-associated BRCs as a mechanism for arthritis-associated subchondral bone changes and provides additional support for their role in physiological trabecular bone remodeling and/or modeling. CLINICAL RELEVANCE Inhibition of synovitis and accompanying abnormal vascularization may limit bone changes associated with arthritis.
Collapse
Affiliation(s)
- Howard J Seeherman
- Orthopedic Research/Pharmaceutical Development Consultant, Cambridge, Massachusetts
| | - X Jian Li
- CBSET, Inc., Lexington, Massachusetts
| | - John M Wozney
- Orthopedics and Pharmaceutical Development Consultant, Hudson, Massachusetts
| |
Collapse
|
50
|
Pulkkinen HH, Kiema M, Lappalainen JP, Toropainen A, Beter M, Tirronen A, Holappa L, Niskanen H, Kaikkonen MU, Ylä-Herttuala S, Laakkonen JP. BMP6/TAZ-Hippo signaling modulates angiogenesis and endothelial cell response to VEGF. Angiogenesis 2021; 24:129-144. [PMID: 33021694 PMCID: PMC7921060 DOI: 10.1007/s10456-020-09748-4] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
The BMP/TGFβ-Smad, Notch and VEGF signaling guides formation of endothelial tip and stalk cells. However, the crosstalk of bone morphogenetic proteins (BMPs) and vascular endothelial growth factor receptor 2 (VEGFR2) signaling has remained largely unknown. We demonstrate that BMP family members regulate VEGFR2 and Notch signaling, and act via TAZ-Hippo signaling pathway. BMPs were found to be regulated after VEGF gene transfer in C57/Bl6 mice and in a porcine myocardial ischemia model. BMPs 2/4/6 were identified as endothelium-specific targets of VEGF. BMP2 modulated VEGF-mediated endothelial sprouting via Delta like Canonical Notch Ligand 4 (DLL4). BMP6 modulated VEGF signaling by regulating VEGFR2 expression and acted via Hippo signaling effector TAZ, known to regulate cell survival/proliferation, and to be dysregulated in cancer. In a matrigel plug assay in nude mice BMP6 was further demonstrated to induce angiogenesis. BMP6 is the first member of BMP family found to directly regulate both Hippo signaling and neovessel formation. It may thus serve as a target in pro/anti-angiogenic therapies.
Collapse
Affiliation(s)
- H H Pulkkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M Kiema
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J P Lappalainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Chemistry, University of Eastern Finland and Eastern Finland Laboratory Centre, Kuopio, Finland
| | - A Toropainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M Beter
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - A Tirronen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - L Holappa
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - H Niskanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Science Service Center, Kuopio University Hospital, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Johanna P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|