1
|
Ng KW, Chaturvedi N, Coté GL, Fisher SA, Mabbott S. Biomarkers and point of care screening approaches for the management of preeclampsia. COMMUNICATIONS MEDICINE 2024; 4:208. [PMID: 39433973 PMCID: PMC11493996 DOI: 10.1038/s43856-024-00642-4] [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: 05/22/2023] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Preeclampsia is a multi-organ pregnancy complication, that is primarily detected when pregnant people have high blood pressure, and is confirmed by testing for the presence of protein in the urine. While more specific and accurate diagnostic and imaging tests are becoming available, they are still in the process of undergoing widespread regulatory adoption, and so are not yet the standard of care. Since biochemical processes are a precursor to the systemic progression of disease, we review some established, emerging, and promising biomarkers that are proposed to be associated with preeclampsia, and newly developed approaches for screening them at the point of care, to reduce the burden of the disease.
Collapse
Affiliation(s)
- Ka Wai Ng
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA
| | - Nandita Chaturvedi
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA
| | - Gerard L Coté
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA
| | - Stephanie A Fisher
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Samuel Mabbott
- Department of Biomedical Engineering, Texas A&M University, 600 Discovery Drive, College Station, TX, 77840-3006, USA.
| |
Collapse
|
2
|
Chen H, Li YY, Nio K, Tang H. Unveiling the Impact of BMP9 in Liver Diseases: Insights into Pathogenesis and Therapeutic Potential. Biomolecules 2024; 14:1013. [PMID: 39199400 PMCID: PMC11353080 DOI: 10.3390/biom14081013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Bone morphogenetic proteins (BMPs) are a group of growth factors belonging to the transforming growth factor β(TGF-β) family. While initially recognized for their role in bone formation, BMPs have emerged as significant players in liver diseases. Among BMPs with various physiological activities, this comprehensive review aims to delve into the involvement of BMP9 specifically in liver diseases and provide insights into the complex BMP signaling pathway. Through an enhanced understanding of BMP9, we anticipate the discovery of new therapeutic options and potential strategies for managing liver diseases.
Collapse
Affiliation(s)
- Han Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China;
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ying-Yi Li
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa 9208641, Japan;
| | - Kouki Nio
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa 9208641, Japan;
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China;
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Li D, Zheng M, Zhang W, He J, Zhang L, Tan Q, Maghsoudloo M, Liu K, Li T, Yao R, Wei C, Cheng J, Fu J. Evaluation of ENG/CD105 expression, methylation, immuno-response, and cordycepin (CD) regulation as a novel biomarker of breast invasive carcinoma (BRCA). J Cancer 2024; 15:5244-5257. [PMID: 39247590 PMCID: PMC11375534 DOI: 10.7150/jca.98767] [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: 05/22/2024] [Accepted: 07/30/2024] [Indexed: 09/10/2024] Open
Abstract
ENG/CD105 encodes a vascular endothelial glycoprotein and plays a crucial role in modulating angiogenesis. However, the significance of ENG expression, DNA methylation, immuno-response, and cordycepin (CD) regulation as diagnostic, prognostic, and therapeutic markers for breast invasive carcinoma (BRCA) remains unclear. As a result, ENG is decreased in BRCA tissues compared with corresponding healthy tissues. Five isoforms were found, and the utilization for ENG isoform (ENG-002) was the highest, suggesting its potential involvement in important roles in BRCA. ENG DNA was frequently altered in most types of cancer, and overall survival (OS) for mutant ENG was significantly longer than for wild-type cases. High expressions of ENG remarkably correlate with long relapse-free survival (RFS) for breast cancer (BC). Additionally, the ENG methylation level was higher in BRCA tissues compared with matched healthy tissues. The ENG expression and DNA methylation showed a significantly reverse correlation, demonstrating that ENG methylation may be a regulatory mechanism. By constructing diagnostic and prognostic models of ENG methylation for BRCA, we found four CpGs (CpG sites) that ranked with high importance. High methylation for cg14185922 of ENG in BRCA tissues showed shorter OS (high risk), indicating that ENG CpGs' methylation has potential as a diagnostic and prognostic biomarker for BRCA. Moreover, ENG might be a novel target for tumor immune response and immunotherapy in pancancer, including BC. CD, an adenosine analog and anti-cancer agent, increased ENG levels in a dose-dependent manner in animal models. This suggests that CD repressed BC growth and metastasis, at least partially through increasing the expression of the tumor suppressor gene ENG. Thus, our study successfully evaluated ENG/CD105 expression, DNA methylation, immune response, and CD regulation, which act as a novel diagnostic, prognostic, and therapeutic biomarker for BRCA. This research also fills critical knowledge gaps in this ENG/cancer field and highlights ENG's potential importance for the diagnosis, prognosis, and treatment of BRCA.
Collapse
Affiliation(s)
- Dabing Li
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Meiling Zheng
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Wenqian Zhang
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- Department of Rehabilitation Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Jiayue He
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lianmei Zhang
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- Department of Pathology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu Province, China
| | - Qi Tan
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Kemeng Liu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- Department of Infection Management, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ting Li
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ranbin Yao
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Chunli Wei
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
4
|
Horvat Mercnik M, Schliefsteiner C, Sanchez-Duffhues G, Wadsack C. TGFβ signalling: a nexus between inflammation, placental health and preeclampsia throughout pregnancy. Hum Reprod Update 2024; 30:442-471. [PMID: 38519450 PMCID: PMC11215164 DOI: 10.1093/humupd/dmae007] [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: 10/18/2023] [Revised: 02/16/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The placenta is a unique and pivotal organ in reproduction, controlling crucial growth and cell differentiation processes that ensure a successful pregnancy. Placental development is a tightly regulated and dynamic process, in which the transforming growth factor beta (TGFβ) superfamily plays a central role. This family of pleiotropic growth factors is heavily involved in regulating various aspects of reproductive biology, particularly in trophoblast differentiation during the first trimester of pregnancy. TGFβ signalling precisely regulates trophoblast invasion and the cell transition from cytotrophoblasts to extravillous trophoblasts, which is an epithelial-to-mesenchymal transition-like process. Later in pregnancy, TGFβ signalling ensures proper vascularization and angiogenesis in placental endothelial cells. Beyond its role in trophoblasts and endothelial cells, TGFβ signalling contributes to the polarization and function of placental and decidual macrophages by promoting maternal tolerance of the semi-allogeneic foetus. Disturbances in early placental development have been associated with several pregnancy complications, including preeclampsia (PE) which is one of the severe complications. Emerging evidence suggests that TGFβ is involved in the pathogenesis of PE, thereby offering a potential target for intervention in the human placenta. OBJECTIVE AND RATIONALE This comprehensive review aims to explore and elucidate the roles of the major members of the TGFβ superfamily, including TGFβs, bone morphogenetic proteins (BMPs), activins, inhibins, nodals, and growth differentiation factors (GDFs), in the context of placental development and function. The review focusses on their interactions within the major cell types of the placenta, namely trophoblasts, endothelial cells, and immune cells, in both normal pregnancies and pregnancies complicated by PE throughout pregnancy. SEARCH METHODS A literature search was carried out using PubMed and Google Scholar, searching terms: 'TGF signalling preeclampsia', 'pregnancy TGF signalling', 'preeclampsia tgfβ', 'preeclampsia bmp', 'preeclampsia gdf', 'preeclampsia activin', 'endoglin preeclampsia', 'endoglin pregnancy', 'tgfβ signalling pregnancy', 'bmp signalling pregnancy', 'gdf signalling pregnancy', 'activin signalling pregnancy', 'Hofbauer cell tgfβ signalling', 'placental macrophages tgfβ', 'endothelial cells tgfβ', 'endothelium tgfβ signalling', 'trophoblast invasion tgfβ signalling', 'trophoblast invasion Smad', 'trophoblast invasion bmp', 'trophoblast invasion tgfβ', 'tgfβ preeclampsia', 'tgfβ placental development', 'TGFβ placental function', 'endothelial dysfunction preeclampsia tgfβ signalling', 'vascular remodelling placenta TGFβ', 'inflammation pregnancy tgfβ', 'immune response pregnancy tgfβ', 'immune tolerance pregnancy tgfβ', 'TGFβ pregnancy NK cells', 'bmp pregnancy NK cells', 'bmp pregnancy tregs', 'tgfβ pregnancy tregs', 'TGFβ placenta NK cells', 'TGFβ placenta tregs', 'NK cells preeclampsia', 'Tregs preeclampsia'. Only articles published in English until 2023 were used. OUTCOMES A comprehensive understanding of TGFβ signalling and its role in regulating interconnected cell functions of the main placental cell types provides valuable insights into the processes essential for successful placental development and growth of the foetus during pregnancy. By orchestrating trophoblast invasion, vascularization, immune tolerance, and tissue remodelling, TGFβ ligands contribute to the proper functioning of a healthy maternal-foetal interface. However, dysregulation of TGFβ signalling has been implicated in the pathogenesis of PE, where the shallow trophoblast invasion, defective vascular remodelling, decreased uteroplacental perfusion, and endothelial cell and immune dysfunction observed in PE, are all affected by an altered TGFβ signalling. WIDER IMPLICATIONS The dysregulation of TGFβ signalling in PE has important implications for research and clinical practice. Further investigation is required to understand the underlying mechanisms, including the role of different ligands and their regulation under pathophysiological conditions, in order to discover new therapeutic targets. Distinguishing between clinically manifested subtypes of PE and studying TGFβ signalling in different placental cell types holistically is an important first step. To put this knowledge into practice, pre-clinical animal models combined with new technologies are needed. This may also lead to improved human research models and identify potential therapeutic targets, ultimately improving outcomes for affected pregnancies and reducing the burden of PE.
Collapse
Affiliation(s)
| | | | - Gonzalo Sanchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Tissue-Specific BMP Signalling ISPA-HUCA, Oviedo, Spain
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
5
|
Li W, Quigley K. Bone morphogenetic protein signalling in pulmonary arterial hypertension: revisiting the BMPRII connection. Biochem Soc Trans 2024; 52:1515-1528. [PMID: 38716930 PMCID: PMC11346422 DOI: 10.1042/bst20231547] [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: 03/04/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and life-threatening vascular disorder, characterised by abnormal remodelling of the pulmonary vessels and elevated pulmonary artery pressure, leading to right ventricular hypertrophy and right-sided heart failure. The importance of bone morphogenetic protein (BMP) signalling in the pathogenesis of PAH is demonstrated by human genetic studies. Many PAH risk genes are involved in the BMP signalling pathway and are highly expressed or preferentially act on vascular endothelial cells. Endothelial dysfunction is recognised as an initial trigger for PAH, and endothelial BMP signalling plays a crucial role in the maintenance of endothelial integrity. BMPR2 is the most prevalent PAH gene, found in over 80% of heritable cases. As BMPRII protein is the major type II receptor for a large family of BMP ligands and expressed ubiquitously in many tissues, dysregulated BMP signalling in other cells may also contribute to PAH pathobiology. Sotatercept, which contains the extracellular domain of another transforming growth factor-β family type II receptor ActRIIA fused to immunoglobin Fc domain, was recently approved by the FDA as a treatment for PAH. Neither its target cells nor its mechanism of action is fully understood. This review will revisit BMPRII function and its extracellular regulation, summarise how dysregulated BMP signalling in endothelial cells and smooth muscle cells may contribute to PAH pathogenesis, and discuss how novel therapeutics targeting the extracellular regulation of BMP signalling, such as BMP9 and Sotatercept, can be related to restoring BMPRII function.
Collapse
Affiliation(s)
- Wei Li
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| | - Kate Quigley
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| |
Collapse
|
6
|
Zhao Y, Wu X, Yang Y, Zhang L, Cai X, Chen S, Vera A, Ji J, Boström KI, Yao Y. Inhibition of endothelial histone deacetylase 2 shifts endothelial-mesenchymal transitions in cerebral arteriovenous malformation models. J Clin Invest 2024; 134:e176758. [PMID: 38781032 PMCID: PMC11290970 DOI: 10.1172/jci176758] [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: 10/18/2023] [Accepted: 05/21/2024] [Indexed: 05/25/2024] Open
Abstract
Cerebral arteriovenous malformations (AVMs) are the most common vascular malformations worldwide and the leading cause of hemorrhagic strokes that may result in crippling neurological deficits. Here, using recently generated mouse models, we uncovered that cerebral endothelial cells (ECs) acquired mesenchymal markers and caused vascular malformations. Interestingly, we found that limiting endothelial histone deacetylase 2 (HDAC2) prevented cerebral ECs from undergoing mesenchymal differentiation and reduced cerebral AVMs. We found that endothelial expression of HDAC2 and enhancer of zeste homolog 1 (EZH1) was altered in cerebral AVMs. These alterations changed the abundance of H4K8ac and H3K27me in the genes regulating endothelial and mesenchymal differentiation, which caused the ECs to acquire mesenchymal characteristics and form AVMs. This investigation demonstrated that the induction of HDAC2 altered specific histone modifications, which resulted in mesenchymal characteristics in the ECs and cerebral AVMs. The results provide insight into the epigenetic impact on AVMs.
Collapse
Affiliation(s)
- Yan Zhao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yang Yang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Sydney Chen
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Abigail Vera
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jaden Ji
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kristina I. Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- The Molecular Biology Institute, UCLA, Los Angeles, California, USA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
7
|
Koh H, Kang W, Mao YY, Park J, Kim S, Hong SH, Lee JH. Employment of diverse in vitro systems for analyzing multiple aspects of disease, hereditary hemorrhagic telangiectasia (HHT). Cell Biosci 2024; 14:65. [PMID: 38778363 PMCID: PMC11110195 DOI: 10.1186/s13578-024-01247-z] [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: 02/10/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND In vitro disease modeling enables translational research by providing insight into disease pathophysiology and molecular mechanisms, leading to the development of novel therapeutics. Nevertheless, in vitro systems have limitations for recapitulating the complexity of tissues, and a single model system is insufficient to gain a comprehensive understanding of a disease. RESULTS Here we explored the potential of using several models in combination to provide mechanistic insight into hereditary hemorrhagic telangiectasia (HHT), a genetic vascular disorder. Genome editing was performed to establish hPSCs (H9) with ENG haploinsufficiency and several in vitro models were used to recapitulate the functional aspects of the cells that constitute blood vessels. In a 2D culture system, endothelial cells showed early senescence, reduced viability, and heightened susceptibility to apoptotic insults, and smooth muscle cells (SMCs) exhibited similar behavior to their wild-type counterparts. Features of HHT were evident in 3D blood-vessel organoid systems, including thickening of capillary structures, decreased interaction between ECs and surrounding SMCs, and reduced cell viability. Features of ENG haploinsufficiency were observed in arterial and venous EC subtypes, with arterial ECs showing significant impairments. Molecular biological approaches confirmed the significant downregulation of Notch signaling in HHT-ECs. CONCLUSIONS Overall, we demonstrated refined research strategies to enhance our comprehension of HHT, providing valuable insights for pathogenic analysis and the exploration of innovative therapeutic interventions. Additionally, these results underscore the importance of employing diverse in vitro systems to assess multiple aspects of disease, which is challenging using a single in vitro system.
Collapse
Affiliation(s)
- Hyebin Koh
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Woojoo Kang
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Ying-Ying Mao
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Daejeon, Republic of Korea
| | - Jisoo Park
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Republic of Korea
| | - Sangjune Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea.
- KW-Bio Co., Ltd, Chuncheon, South Korea.
| | - Jong-Hee Lee
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea.
| |
Collapse
|
8
|
Redgrave RE, Singh E, Tual-Chalot S, Park C, Hall D, Bennaceur K, Smyth DJ, Maizels RM, Spyridopoulos I, Arthur HM. Exogenous Transforming Growth Factor-β1 and Its Helminth-Derived Mimic Attenuate the Heart's Inflammatory Response to Ischemic Injury and Reduce Mature Scar Size. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:562-573. [PMID: 37832870 DOI: 10.1016/j.ajpath.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/29/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023]
Abstract
Coronary reperfusion after acute ST-elevation myocardial infarction (STEMI) is standard therapy to salvage ischemic heart muscle. However, subsequent inflammatory responses within the infarct lead to further loss of viable myocardium. Transforming growth factor (TGF)-β1 is a potent anti-inflammatory cytokine released in response to tissue injury. The aim of this study was to investigate the protective effects of TGF-β1 after MI. In patients with STEMI, there was a significant correlation (P = 0.003) between higher circulating TGF-β1 levels at 24 hours after MI and a reduction in infarct size after 3 months, suggesting a protective role of early increase in circulating TGF-β1. A mouse model of cardiac ischemia reperfusion was used to demonstrate multiple benefits of exogenous TGF-β1 delivered in the acute phase. It led to a significantly smaller infarct size (30% reduction, P = 0.025), reduced inflammatory infiltrate (28% reduction, P = 0.015), lower intracardiac expression of inflammatory cytokines IL-1β and chemokine (C-C motif) ligand 2 (>50% reduction, P = 0.038 and 0.0004, respectively) at 24 hours, and reduced scar size at 4 weeks (21% reduction, P = 0.015) after reperfusion. Furthermore, a low-fibrogenic mimic of TGF-β1, secreted by the helminth parasite Heligmosomoides polygyrus, had an almost identical protective effect on injured mouse hearts. Finally, genetic studies indicated that this benefit was mediated by TGF-β signaling in the vascular endothelium.
Collapse
Affiliation(s)
- Rachael E Redgrave
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Esha Singh
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Simon Tual-Chalot
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Catherine Park
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Darroch Hall
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Karim Bennaceur
- Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Ioakim Spyridopoulos
- Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Helen M Arthur
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom.
| |
Collapse
|
9
|
Sun C, Xie K, Yang L, Cai S, Wang M, Zhu Y, Tao B, Zhu Y. HDAC6 Enhances Endoglin Expression through Deacetylation of Transcription Factor SP1, Potentiating BMP9-Induced Angiogenesis. Cells 2024; 13:490. [PMID: 38534334 DOI: 10.3390/cells13060490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Histone deacetylase 6 (HDAC6) plays a crucial role in the acetylation of non-histone proteins and is notably implicated in angiogenesis, though its underlying mechanisms were previously not fully understood. This study conducted transcriptomic and proteomic analyses on vascular endothelial cells with HDAC6 knockdown, identifying endoglin (ENG) as a key downstream protein regulated by HDAC6. This protein is vital for maintaining vascular integrity and plays a complex role in angiogenesis, particularly in its interaction with bone morphogenetic protein 9 (BMP9). In experiments using human umbilical vein endothelial cells (HUVECs), the pro-angiogenic effects of BMP9 were observed, which diminished following the knockdown of HDAC6 and ENG. Western blot analysis revealed that BMP9 treatment increased SMAD1/5/9 phosphorylation, a process hindered by HDAC6 knockdown, correlating with reduced ENG expression. Mechanistically, our study indicates that HDAC6 modulates ENG transcription by influencing promoter activity, leading to increased acetylation of transcription factor SP1 and consequently altering its transcriptional activity. Additionally, the study delves into the structural role of HDAC6, particularly its CD2 domain, in regulating SP1 acetylation and subsequently ENG expression. In conclusion, the present study underscores the critical function of HDAC6 in modulating SP1 acetylation and ENG expression, thereby significantly affecting BMP9-mediated angiogenesis. This finding highlights the potential of HDAC6 as a therapeutic target in angiogenesis-related processes.
Collapse
Affiliation(s)
- Chen Sun
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Avenida WaiLong, Taipa, Macau 999078, China
| | - Kuifang Xie
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Lejie Yang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Shengyang Cai
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Mingjie Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yizhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Avenida WaiLong, Taipa, Macau 999078, China
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 200433, China
| | - Beibei Tao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Yichun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University Shanghai Medical College, Shanghai 200032, China
| |
Collapse
|
10
|
Liu K, Fu J, Guo K, Maghsoudloo M, Cheng J, Fu J. The ENG/VEGFα Pathway Is Likely Affected by a Nonsense Variant of Endoglin (ENG)/CD105, Causing Hereditary Hemorrhagic Telangiectasia Type 1 (HHT1) in a Chinese Family. Genes (Basel) 2024; 15:304. [PMID: 38540362 PMCID: PMC10970080 DOI: 10.3390/genes15030304] [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: 02/02/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 06/14/2024] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT), also called Rendu-Osler syndrome, is a group of rare genetic diseases characterized by autosomal dominance, multisystemic vascular dysplasia, and age-related penetrance. This includes arteriovenous malformations (AVMs) in the skin, brain, lung, liver, and mucous membranes. The correlations between the phenotype and genotype for HHT are not clear. An HHT Chinese pedigree was recruited. Whole exome sequencing (WES) analysis, Sanger verification, and co-segregation were conducted. Western blotting was performed for monitoring ENG/VEGFα signaling. As a result, a nonsense, heterozygous variant for ENG/CD105: c.G1169A:p. Trp390Ter of the proband with hereditary hemorrhagic telangiectasia type 1 (HHT1) was identified, which co-segregated with the disease in the M666 pedigree. Western blotting found that, compared with the normal levels associated with non-carrier family members, the ENG protein levels in the proband showed approximately a one-half decrease (47.4% decrease), while levels of the VEGFα protein, in the proband, showed approximately a one-quarter decrease (25.6% decrease), implying that ENG haploinsufficiency, displayed in the carrier of this variant, may affect VEGFα expression downregulation. Pearson and Spearman correlation analyses further supported TGFβ/ENG/VEGFα signaling, implying ENG regulation in the blood vessels. Thus, next-generation sequencing including WES should provide an accurate strategy for gene diagnosis, therapy, genetic counseling, and clinical management for rare genetic diseases including that in HHT1 patients.
Collapse
Affiliation(s)
- Kemeng Liu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China; (K.L.); (J.F.); (K.G.); (M.M.); (J.C.)
| | - Jiewen Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China; (K.L.); (J.F.); (K.G.); (M.M.); (J.C.)
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Kan Guo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China; (K.L.); (J.F.); (K.G.); (M.M.); (J.C.)
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China; (K.L.); (J.F.); (K.G.); (M.M.); (J.C.)
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China; (K.L.); (J.F.); (K.G.); (M.M.); (J.C.)
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China; (K.L.); (J.F.); (K.G.); (M.M.); (J.C.)
| |
Collapse
|
11
|
Al Tabosh T, Al Tarrass M, Tourvieilhe L, Guilhem A, Dupuis-Girod S, Bailly S. Hereditary hemorrhagic telangiectasia: from signaling insights to therapeutic advances. J Clin Invest 2024; 134:e176379. [PMID: 38357927 PMCID: PMC10866657 DOI: 10.1172/jci176379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Hereditary hemorrhagic telangiectsia (HHT) is an inherited vascular disorder with highly variable expressivity, affecting up to 1 in 5,000 individuals. This disease is characterized by small arteriovenous malformations (AVMs) in mucocutaneous areas (telangiectases) and larger visceral AVMs in the lungs, liver, and brain. HHT is caused by loss-of-function mutations in the BMP9-10/ENG/ALK1/SMAD4 signaling pathway. This Review presents up-to-date insights on this mutated signaling pathway and its crosstalk with proangiogenic pathways, in particular the VEGF pathway, that has allowed the repurposing of new drugs for HHT treatment. However, despite the substantial benefits of these new treatments in terms of alleviating symptom severity, this not-so-uncommon bleeding disorder still currently lacks any FDA- or European Medicines Agency-approved (EMA-approved) therapies.
Collapse
Affiliation(s)
- Tala Al Tabosh
- Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France
| | - Mohammad Al Tarrass
- Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France
| | - Laura Tourvieilhe
- Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France
| | - Alexandre Guilhem
- Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France
- TAI-IT Autoimmunité Unit RIGHT-UMR1098, Burgundy University, INSERM, EFS-BFC, Besancon, France
| | - Sophie Dupuis-Girod
- Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France
- Hospices Civils de Lyon, National HHT Reference Center and Genetics Department, Femme-Mère-Enfants Hospital, Bron, France
| | - Sabine Bailly
- Biosanté Unit U1292, Grenoble Alpes University, INSERM, CEA, Grenoble, France
| |
Collapse
|
12
|
Wu M, Wu S, Chen W, Li YP. The roles and regulatory mechanisms of TGF-β and BMP signaling in bone and cartilage development, homeostasis and disease. Cell Res 2024; 34:101-123. [PMID: 38267638 PMCID: PMC10837209 DOI: 10.1038/s41422-023-00918-9] [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: 02/26/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Transforming growth factor-βs (TGF-βs) and bone morphometric proteins (BMPs) belong to the TGF-β superfamily and perform essential functions during osteoblast and chondrocyte lineage commitment and differentiation, skeletal development, and homeostasis. TGF-βs and BMPs transduce signals through SMAD-dependent and -independent pathways; specifically, they recruit different receptor heterotetramers and R-Smad complexes, resulting in unique biological readouts. BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner. BMPs and TGF-β have opposite functions in articular cartilage homeostasis. Moreover, TGF-β has a specific role in maintaining the osteocyte network. The precise activation of BMP and TGF-β signaling requires regulatory machinery at multiple levels, including latency control in the matrix, extracellular antagonists, ubiquitination and phosphorylation in the cytoplasm, nucleus-cytoplasm transportation, and transcriptional co-regulation in the nuclei. This review weaves the background information with the latest advances in the signaling facilitated by TGF-βs and BMPs, and the advanced understanding of their diverse physiological functions and regulations. This review also summarizes the human diseases and mouse models associated with disordered TGF-β and BMP signaling. A more precise understanding of the BMP and TGF-β signaling could facilitate the development of bona fide clinical applications in treating bone and cartilage disorders.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
13
|
Graham BB, Hilton JF, Lee MH, Kumar R, Balladares DF, Rahaghi FN, Estépar RSJ, Mickael C, Lima RLB, Loureiro CM, Lucena J, Oliveira RK, Corrêa RDA. Is pulmonary arterial hypertension associated with schistosomiasis distinct from pulmonary arterial hypertension associated with portal hypertension? JHLT OPEN 2023; 1:100007. [PMID: 38050478 PMCID: PMC10695267 DOI: 10.1016/j.jhlto.2023.100007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Pulmonary arterial hypertension associated with schistosomiasis (SchPAH) and pulmonary arterial hypertension associated with portal hypertension (PoPAH) are lung diseases that develop in the presence of liver diseases. However, mechanistic pathways by which the underlying liver conditions and other drivers contribute to the development and progression of pulmonary arterial hypertension (PAH) are unclear for both etiologies. In turn, these unknowns limit certainty of strategies to prevent, diagnose, and reverse the resultant PAH. Here we consider specific mechanisms that contribute to SchPAH and PoPAH, identifying those that may be shared and those that appear to be unique to each etiology, in the hope that this exploration will both highlight known causal drivers and identify knowledge gaps appropriate for future research. Overall, the key pathophysiologic differences that we identify between SchPAH and PoPAH suggest that they are not variants of a single condition.
Collapse
Affiliation(s)
- Brian B. Graham
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Joan F. Hilton
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Michael H. Lee
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Rahul Kumar
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Dara Fonseca Balladares
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Farbod N. Rahaghi
- Pulmonary Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Raúl San José Estépar
- Applied Chest Imaging Laboratory, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Claudia Mickael
- Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | | | - Juliana Lucena
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Rudolf K.F. Oliveira
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ricardo de Amorim Corrêa
- Internal Medicine/Pulmonary Division, Medical School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
14
|
Atreya MR, Cvijanovich NZ, Fitzgerald JC, Weiss SL, Bigham MT, Jain PN, Schwarz AJ, Lutfi R, Nowak J, Thomas NJ, Quasney M, Haileselassie B, Baines TD, Zingarelli B. SERUM SOLUBLE ENDOGLIN IN PEDIATRIC SEPTIC SHOCK-ASSOCIATED MULTIPLE ORGAN DYSFUNCTION SYNDROME. Shock 2023; 60:379-384. [PMID: 37493567 PMCID: PMC10529838 DOI: 10.1097/shk.0000000000002183] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
ABSTRACT Background: Endothelial activation is a key driver of multiple organ dysfunction syndrome (MODS). Soluble endoglin (sENG) is expressed by mature and progenitor endothelial cells and thought to have angiogenic properties. We sought to determine the association between sENG and pediatric sepsis-associated MODS. Methods: Prospective observational study of pediatric septic shock. Primary outcome of interest was complicated course-a composite of death by (or) MODS on day 7 of illness. Secondary outcomes included individual organ dysfunctions. Endothelial biomarkers including sENG were measured using multiplex Luminex assays among patients with existing data on the Pediatric Sepsis Biomarker Risk Model (PERSEVERE-II) data. Multivariable regression was used to test the independent association between sENG and clinical outcomes. Serum sENG concentrations across PERSEVERE-II mortality risk strata and correlations with established markers of endothelial dysfunction were determined. Results: Three hundred six critically ill children with septic shock were included. Serum sENG concentrations were higher among those with primary and secondary outcomes of interest, with the exception of acute neurological dysfunction. Soluble endoglin was independently associated with increased odds of complicated course (adjusted odds ratio, 1.53; 95% confidence interval, 1.02-2.27; P = 0.038) and acute renal dysfunction (adjusted odds ratio, 1.84; 95% confidence interval, 1.18-2.876; P = 0.006). Soluble endoglin demonstrated graded responses across PERSEVERE-II risk strata and was positively correlated with endothelial biomarkers, except angiopoietin-1. Conclusions: Serum sENG is independently associated with complicated course and acute renal dysfunction in pediatric septic shock. Future studies are required to validate our observational data, and mechanistic studies are necessary to elucidate whether endoglin plays an organ-specific role in the development or resolution of acute renal dysfunction in sepsis.
Collapse
Affiliation(s)
- Mihir R. Atreya
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center and Cincinnati Children’s Research Foundation, Cincinnati, 45229, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | - Scott L. Weiss
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Parag N. Jain
- Texas Children’s Hospital and Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Riad Lutfi
- Riley Hospital for Children, Indianapolis, IN 46202, USA
| | - Jeffrey Nowak
- Children’s Hospital and Clinics of Minnesota, Minneapolis, MN 55404, USA
| | - Neal J. Thomas
- Penn State Hershey Children’s Hospital, Hershey, PA 17033, USA
| | - Michael Quasney
- CS Mott Children’s Hospital at the University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Torrey D. Baines
- University of Florida Health Shands Children’s Hospital, Gainesville, FL 32610, USA
| | - Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center and Cincinnati Children’s Research Foundation, Cincinnati, 45229, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | |
Collapse
|
15
|
Rossi E, Bernabeu C. Novel vascular roles of human endoglin in pathophysiology. J Thromb Haemost 2023; 21:2327-2338. [PMID: 37315795 DOI: 10.1016/j.jtha.2023.06.007] [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: 03/16/2023] [Revised: 05/19/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
Endoglin, alias CD105, is a human membrane glycoprotein highly expressed in vascular endothelial cells. It is involved in angiogenesis and angiogenesis-related diseases, including the rare vascular pathology known as hereditary hemorrhagic telangiectasia type 1. Although endoglin acts as an accessory receptor for members of the transforming growth factor-β family, in recent years, emerging evidence has shown a novel functional role for this protein beyond the transforming growth factor-β system. In fact, endoglin has been found to be an integrin counterreceptor involved in endothelial cell adhesion processes during pathological inflammatory conditions and primary hemostasis. Furthermore, a circulating form of endoglin, also named as soluble endoglin, whose levels are abnormally increased in different pathological conditions, such as preeclampsia, seems to act as an antagonist of membrane-bound endoglin and as a competitor of the fibrinogen-integrin interaction in platelet-dependent thrombus formation. These studies suggest that membrane-bound endoglin and circulating endoglin are important components involved in vascular homeostasis and hemostasis.
Collapse
Affiliation(s)
- Elisa Rossi
- Université Paris Cité, INSERM U1140, Innovative Therapies in Haemostasis, Paris, France.
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
16
|
Oladejo M, Nguyen HM, Wood L. CD105 in the progression and therapy of renal cell carcinoma. Cancer Lett 2023; 570:216327. [PMID: 37499740 DOI: 10.1016/j.canlet.2023.216327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Molecular biomarkers that interact with the vascular and immune compartments play an important role in the progression of solid malignancies. CD105, which is a component of the transforming growth factor beta (TGF β) signaling cascade, has long been studied for its role in potentiating angiogenesis in numerous cancers. In renal cell carcinoma (RCC), the role of CD105 is more complicated due to its diverse expression profile on the tumor cells, tumor vasculature, and the components of the immune system. Since its discovery, its angiogenic role has overshadowed other potential functions, especially in cancers. In this review, we aim to summarize the recent evidence and findings of the multifunctional roles of CD105 in angiogenesis and immunomodulation in the context of the various subtypes of RCC, with a specific emphasis on the clear cell RCC subtype. Since CD105 is an established biomarker and tumor antigen, we also provide an update on the preclinical and clinical applications of CD105 as a therapeutic platform in RCC.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Hong-My Nguyen
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Laurence Wood
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA.
| |
Collapse
|
17
|
Jose A, Elwing JM, Kawut SM, Pauciulo MW, Sherman KE, Nichols WC, Fallon MB, McCormack FX. Human liver single nuclear RNA sequencing implicates BMPR2, GDF15, arginine, and estrogen in portopulmonary hypertension. Commun Biol 2023; 6:826. [PMID: 37558836 PMCID: PMC10412637 DOI: 10.1038/s42003-023-05193-3] [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: 03/27/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
Portopulmonary hypertension (PoPH) is a type of pulmonary vascular disease due to portal hypertension that exhibits high morbidity and mortality. The mechanisms driving disease are unknown, and transcriptional characteristics unique to the PoPH liver remain unexplored. Here, we apply single nuclear RNA sequencing to compare cirrhotic livers from patients with and without PoPH. We identify characteristics unique to PoPH in cells surrounding the central hepatic vein, including increased growth differentiation factor signaling, enrichment of the arginine biosynthesis pathway, and differential expression of the bone morphogenic protein type II receptor and estrogen receptor type I genes. These results provide insight into the transcriptomic characteristics of the PoPH liver and mechanisms by which PoPH cellular dysfunction might contribute to pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Arun Jose
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Jean M Elwing
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kenneth E Sherman
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Francis X McCormack
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
18
|
Hakuno SK, Janson SGT, Trietsch MD, de Graaf M, de Jonge-Muller E, Crobach S, Harryvan TJ, Boonstra JJ, Dinjens WNM, Slingerland M, Hawinkels LJAC. Endoglin and squamous cell carcinomas. Front Med (Lausanne) 2023; 10:1112573. [PMID: 37396898 PMCID: PMC10313935 DOI: 10.3389/fmed.2023.1112573] [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: 11/30/2022] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Despite the fact that the role of endoglin on endothelial cells has been extensively described, its expression and biological role on (epithelial) cancer cells is still debatable. Especially its function on squamous cell carcinoma (SCC) cells is largely unknown. Therefore, we investigated SCC endoglin expression and function in three types of SCCs; head and neck (HNSCC), esophageal (ESCC) and vulvar (VSCC) cancers. Endoglin expression was evaluated in tumor specimens and 14 patient-derived cell lines. Next to being expressed on angiogenic endothelial cells, endoglin is selectively expressed by individual SCC cells in tumor nests. Patient derived HNSCC, ESCC and VSCC cell lines express varying levels of endoglin with high interpatient variation. To assess the function of endoglin in signaling of TGF-β ligands, endoglin was overexpressed or knocked out or the signaling was blocked using TRC105, an endoglin neutralizing antibody. The endoglin ligand BMP-9 induced strong phosphorylation of SMAD1 independent of expression of the type-I receptor ALK1. Interestingly, we observed that endoglin overexpression leads to strongly increased soluble endoglin levels, which in turn decreases BMP-9 signaling. On the functional level, endoglin, both in a ligand dependent and independent manner, did not influence proliferation or migration of the SCC cells. In conclusion, these data show endoglin expression on individual cells in the tumor nests in SCCs and a role for (soluble) endoglin in paracrine signaling, without directly affecting proliferation or migration in an autocrine manner.
Collapse
Affiliation(s)
- Sarah K. Hakuno
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
| | - Stefanus G. T. Janson
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
| | - Marjolijn D. Trietsch
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
- Department of Gynecology, Leiden University Medical Center, Leiden, Netherlands
| | - Manon de Graaf
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Eveline de Jonge-Muller
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
| | - Stijn Crobach
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Tom J. Harryvan
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
| | - Jurjen J. Boonstra
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
| | - Winand N. M. Dinjens
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
| | - Marije Slingerland
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Lukas J. A. C. Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
19
|
Andersson-Rusch C, Liu B, Quist-Løkken I, Upton PD, Olsen OE, Hella H, Yang X, Tong Z, Morrell NW, Holien T, Li W. High concentrations of soluble endoglin can inhibit BMP9 signaling in non-endothelial cells. Sci Rep 2023; 13:6639. [PMID: 37095146 PMCID: PMC10126157 DOI: 10.1038/s41598-023-33352-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
Endoglin (ENG) is a single-pass transmembrane protein highly expressed on vascular endothelial cells, although low expression levels can be detected in many other cell types. Its extracellular domain can be found in circulation known as soluble endoglin (sENG). Levels of sENG are elevated in many pathological conditions, in particular preeclampsia. We have shown that while loss of cell surface ENG decreases BMP9 signaling in endothelial cells, knocking down ENG in blood cancer cells enhances BMP9 signaling. Despite sENG binding to BMP9 with high affinity and blocking the type II receptor binding site on BMP9, sENG did not inhibit BMP9 signaling in vascular endothelial cells, but the dimeric form of sENG inhibited BMP9 signaling in blood cancer cells. Here we report that in non-endothelial cells such as human multiple myeloma cell lines and the mouse myoblast cell line C2C12, both monomeric and dimeric forms of sENG inhibit BMP9 signaling when present at high concentrations. Such inhibition can be alleviated by the overexpression of ENG and ACVRL1 (encoding ALK1) in the non-endothelial cells. Our findings suggest that the effects of sENG on BMP9 signaling is cell-type specific. This is an important consideration when developing therapies targeting the ENG and ALK1 pathway.
Collapse
Affiliation(s)
- Clara Andersson-Rusch
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
- Department of Hematology, St. Olav's University Hospital, Trondheim, Norway
| | - Bin Liu
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Ingrid Quist-Løkken
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Paul D Upton
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Oddrun Elise Olsen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Hanne Hella
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Xudong Yang
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Zhen Tong
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Nicholas W Morrell
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Toril Holien
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway.
- Department of Hematology, St. Olav's University Hospital, Trondheim, Norway.
- Department of Biomedical Laboratory Science, NTNU, Trondheim, Norway.
- Department of Immunology and Transfusion Medicine, St. Olav's University Hospital, Trondheim, Norway.
| | - Wei Li
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| |
Collapse
|
20
|
Arthur HM, Roman BL. An update on preclinical models of hereditary haemorrhagic telangiectasia: Insights into disease mechanisms. Front Med (Lausanne) 2022; 9:973964. [PMID: 36250069 PMCID: PMC9556665 DOI: 10.3389/fmed.2022.973964] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Endoglin (ENG) is expressed on the surface of endothelial cells (ECs) where it efficiently binds circulating BMP9 and BMP10 ligands to initiate activin A receptor like type 1 (ALK1) protein signalling to protect the vascular architecture. Patients heterozygous for ENG or ALK1 mutations develop the vascular disorder known as hereditary haemorrhagic telangiectasia (HHT). Many patients with this disorder suffer from anaemia, and are also at increased risk of stroke and high output heart failure. Recent work using animal models of HHT has revealed new insights into cellular and molecular mechanisms causing this disease. Loss of the ENG (HHT1) or ALK1 (HHT2) gene in ECs leads to aberrant arteriovenous connections or malformations (AVMs) in developing blood vessels. Similar phenotypes develop following combined EC specific loss of SMAD1 and 5, or EC loss of SMAD4. Taken together these data point to the essential role of the BMP9/10-ENG-ALK1-SMAD1/5-SMAD4 pathway in protecting the vasculature from AVMs. Altered directional migration of ECs in response to shear stress and increased EC proliferation are now recognised as critical factors driving AVM formation. Disruption of the ENG/ALK1 signalling pathway also affects EC responses to vascular endothelial growth factor (VEGF) and crosstalk between ECs and vascular smooth muscle cells. It is striking that the vascular lesions in HHT are both localised and tissue specific. Increasing evidence points to the importance of a second genetic hit to generate biallelic mutations, and the sporadic nature of such somatic mutations would explain the localised formation of vascular lesions. In addition, different pro-angiogenic drivers of AVM formation are likely to be at play during the patient’s life course. For example, inflammation is a key driver of vessel remodelling in postnatal life, and may turn out to be an important driver of HHT disease. The current wealth of preclinical models of HHT has led to increased understanding of AVM development and revealed new therapeutic approaches to treat AVMs, and form the topic of this review.
Collapse
Affiliation(s)
- Helen M. Arthur
- Biosciences Institute, Centre for Life, University of Newcastle, Newcastle, United Kingdom
- *Correspondence: Helen M. Arthur,
| | - Beth L. Roman
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
21
|
Song YS, Annalora AJ, Marcus CB, Jefcoate CR, Sorenson CM, Sheibani N. Cytochrome P450 1B1: A Key Regulator of Ocular Iron Homeostasis and Oxidative Stress. Cells 2022; 11:2930. [PMID: 36230892 PMCID: PMC9563809 DOI: 10.3390/cells11192930] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/12/2022] [Accepted: 09/18/2022] [Indexed: 11/22/2022] Open
Abstract
Cytochrome P450 (CYP) 1B1 belongs to the superfamily of heme-containing monooxygenases. Unlike other CYP enzymes, which are highly expressed in the liver, CYP1B1 is predominantly found in extrahepatic tissues, such as the brain, and ocular tissues including retina and trabecular meshwork. CYP1B1 metabolizes exogenous chemicals such as polycyclic aromatic hydrocarbons. CYP1B1 also metabolizes endogenous bioactive compounds including estradiol and arachidonic acid. These metabolites impact various cellular and physiological processes during development and pathological processes. We previously showed that CYP1B1 deficiency mitigates ischemia-mediated retinal neovascularization and drives the trabecular meshwork dysgenesis through increased levels of oxidative stress. However, the underlying mechanisms responsible for CYP1B1-deficiency-mediated increased oxidative stress remain largely unresolved. Iron is an essential element and utilized as a cofactor in a variety of enzymes. However, excess iron promotes the production of hydroxyl radicals, lipid peroxidation, increased oxidative stress, and cell damage. The retinal endothelium is recognized as a major component of the blood-retinal barrier, which controls ocular iron levels through the modulation of proteins involved in iron regulation present in retinal endothelial cells, as well as other ocular cell types including trabecular meshwork cells. We previously showed increased levels of reactive oxygen species and lipid peroxidation in the absence of CYP1B1, and in the retinal vasculature and trabecular meshwork, which was reversed by administration of antioxidant N-acetylcysteine. Here, we review the important role CYP1B1 expression and activity play in maintaining retinal redox homeostasis through the modulation of iron levels by retinal endothelial cells. The relationship between CYP1B1 expression and activity and iron levels has not been previously delineated. We review the potential significance of CYP1B1 expression, estrogen metabolism, and hepcidin-ferroportin regulatory axis in the local regulation of ocular iron levels.
Collapse
Affiliation(s)
- Yong-Seok Song
- Departments of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Andrew J. Annalora
- Department of Environmental and Molecular Toxicology, Organ State University, Corvallis, OR 97331, USA
| | - Craig B. Marcus
- Department of Environmental and Molecular Toxicology, Organ State University, Corvallis, OR 97331, USA
| | - Colin R. Jefcoate
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Christine M. Sorenson
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
22
|
Betterton RD, Abdullahi W, Williams EI, Lochhead JJ, Brzica H, Stanton J, Reddell E, Ogbonnaya C, Davis TP, Ronaldson PT. Regulation of Blood-Brain Barrier Transporters by Transforming Growth Factor- β/Activin Receptor-Like Kinase 1 Signaling: Relevance to the Brain Disposition of 3-Hydroxy-3-Methylglutaryl Coenzyme A Reductase Inhibitors (i.e., Statins). Drug Metab Dispos 2022; 50:942-956. [PMID: 35504656 PMCID: PMC11022862 DOI: 10.1124/dmd.121.000781] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/08/2022] [Indexed: 11/22/2022] Open
Abstract
Our laboratory has shown that activation of transforming growth factor- β (TGF- β )/activin receptor-like kinase 1 (ALK1) signaling can increase protein expression and transport activity of organic anion transporting polypeptide 1a4 (Oatp1a4) at the blood-brain barrier (BBB). These results are relevant to treatment of ischemic stroke because Oatp transport substrates such as 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (i.e., statins) improve functional neurologic outcomes in patients. Advancement of our work requires determination if TGF- β /ALK1 signaling alters Oatp1a4 functional expression differently across brain regions and if such disparities affect central nervous system (CNS) statin disposition. Therefore, we studied regulation of Oatp1a4 by the TGF- β /ALK1 pathway, in vivo, in rat brain microvessels isolated from cerebral cortex, hippocampus, and cerebellum using the ALK1 agonist bone morphogenetic protein-9 (BMP-9) and the ALK1 inhibitor 4-[6-[4-(1-piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride 193189. We showed that Oatp1a4 protein expression and brain distribution of three currently marketed statin drugs (i.e., atorvastatin, pravastatin, and rosuvastatin) were increased in cortex relative to hippocampus and cerebellum. Additionally, BMP-9 treatment enhanced Oatp-mediated statin transport in cortical tissue but not in hippocampus or cerebellum. Although brain drug delivery is also dependent upon efflux transporters, such as P-glycoprotein and/or Breast Cancer Resistance Protein, our data showed that administration of BMP-9 did not alter the relative contribution of these transporters to CNS disposition of statins. Overall, this study provides evidence for differential regulation of Oatp1a4 by TGF- β /ALK1 signaling across brain regions, knowledge that is critical for development of therapeutic strategies to target Oatps at the BBB for CNS drug delivery. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (Oatps) represent transporter targets for brain drug delivery. We have shown that Oatp1a4 statin uptake is higher in cortex versus hippocampus and cerebellum. Additionally, we report that the transforming growth factor- β /activin receptor-like kinase 1 agonist bone morphogenetic protein-9 increases Oatp1a4 functional expression, but not efflux transporters P-glycoprotein and Breast Cancer Resistance Protein, in cortical brain microvessels. Overall, this study provides critical data that will advance treatment for neurological diseases where drug development has been challenging.
Collapse
Affiliation(s)
- Robert D Betterton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Wazir Abdullahi
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Erica I Williams
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Jeffrey J Lochhead
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Hrvoje Brzica
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Joshua Stanton
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Elizabeth Reddell
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Chidinma Ogbonnaya
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
23
|
Imbalances in circulating angiogenic factors in the pathophysiology of preeclampsia and related disorders. Am J Obstet Gynecol 2022; 226:S1019-S1034. [PMID: 33096092 PMCID: PMC8884164 DOI: 10.1016/j.ajog.2020.10.022] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/26/2020] [Accepted: 10/17/2020] [Indexed: 12/11/2022]
Abstract
Preeclampsia is a devastating medical complication of pregnancy that can lead to significant maternal and fetal morbidity and mortality. It is currently believed that there is abnormal placentation in as early as the first trimester in women destined to develop preeclampsia. Although the etiology of the abnormal placentation is being debated, numerous epidemiologic and experimental studies suggest that imbalances in circulating angiogenic factors released from the placenta are responsible for the maternal signs and symptoms of preeclampsia. In particular, circulating levels of soluble fms-like tyrosine kinase 1, an antiangiogenic factor, are markedly increased in women with preeclampsia, whereas free levels of its ligand, placental, growth factor are markedly diminished. Alterations in these angiogenic factors precede the onset of clinical signs of preeclampsia and correlate with disease severity. Recently, the availability of automated assays for the measurement of angiogenic biomarkers in the plasma, serum, and urine has helped investigators worldwide to demonstrate a key role for these factors in the clinical diagnosis and prediction of preeclampsia. Numerous studies have reported that circulating angiogenic biomarkers have a very high negative predictive value to rule out clinical disease among women with suspected preeclampsia. These blood-based biomarkers have provided a valuable tool to clinicians to accelerate the time to clinical diagnosis and minimize maternal adverse outcomes in women with preeclampsia. Angiogenic biomarkers have also been useful to elucidate the pathogenesis of related disorders of abnormal placentation such as intrauterine growth restriction, intrauterine fetal death, twin-to-twin transfusion syndrome, and fetal hydrops. In summary, the discovery and characterization of angiogenic proteins of placental origin have provided clinicians a noninvasive blood-based tool to monitor placental function and health and for early detection of disorders of placentation. Uncovering the mechanisms of altered angiogenic factors in preeclampsia and related disorders of placentation may provide insights into novel preventive and therapeutic options.
Collapse
|
24
|
Margioula-Siarkou G, Margioula-Siarkou C, Petousis S, Margaritis K, Vavoulidis E, Gullo G, Alexandratou M, Dinas K, Sotiriadis A, Mavromatidis G. The role of endoglin and its soluble form in pathogenesis of preeclampsia. Mol Cell Biochem 2022; 477:479-491. [PMID: 34783962 DOI: 10.1007/s11010-021-04294-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022]
Abstract
Preeclampsia remains till today a leading cause of maternal and fetal morbidity and mortality. Pathophysiology of the disease is not yet fully elucidated, though it is evident that it revolves around placenta. Cellular ischemia in the preeclamptic placenta creates an imbalance between angiogenic and anti-angiogenic factors in maternal circulation. Endoglin, a transmembrane co-receptor of transforming growth factor β (TGF-β) demonstrating angiogenic effects, is involved in a variety of angiogenesis-dependent diseases with endothelial dysfunction, including preeclampsia. Endoglin expression is up-regulated in preeclamptic placentas, through mechanisms mainly induced by hypoxia, oxidative stress and oxysterol-mediated activation of liver X receptors. Overexpression of endoglin results in an increase of its soluble form in maternal circulation. Soluble endoglin represents the extracellular domain of membrane endoglin, cleaved by the action of metalloproteinases, predominantly matrix metalloproteinase-14. Released in circulation, soluble endoglin interferes in TGF-β1 and activin receptor-like kinase 1 signaling pathways and inhibits endothelial nitric oxide synthase activation, consequently deranging angiogenesis and promoting vasoconstriction. Due to these properties, soluble endoglin actively contributes to the impaired placentation observed in preeclampsia, as well as to the pathogenesis and manifestation of its clinical signs and symptoms, especially hypertension and proteinuria. The significant role of endoglin and soluble endoglin in pathophysiology of preeclampsia could have prognostic, diagnostic and therapeutic perspectives. Further research is essential to extensively explore the potential use of these molecules in the management of preeclampsia in clinical settings.
Collapse
Affiliation(s)
- Georgia Margioula-Siarkou
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54624, Thessaloniki, Greece.
| | - Chrysoula Margioula-Siarkou
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54624, Thessaloniki, Greece
| | - Stamatios Petousis
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54624, Thessaloniki, Greece
| | - Kosmas Margaritis
- 2nd Department of Pediatrics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleftherios Vavoulidis
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54624, Thessaloniki, Greece
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, IVF Unit, Villa Sofia Cervello Hospital, University of Palermo, Palermo, Italy
| | - Maria Alexandratou
- Department of Radiology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Dinas
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54624, Thessaloniki, Greece
| | - Alexandros Sotiriadis
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54624, Thessaloniki, Greece
| | - Georgios Mavromatidis
- 2nd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54624, Thessaloniki, Greece
| |
Collapse
|
25
|
Soluble Endoglin Stimulates Inflammatory and Angiogenic Responses in Microglia That Are Associated with Endothelial Dysfunction. Int J Mol Sci 2022; 23:ijms23031225. [PMID: 35163148 PMCID: PMC8835690 DOI: 10.3390/ijms23031225] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/16/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Increased soluble endoglin (sENG) has been observed in human brain arteriovenous malformations (bAVMs). In addition, the overexpression of sENG in concurrence with vascular endothelial growth factor (VEGF)-A has been shown to induce dysplastic vessel formation in mouse brains. However, the underlying mechanism of sENG-induced vascular malformations is not clear. The evidence suggests the role of sENG as a pro-inflammatory modulator, and increased microglial accumulation and inflammation have been observed in bAVMs. Therefore, we hypothesized that microglia mediate sENG-induced inflammation and endothelial cell (EC) dysfunction in bAVMs. In this study, we confirmed that the presence of sENG along with VEGF-A overexpression induced dysplastic vessel formation. Remarkably, we observed increased microglial activation around dysplastic vessels with the expression of NLRP3, an inflammasome marker. We found that sENG increased the gene expression of VEGF-A, pro-inflammatory cytokines/inflammasome mediators (TNF-α, IL-6, NLRP3, ASC, Caspase-1, and IL-1β), and proteolytic enzyme (MMP-9) in BV2 microglia. The conditioned media from sENG-treated BV2 (BV2-sENG-CM) significantly increased levels of angiogenic factors (Notch-1 and TGFβ) and pERK1/2 in ECs but it decreased the level of IL-17RD, an anti-angiogenic mediator. Finally, the BV2-sENG-CM significantly increased EC migration and tube formation. Together, our study demonstrates that sENG provokes microglia to express angiogenic/inflammatory molecules which may be involved in EC dysfunction. Our study corroborates the contribution of microglia to the pathology of sENG-associated vascular malformations.
Collapse
|
26
|
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: 15] [Impact Index Per Article: 7.5] [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
|
27
|
Pawlak JB, Blobe GC. TGF-β superfamily co-receptors in cancer. Dev Dyn 2022; 251:137-163. [PMID: 33797167 PMCID: PMC8484463 DOI: 10.1002/dvdy.338] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 01/03/2023] Open
Abstract
Transforming growth factor-β (TGF-β) superfamily signaling via their cognate receptors is frequently modified by TGF-β superfamily co-receptors. Signaling through SMAD-mediated pathways may be enhanced or depressed depending on the specific co-receptor and cell context. This dynamic effect on signaling is further modified by the release of many of the co-receptors from the membrane to generate soluble forms that are often antagonistic to the membrane-bound receptors. The co-receptors discussed here include TβRIII (betaglycan), endoglin, BAMBI, CD109, SCUBE proteins, neuropilins, Cripto-1, MuSK, and RGMs. Dysregulation of these co-receptors can lead to altered TGF-β superfamily signaling that contributes to the pathophysiology of many cancers through regulation of growth, metastatic potential, and the tumor microenvironment. Here we describe the role of several TGF-β superfamily co-receptors on TGF-β superfamily signaling and the impact on cellular and physiological functions with a particular focus on cancer, including a discussion on recent pharmacological advances and potential clinical applications targeting these co-receptors.
Collapse
Affiliation(s)
| | - Gerard C. Blobe
- Department of Medicine, Duke University Medical Center,Department of Pharmacology and Cancer Biology, Duke University Medical Center,Corresponding author: Gerard Blobe, B354 LSRC, Box 91004 DUMC, Durham, NC 27708, , 919-668-1352
| |
Collapse
|
28
|
The Dual Effect of the BMP9-ALK1 Pathway in Blood Vessels: An Opportunity for Cancer Therapy Improvement? Cancers (Basel) 2021; 13:cancers13215412. [PMID: 34771575 PMCID: PMC8582496 DOI: 10.3390/cancers13215412] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The modulation of tumor blood vessels is a great opportunity for improving cancer therapies. Understanding the cellular and molecular players that regulate the biology of tumor blood vessels and tumor angiogenesis is necessary for the development of new anti-tumor strategies. Bone morphogenetic protein 9 (BMP9) is a circulating factor with multiple effects in vascular biology through its receptor activin receptor-like kinase 1 (ALK1). In this review, we give an overview of the possible benefits of modulating BMP9–ALK1 functions for cancer therapy improvement. Abstract The improvement of cancer therapy efficacy, the extension of patient survival and the reduction of adverse side effects are major challenges in cancer research. Targeting blood vessels has been considered a promising strategy in cancer therapy. Since the tumor vasculature is disorganized, leaky and triggers immunosuppression and tumor hypoxia, several strategies have been studied to modify tumor vasculature for cancer therapy improvement. Anti-angiogenesis was first described as a mechanism to prevent the formation of new blood vessels and prevent the oxygen supply to tumor cells, showing numerous limitations. Vascular normalization using low doses of anti-angiogenic drugs was purposed to overcome the limitations of anti-angiogenic therapies. Other strategies such as vascular promotion or the induction of high endothelial venules are being studied now to improve cancer therapy. Bone morphogenetic protein 9 (BMP9) exerts a dual effect through the activin receptor-like kinase 1 (ALK1) receptor in blood vessel maturation or activation phase of angiogenesis. Thus, it is an interesting pathway to target in combination with chemotherapies or immunotherapies. This review manuscript explores the effect of the BMP9–ALK1 pathway in tumor angiogenesis and the possible usefulness of targeting this pathway in anti-angiogenesis, vascular normalization or vascular promotion therapies.
Collapse
|
29
|
Ruiz-Llorente L, Vega MC, Fernández FJ, Langa C, Morrell NW, Upton PD, Bernabeu C. Generation of a Soluble Form of Human Endoglin Fused to Green Fluorescent Protein. Int J Mol Sci 2021; 22:ijms222011282. [PMID: 34681942 PMCID: PMC8539536 DOI: 10.3390/ijms222011282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 10/08/2021] [Accepted: 10/15/2021] [Indexed: 01/06/2023] Open
Abstract
Endoglin (Eng, CD105) is a type I membrane glycoprotein that functions in endothelial cells as an auxiliary receptor for transforming growth factor β (TGF-β)/bone morphogenetic protein (BMP) family members and as an integrin ligand, modulating the vascular pathophysiology. Besides the membrane-bound endoglin, there is a soluble form of endoglin (sEng) that can be generated by the action of the matrix metalloproteinase (MMP)-14 or -12 on the juxtamembrane region of its ectodomain. High levels of sEng have been reported in patients with preeclampsia, hypercholesterolemia, atherosclerosis and cancer. In addition, sEng is a marker of cardiovascular damage in patients with hypertension and diabetes, plays a pathogenic role in preeclampsia, and inhibits angiogenesis and tumor proliferation, migration, and invasion in cancer. However, the mechanisms of action of sEng have not yet been elucidated, and new tools and experimental approaches are necessary to advance in this field. To this end, we aimed to obtain a fluorescent form of sEng as a new tool for biological imaging. Thus, we cloned the extracellular domain of endoglin in the pEGFP-N1 plasmid to generate a fusion protein with green fluorescent protein (GFP), giving rise to pEGFP-N1/Eng.EC. The recombinant fusion protein was characterized by transient and stable transfections in CHO-K1 cells using fluorescence microscopy, SDS-PAGE, immunodetection, and ELISA techniques. Upon transfection with pEGFP-N1/Eng.EC, fluorescence was readily detected in cells, indicating that the GFP contained in the recombinant protein was properly folded into the cytosol. Furthermore, as evidenced by Western blot analysis, the secreted fusion protein yielded the expected molecular mass and displayed a specific fluorescent signal. The fusion protein was also able to bind to BMP9 and BMP10 in vitro. Therefore, the construct described here could be used as a tool for functional in vitro studies of the extracellular domain of endoglin.
Collapse
Affiliation(s)
- Lidia Ruiz-Llorente
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain; (L.R.-L.); (M.C.V.); (F.J.F.); (C.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain
- Biochemistry and Molecular Biology Unit, Department of System Biology, School of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid, Spain
| | - M. Cristina Vega
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain; (L.R.-L.); (M.C.V.); (F.J.F.); (C.L.)
| | - Francisco J. Fernández
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain; (L.R.-L.); (M.C.V.); (F.J.F.); (C.L.)
| | - Carmen Langa
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain; (L.R.-L.); (M.C.V.); (F.J.F.); (C.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK; (N.W.M.); (P.D.U.)
| | - Paul D. Upton
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK; (N.W.M.); (P.D.U.)
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain; (L.R.-L.); (M.C.V.); (F.J.F.); (C.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
30
|
Song T, Huang D, Song D. The potential regulatory role of BMP9 in inflammatory responses. Genes Dis 2021; 9:1566-1578. [PMID: 36157503 PMCID: PMC9485205 DOI: 10.1016/j.gendis.2021.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/24/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Inflammation is a protective response of the body to pathogens and injury. Hence, it is particularly important to explore the pathogenesis and key regulatory factors of inflammation. BMP9 is a unique member of the BMP family, which is widely known for its strong osteogenic potential and insensitivity to the inhibition of BMP3. Recently, several studies have reported an underlying pivotal link between BMP9 and inflammation. What is clear, though not well understood, is that BMP9 plays a role in inflammation in a carefully choreographed manner in different contexts. In this review, we have summarized current studies focusing on BMP9 and inflammation in various tissues and the latest advances in BMP9 expression, signal transduction, and crystal structure to better understand the relationship between BMP9 and inflammation. In addition, we also briefly summarized the inflammatory characteristics of some TGF-β superfamily members to provide better insights and ideas for the study of BMP9 and inflammation.
Collapse
Affiliation(s)
- Tianzhu Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
- Key Laboratory of Oral Diseases of Gansu Province, Northwest Minzu University, Key Laboratory of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, Gansu 730030, PR China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
- Corresponding author.
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
- Corresponding author.
| |
Collapse
|
31
|
Zakrzewski PK. Canonical TGFβ Signaling and Its Contribution to Endometrial Cancer Development and Progression-Underestimated Target of Anticancer Strategies. J Clin Med 2021; 10:3900. [PMID: 34501347 PMCID: PMC8432036 DOI: 10.3390/jcm10173900] [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: 08/12/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023] Open
Abstract
Endometrial cancer is one of the leading gynecological cancers diagnosed among women in their menopausal and postmenopausal age. Despite the progress in molecular biology and medicine, no efficient and powerful diagnostic and prognostic marker is dedicated to endometrial carcinogenesis. The canonical TGFβ pathway is a pleiotropic signaling cascade orchestrating a variety of cellular and molecular processes, whose alterations are responsible for carcinogenesis that originates from different tissue types. This review covers the current knowledge concerning the canonical TGFβ pathway (Smad-dependent) induced by prototypical TGFβ isoforms and the involvement of pathway alterations in the development and progression of endometrial neoplastic lesions. Since Smad-dependent signalization governs opposed cellular processes, such as growth arrest, apoptosis, tumor cells growth and differentiation, as well as angiogenesis and metastasis, TGFβ cascade may act both as a tumor suppressor or tumor promoter. However, the final effect of TGFβ signaling on endometrial cancer cells depends on the cancer disease stage. The multifunctional role of the TGFβ pathway indicates the possible utilization of alterations in the TGFβ cascade as a potential target of novel anticancer strategies.
Collapse
Affiliation(s)
- Piotr K Zakrzewski
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
32
|
FOXA1 promotes prostate cancer angiogenesis by inducing multiple pro-angiogenic factors expression. J Cancer Res Clin Oncol 2021; 147:3225-3243. [PMID: 34258652 DOI: 10.1007/s00432-021-03730-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 07/05/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE FOXA1, as a pioneering transcription factor, has been shown to drive prostate cancer progression. Previous studies showed that FOXA1 expression in prostate cancer was positively associated with cancer angiolymphatic invasion and metastasis. However, the mechanism underlying the correlation between FOXA1 and prostate cancer angiolymphatic invasion and metastasis remains largely unclear. METHODS Herein, we set out to investigate the role of FOXA1 in the interactions between prostate cancer cells and endothelial cells. Endothelial cells' phenotypes were assessed through CCK-8 assay, Transwell migration assay, and tube formation assay. The angiogenic factors acting on endothelial cells mediated by FOXA1were characterized by RNA-seq, qPCR array, angiogenesis cytokines array, and ELISA assay. The impact of FOXA1 on tumor angiogenesis was examined in a xenograft model in nude mice. The effect of FOXA1 on prostate cancer angiogenesis was validated on a primary prostate cancer tissue microarray. RESULTS FOXA1 expression in prostate cancer cells promoted endothelial cell proliferation, migration, and tube formation in vitro. Mechanistically, FOXA1 increased pro-angiogenic factors production, including EGF, Endothelin-1, and Endoglin. Moreover, in vivo study showed that FOXA1 facilitated tumor angiogenesis. Furthermore, clinical samples investigation indicated that FOXA1 enhanced prostate cancer angiogenesis. CONCLUSION Overall, these findings illustrated a tumor angiogenesis-promoting role of FOXA1 in prostate cancer.
Collapse
|
33
|
Desroches-Castan A, Tillet E, Bouvard C, Bailly S. BMP9 and BMP10: two close vascular quiescence partners that stand out. Dev Dyn 2021; 251:178-197. [PMID: 34240497 DOI: 10.1002/dvdy.395] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are dimeric transforming growth factor ß (TGFß) family cytokines that were first described in bone and cartilage formation but have since been shown to be involved in many pleiotropic functions. In human, there are 15 BMP ligands, which initiate their cellular signaling by forming a complex with two copies of type I receptors and two copies of type II receptors, both of which are transmembrane receptors with an intracellular serine/threonine kinase domain. Within this receptor family, ALK1 (Activin receptor-Like Kinase 1), which is a type I receptor mainly expressed on endothelial cells, and BMPRII (BMP Receptor type II), a type II receptor also highly expressed on endothelial cells, have been directly linked to two rare vascular diseases: hereditary haemorrhagic telangiectasia (HHT), and pulmonary arterial hypertension (PAH), respectively. BMP9 (gene name GDF2) and BMP10, two close members of the BMP family, are the only known ligands for the ALK1 receptor. This specificity gives them a unique role in physiological and pathological angiogenesis and tissue homeostasis. The aim of this current review is to present an overview of what is known about BMP9 and BMP10 on vascular regulation with a particular emphasis on recent results and the many questions that remain unanswered regarding the roles and specificities between BMP9 and BMP10. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Emmanuelle Tillet
- Laboratory BioSanté, Univ. Grenoble Alpes, INSERM, CEA, Grenoble, France
| | - Claire Bouvard
- Laboratory BioSanté, Univ. Grenoble Alpes, INSERM, CEA, Grenoble, France
| | - Sabine Bailly
- Laboratory BioSanté, Univ. Grenoble Alpes, INSERM, CEA, Grenoble, France
| |
Collapse
|
34
|
New insights into BMP9 signaling in liver diseases. Mol Cell Biochem 2021; 476:3591-3600. [PMID: 34019202 DOI: 10.1007/s11010-021-04182-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/12/2021] [Indexed: 02/08/2023]
Abstract
Bone morphogenetic protein 9 (BMP9) is a recently discovered cytokine mainly secreted by the liver and is a member of the transforming growth factor β (TGF-β) superfamily. In recent years, an increasing number of studies have shown that BMP9 is associated with liver diseases, including nonalcoholic fatty liver disease (NAFLD), liver fibrosis and hepatocellular carcinoma (HCC), and BMP9 signaling may play dual roles in liver diseases. In this review, we mainly summarized and discussed the roles and potential mechanisms of BMP9 signaling in NAFLD, liver fibrosis and HCC. Specifically, this article will provide a better understanding of BMP9 signaling and new clues for the treatment of liver diseases.
Collapse
|
35
|
Gebara N, Correia Y, Wang K, Bussolati B. Angiogenic Properties of Placenta-Derived Extracellular Vesicles in Normal Pregnancy and in Preeclampsia. Int J Mol Sci 2021; 22:5402. [PMID: 34065595 PMCID: PMC8160914 DOI: 10.3390/ijms22105402] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the main processes that coordinate the biological events leading to a successful pregnancy, and its imbalance characterizes several pregnancy-related diseases, including preeclampsia. Intracellular interactions via extracellular vesicles (EVs) contribute to pregnancy's physiology and pathophysiology, and to the fetal-maternal interaction. The present review outlines the implications of EV-mediated crosstalk in the angiogenic process in healthy pregnancy and its dysregulation in preeclampsia. In particular, the effect of EVs derived from gestational tissues in pro and anti-angiogenic processes in the physiological and pathological setting is described. Moreover, the application of EVs from placental stem cells in the clinical setting is reported.
Collapse
Affiliation(s)
- Natalia Gebara
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Yolanda Correia
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Keqing Wang
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| |
Collapse
|
36
|
Ollauri-Ibáñez C, Ayuso-Íñigo B, Pericacho M. Hot and Cold Tumors: Is Endoglin (CD105) a Potential Target for Vessel Normalization? Cancers (Basel) 2021; 13:1552. [PMID: 33800564 PMCID: PMC8038031 DOI: 10.3390/cancers13071552] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Tumors are complex masses formed by malignant but also by normal cells. The interaction between these cells via cytokines, chemokines, growth factors, and enzymes that remodel the extracellular matrix (ECM) constitutes the tumor microenvironment (TME). This TME can be determinant in the prognosis and the response to some treatments such as immunotherapy. Depending on their TME, two types of tumors can be defined: hot tumors, characterized by an immunosupportive TME and a good response to immunotherapy; and cold tumors, which respond poorly to this therapy and are characterized by an immunosuppressive TME. A therapeutic strategy that has been shown to be useful for the conversion of cold tumors into hot tumors is vascular normalization. In this review we propose that endoglin (CD105) may be a useful target of this strategy since it is involved in the three main processes involved in the generation of the TME: angiogenesis, inflammation, and cancer-associated fibroblast (CAF) accumulation. Moreover, the analysis of endoglin expression in tumors, which is already used in the clinic to study the microvascular density and that is associated with worse prognosis, could be used to predict a patient's response to immunotherapy.
Collapse
Affiliation(s)
| | | | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Group of Physiopathology of the Vascular Endothelium (ENDOVAS), Biomedical Research Institute of Salamanca (IBSAL), Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain; (C.O.-I.); (B.A.-Í.)
| |
Collapse
|
37
|
Burghardt I, Schroeder JJ, Weiss T, Gramatzki D, Weller M. A tumor-promoting role for soluble TβRIII in glioblastoma. Mol Cell Biochem 2021; 476:2963-2973. [PMID: 33772427 PMCID: PMC8263459 DOI: 10.1007/s11010-021-04128-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 03/04/2021] [Indexed: 12/21/2022]
Abstract
Purpose Members of the transforming growth factor (TGF)-β superfamily play a key role in the regulation of the malignant phenotype of glioblastoma by promoting invasiveness, angiogenesis, immunosuppression, and maintaining stem cell-like properties. Betaglycan, a TGF-β coreceptor also known as TGF-β receptor III (TβRIII), interacts with members of the TGF-β superfamily and acts as membrane-associated or shed molecule. Shed, soluble TβRIII (sTβRIII) is produced upon ectodomain cleavage of the membrane-bound form. Elucidating the role of TβRIII may improve our understanding of TGF-β pathway activity in glioblastoma Methods Protein levels of TβRIII were determined by immunohistochemical analyses and ex vivo single-cell gene expression profiling of glioblastoma tissue respectively. In vitro, TβRIII levels were assessed investigating long-term glioma cell lines (LTCs), cultured human brain-derived microvascular endothelial cells (hCMECs), glioblastoma-derived microvascular endothelial cells, and glioma-initiating cell lines (GICs). The impact of TβRIII on TGF-β signaling was investigated, and results were validated in a xenograft mouse glioma model Results Immunohistochemistry and ex vivo single-cell gene expression profiling of glioblastoma tissue showed that TβRIII was expressed in the tumor tissue, predominantly in the vascular compartment. We confirmed this pattern of TβRIII expression in vitro. Specifically, we detected sTβRIII in glioblastoma-derived microvascular endothelial cells. STβRIII facilitated TGF-β-induced Smad2 phosphorylation in vitro and overexpression of sTβRIII in a xenograft mouse glioma model led to increased levels of Smad2 phosphorylation, increased tumor volume, and decreased survival Conclusions These data shed light on the potential tumor-promoting role of extracellular shed TβRIII which may be released by glioblastoma endothelium with high sTβRIII levels. Supplementary Information The online version contains supplementary material available at 10.1007/s11010-021-04128-y.
Collapse
Affiliation(s)
- Isabel Burghardt
- Laboratory of Molecular Neuro-Oncology, Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Judith Johanna Schroeder
- Laboratory of Molecular Neuro-Oncology, Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Tobias Weiss
- Laboratory of Molecular Neuro-Oncology, Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Dorothee Gramatzki
- Laboratory of Molecular Neuro-Oncology, Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091, Zurich, Switzerland.
| |
Collapse
|
38
|
Kang Q, Li W, Xiao J, Yu N, Fan L, Sha M, Ma S, Wu J, Chen S. Identification of potential crucial genes associated with early-onset preeclampsia via bioinformatic analysis. Pregnancy Hypertens 2021; 24:27-36. [PMID: 33640831 DOI: 10.1016/j.preghy.2021.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 01/04/2021] [Accepted: 02/16/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Early-onset preeclampsia is a pregnancy complication associated with high maternal and perinatal morbidity, mortality. Intense efforts have been made to elucidate the pathogenesis, but the molecular mechanism is still elusive. This study aimed to identify potential key genes related to early-onset preeclampsia, and to obtain a better understanding of the molecular mechanisms of this disease. METHODS We performed a multi-step integrative bioinformatics analysis of microarray dataset GSE74341 downloaded from Gene Expression Omnibus (GEO) database including 7 early-onset preeclampsia and 5 gestational age matched normotensive controls. The differentially expressed genes (DEGs) were identified using the "limma" package, and their potential functions were predicted by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Furthermore, the protein-protein interaction network (PPI) was obtained from the STRING database and the PPI network was visualized by Cytoscape software. Then, hub modules and hub genes were screened out from the PPI network, and enrichment analysis was performed for them. Also, validation of hub genes expression in early-onset PE was down by using microarray dataset GSE44711. RESULTS A total of 628 DEGs (256 down- and 372 up-regulated) were identified in early-onset PE compared to controls. A total of 4 significant hub modules and 26 significant hub genes were identified. CONCLUSION In conclusion, the DEGs related to cell-cell or cell-extracellular matrix interaction (ITGA5, SPP1, LUM, VCAN, APP), placenta metabolic or oxidative stress (CCR7, NT5E, CYBB) were predicted to be newly potential crucial genes that may play significant roles in the pathogenesis of early-onset PE.
Collapse
Affiliation(s)
- Qingling Kang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Wei Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Juan Xiao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Nan Yu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Lei Fan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Menghan Sha
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Songyan Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Jianli Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Suhua Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
39
|
Vicen M, Igreja Sá IC, Tripská K, Vitverová B, Najmanová I, Eissazadeh S, Micuda S, Nachtigal P. Membrane and soluble endoglin role in cardiovascular and metabolic disorders related to metabolic syndrome. Cell Mol Life Sci 2021; 78:2405-2418. [PMID: 33185696 PMCID: PMC11072708 DOI: 10.1007/s00018-020-03701-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/05/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
Membrane endoglin (Eng, CD105) is a transmembrane glycoprotein essential for the proper function of vascular endothelium. It might be cleaved by matrix metalloproteinases to form soluble endoglin (sEng), which is released into the circulation. Metabolic syndrome comprises conditions/symptoms that usually coincide (endothelial dysfunction, arterial hypertension, hyperglycemia, obesity-related insulin resistance, and hypercholesterolemia), and are considered risk factors for cardiometabolic disorders such as atherosclerosis, type II diabetes mellitus, and liver disorders. The purpose of this review is to highlight current knowledge about the role of Eng and sEng in the disorders mentioned above, in vivo and in vitro extent, where we can find a wide range of contradictory results. We propose that reduced Eng expression is a hallmark of endothelial dysfunction development in chronic pathologies related to metabolic syndrome. Eng expression is also essential for leukocyte transmigration and acute inflammation, suggesting that Eng is crucial for the regulation of endothelial function during the acute phase of vascular defense reaction to harmful conditions. sEng was shown to be a circulating biomarker of preeclampsia, and we propose that it might be a biomarker of metabolic syndrome-related symptoms and pathologies, including hypercholesterolemia, hyperglycemia, arterial hypertension, and diabetes mellitus as well, despite the fact that some contradictory findings have been reported. Besides, sEng can participate in the development of endothelial dysfunction and promote the development of arterial hypertension, suggesting that high levels of sEng promote metabolic syndrome symptoms and complications. Therefore, we suggest that the treatment of metabolic syndrome should take into account the importance of Eng in the endothelial function and levels of sEng as a biomarker and risk factor of related pathologies.
Collapse
Affiliation(s)
- Matej Vicen
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Ivone Cristina Igreja Sá
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Katarína Tripská
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Barbora Vitverová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Iveta Najmanová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Samira Eissazadeh
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Stanislav Micuda
- Faculty of Medicine in Hradec Kralove, Department of Pharmacology, Charles University, Simkova 870, Hradec Kralove, 500 03, Czech Republic
| | - Petr Nachtigal
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic.
| |
Collapse
|
40
|
Snodgrass RO, Chico TJA, Arthur HM. Hereditary Haemorrhagic Telangiectasia, an Inherited Vascular Disorder in Need of Improved Evidence-Based Pharmaceutical Interventions. Genes (Basel) 2021; 12:174. [PMID: 33513792 PMCID: PMC7911152 DOI: 10.3390/genes12020174] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022] Open
Abstract
Hereditary haemorrhagic telangiectasia (HHT) is characterised by arteriovenous malformations (AVMs). These vascular abnormalities form when arteries and veins directly connect, bypassing the local capillary system. Large AVMs may occur in the lungs, liver and brain, increasing the risk of morbidity and mortality. Smaller AVMs, known as telangiectases, are prevalent on the skin and mucosal lining of the nose, mouth and gastrointestinal tract and are prone to haemorrhage. HHT is primarily associated with a reduction in endoglin (ENG) or ACVRL1 activity due to loss-of-function mutations. ENG and ACVRL1 transmembrane receptors are expressed on endothelial cells (ECs) and bind to circulating ligands BMP9 and BMP10 with high affinity. Ligand binding to the receptor complex leads to activation of the SMAD1/5/8 signalling pathway to regulate downstream gene expression. Various genetic animal models demonstrate that disruption of this pathway in ECs results in AVMs. The vascular abnormalities underlying AVM formation result from abnormal EC responses to angiogenic and haemodynamic cues, and include increased proliferation, reduced migration against the direction of blood flow and an increased EC footprint. There is growing evidence that targeting VEGF signalling has beneficial outcomes in HHT patients and in animal models of this disease. The anti-VEGF inhibitor bevacizumab reduces epistaxis and has a normalising effect on high cardiac output in HHT patients with hepatic AVMs. Blocking VEGF signalling also reduces vascular malformations in mouse models of HHT1 and HHT2. However, VEGF signalling is complex and drives numerous downstream pathways, and it is not yet clear which pathway (or combination of pathways) is critical to target. This review will consider the recent evidence gained from HHT clinical and preclinical studies that are increasing our understanding of HHT pathobiology and informing therapeutic strategies.
Collapse
Affiliation(s)
- Ryan O. Snodgrass
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, UK; (R.O.S.); (T.J.A.C.)
| | - Timothy J. A. Chico
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, UK; (R.O.S.); (T.J.A.C.)
| | - Helen M. Arthur
- Biosciences Institute, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| |
Collapse
|
41
|
SOLUBLE ENDOGLIN AS AN EARLY PREDICTION MARKER OF INTRAUTERINE GROWTH RETARDATION. WORLD OF MEDICINE AND BIOLOGY 2021. [DOI: 10.26724/2079-8334-2021-4-78-41-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Liu Y, Paauwe M, Nixon AB, Hawinkels LJ. Endoglin Targeting: Lessons Learned and Questions That Remain. Int J Mol Sci 2020; 22:ijms22010147. [PMID: 33375670 PMCID: PMC7795616 DOI: 10.3390/ijms22010147] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Approximately 30 years ago, endoglin was identified as a transforming growth factor (TGF)-β coreceptor with a crucial role in developmental biology and tumor angiogenesis. Its selectively high expression on tumor vessels and its correlation with poor survival in cancer patients led to the exploration of endoglin as a therapeutic target for cancer. The endoglin neutralizing antibody TRC105 (Carotuximab®, Tracon Pharmaceuticals (San Diego, CA, USA) was subsequently tested in a wide variety of preclinical cancer models before being tested in phase I-III clinical studies in cancer patients as both a monotherapy and in combination with other chemotherapeutic and anti-angiogenic therapies. The combined data of these studies have revealed new insights into the role of endoglin in angiogenesis and its expression and functional role on other cells in the tumor microenvironment. In this review, we will summarize the preclinical work, clinical trials and biomarker studies of TRC105 and explore what these studies have enabled us to learn and what questions remain unanswered.
Collapse
Affiliation(s)
- Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (Y.L.); (A.B.N.)
| | - Madelon Paauwe
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Andrew B. Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (Y.L.); (A.B.N.)
| | - Lukas J.A.C. Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Correspondence: ; Tel.: +31-71-526-6736
| |
Collapse
|
43
|
Migliorini E, Guevara-Garcia A, Albiges-Rizo C, Picart C. Learning from BMPs and their biophysical extracellular matrix microenvironment for biomaterial design. Bone 2020; 141:115540. [PMID: 32730925 PMCID: PMC7614069 DOI: 10.1016/j.bone.2020.115540] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 01/19/2023]
Abstract
It is nowadays well-accepted that the extracellular matrix (ECM) is not a simple reservoir for growth factors but is an organization center of their biological activity. In this review, we focus on the ability of the ECM to regulate the biological activity of BMPs. In particular, we survey the role of the ECM components, notably the glycosaminoglycans and fibrillary ECM proteins, which can be promoters or repressors of the biological activities mediated by the BMPs. We examine how a process called mechano-transduction induced by the ECM can affect BMP signaling, including BMP internalization by the cells. We also focus on the spatio-temporal regulation of the BMPs, including their release from the ECM, which enables to modulate their spatial localization as well as their local concentration. We highlight how biomaterials can recapitulate some aspects of the BMPs/ECM interactions and help to answer fundamental questions to reveal previously unknown molecular mechanisms. Finally, the design of new biomaterials inspired by the ECM to better present BMPs is discussed, and their use for a more efficient bone regeneration in vivo is also highlighted.
Collapse
Affiliation(s)
- Elisa Migliorini
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| | - Amaris Guevara-Garcia
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France; Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Corinne Albiges-Rizo
- Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Catherine Picart
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| |
Collapse
|
44
|
Yang Z, Li P, Shang Q, Wang Y, He J, Ge S, Jia R, Fan X. CRISPR-mediated BMP9 ablation promotes liver steatosis via the down-regulation of PPARα expression. SCIENCE ADVANCES 2020; 6:6/48/eabc5022. [PMID: 33246954 PMCID: PMC7695473 DOI: 10.1126/sciadv.abc5022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/14/2020] [Indexed: 05/09/2023]
Abstract
Obesity drives the development of nonalcoholic fatty liver disease (NAFLD) characterized by hepatic steatosis. Several bone morphogenetic proteins (BMPs) except BMP9 were reported related to metabolic syndrome. This study demonstrates that liver cytokine BMP9 is decreased in the liver and serum of NAFLD model mice and patients. BMP9 knockdown induces lipid accumulation in Hepa 1-6 cells. BMP9-knockout mice exhibit hepatosteatosis due to down-regulated peroxisome proliferator-activated receptor α (PPARα) expression and reduced fatty acid oxidation. In vitro, recombinant BMP9 treatment attenuates triglyceride accumulation by enhancing PPARα promoter activity via the activation of p-smad. PPARα-specific antagonist GW6471 abolishes the effect of BMP9 knockdown. Furthermore, adeno-associated virus-mediated BMP9 overexpression in mouse liver markedly relieves liver steatosis and obesity-related metabolic syndrome. These findings indicate that BMP9 plays a critical role in regulating hepatic lipid metabolism in a PPARα-dependent manner and may provide a previously unknown insight into NAFLD therapeutic approaches.
Collapse
Affiliation(s)
- Z Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200032, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| | - P Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| | - Q Shang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| | - Y Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| | - J He
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| | - S Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| | - R Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| | - X Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 833 Zhizaoju Road, Shanghai 200011, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200032, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, 833 Zhizaoju Road, Shanghai 200011, China
| |
Collapse
|
45
|
Influence of the TGF-β Superfamily on Osteoclasts/Osteoblasts Balance in Physiological and Pathological Bone Conditions. Int J Mol Sci 2020; 21:ijms21207597. [PMID: 33066607 PMCID: PMC7589189 DOI: 10.3390/ijms21207597] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/19/2022] Open
Abstract
The balance between bone forming cells (osteoblasts/osteocytes) and bone resorbing cells (osteoclasts) plays a crucial role in tissue homeostasis and bone repair. Several hormones, cytokines, and growth factors-in particular the members of the TGF-β superfamily such as the bone morphogenetic proteins-not only regulate the proliferation, differentiation, and functioning of these cells, but also coordinate the communication between them to ensure an appropriate response. Therefore, this review focuses on TGF-β superfamily and its influence on bone formation and repair, through the regulation of osteoclastogenesis, osteogenic differentiation of stem cells, and osteoblasts/osteoclasts balance. After introducing the main types of bone cells, their differentiation and cooperation during bone remodeling and fracture healing processes are discussed. Then, the TGF-β superfamily, its signaling via canonical and non-canonical pathways, as well as its regulation by Wnt/Notch or microRNAs are described and discussed. Its important role in bone homeostasis, repair, or disease is also highlighted. Finally, the clinical therapeutic uses of members of the TGF-β superfamily and their associated complications are debated.
Collapse
|
46
|
Yang Y, Lin F, Xiao Z, Sun B, Wei Z, Liu B, Xue L, Xiong C. Investigational pharmacotherapy and immunotherapy of pulmonary arterial hypertension: An update. Biomed Pharmacother 2020; 129:110355. [DOI: 10.1016/j.biopha.2020.110355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/22/2020] [Accepted: 05/30/2020] [Indexed: 12/13/2022] Open
|
47
|
Hwan Kim Y, Vu PN, Choe SW, Jeon CJ, Arthur HM, Vary CPH, Lee YJ, Oh SP. Overexpression of Activin Receptor-Like Kinase 1 in Endothelial Cells Suppresses Development of Arteriovenous Malformations in Mouse Models of Hereditary Hemorrhagic Telangiectasia. Circ Res 2020; 127:1122-1137. [PMID: 32762495 DOI: 10.1161/circresaha.119.316267] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RATIONALE Hereditary hemorrhagic telangiectasia (HHT) is a genetic disease caused by mutations in ENG, ALK1, or SMAD4. Since proteins from all 3 HHT genes are components of signal transduction of TGF-β (transforming growth factor β) family members, it has been hypothesized that HHT is a disease caused by defects in the ENG-ALK1-SMAD4 linear signaling. However, in vivo evidence supporting this hypothesis is scarce. OBJECTIVE We tested this hypothesis and investigated the therapeutic effects and potential risks of induced-ALK1 or -ENG overexpression (OE) for HHT. METHODS AND RESULTS We generated a novel mouse allele (ROSA26Alk1) in which HA (human influenza hemagglutinin)-tagged ALK1 and bicistronic eGFP expression are induced by Cre activity. We examined whether ALK1-OE using the ROSA26Alk1 allele could suppress the development of arteriovenous malformations (AVMs) in wounded adult skin and developing retinas of Alk1- and Eng-inducible knockout (iKO) mice. We also used a similar approach to investigate whether ENG-OE could rescue AVMs. Biochemical and immunofluorescence analyses confirmed the Cre-dependent OE of the ALK1-HA transgene. We could not detect any pathological signs in ALK1-OE mice up to 3 months after induction. ALK1-OE prevented the development of retinal AVMs and wound-induced skin AVMs in Eng-iKO as well as Alk1-iKO mice. ALK1-OE normalized expression of SMAD and NOTCH target genes in ENG-deficient endothelial cells (ECs) and restored the effect of BMP9 (bone morphogenetic protein 9) on suppression of phosphor-AKT levels in these endothelial cells. On the other hand, ENG-OE could not inhibit the AVM development in Alk1-iKO models. CONCLUSIONS These data support the notion that ENG and ALK1 form a linear signaling pathway for the formation of a proper arteriovenous network during angiogenesis. We suggest that ALK1 OE or activation can be an effective therapeutic strategy for HHT. Further research is required to study whether this therapy could be translated into treatment for humans.
Collapse
Affiliation(s)
- Yong Hwan Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (Y.H.K., S.-w.C., S.P.O.).,Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ (Y.H.K., S.P.O.)
| | - Phuong-Nhung Vu
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea (N.V.P., Y.J.L.)
| | - Se-Woon Choe
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (Y.H.K., S.-w.C., S.P.O.).,Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, Republic of Korea (S.-w.C.)
| | - Chang-Jin Jeon
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, Korea (C.J.J.)
| | - Helen M Arthur
- Institute of Genetic Medicine, Newcastle University, United Kingdom (H.M.A.)
| | - Calvin P H Vary
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough (C.P.V.)
| | - Young Jae Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea (N.V.P., Y.J.L.)
| | - S Paul Oh
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (Y.H.K., S.-w.C., S.P.O.).,Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ (Y.H.K., S.P.O.)
| |
Collapse
|
48
|
Aukema SM, Ten Brinke GA, Timens W, Vos YJ, Accord RE, Kraft KE, Santing MJ, Morssink LP, Streefland E, van Diemen CC, Vrijlandt EJ, Hulzebos CV, Kerstjens-Frederikse WS. A homozygous variant in growth and differentiation factor 2 (GDF2) may cause lymphatic dysplasia with hydrothorax and nonimmune hydrops fetalis. Am J Med Genet A 2020; 182:2152-2160. [PMID: 32618121 DOI: 10.1002/ajmg.a.61743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/08/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023]
Abstract
The etiology of nonimmune hydrops fetalis is extensive and includes genetic disorders. We describe a term-born female neonate with late onset extensive nonimmune hydrops, that is, polyhydramnios, edema, and congenital bilateral chylothorax. This newborn was successfully treated with repetitive thoracocentesis, total parenteral feeding, octreotide intravenously and finally surgical pleurodesis and corticosteroids. A genetic cause seemed plausible as the maternal history revealed a fatal nonimmune hydrops fetalis. A homozygous truncating variant in GDF2 (c.451C>T, p.(Arg151*)) was detected with exome sequencing. Genetic analysis of tissue obtained from the deceased fetal sibling revealed the same homozygous variant. The parents and two healthy siblings were heterozygous for the GDF2 variant. Skin and lung biopsies in the index patient, as well as the revised lung biopsy of the deceased fetal sibling, showed lymphatic dysplasia and lymphangiectasia. To the best of our knowledge, this is the first report of an association between a homozygous variant in GDF2 with lymphatic dysplasia, hydrothorax and nonimmune hydrops fetalis.
Collapse
Affiliation(s)
- Sietse M Aukema
- Department of Clinical Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerdien A Ten Brinke
- Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Yvonne J Vos
- Department of Clinical Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ryan E Accord
- Department of Congenital Cardiothoracic Surgery, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, Center for Congenital Heart Diseases, Groningen, The Netherlands
| | - Karianne E Kraft
- Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel J Santing
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Leonard P Morssink
- Department of Obstetrics and Gynaecology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Esther Streefland
- Department of Obstetrics and Gynecology/Prenatal diagnosis, University Medical Centre of Groningen, University of Groningen, Groningen, The Netherlands
| | - Cleo C van Diemen
- Department of Clinical Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elianne Jle Vrijlandt
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Christian V Hulzebos
- Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
| | | |
Collapse
|
49
|
Singh E, Redgrave RE, Phillips HM, Arthur HM. Arterial endoglin does not protect against arteriovenous malformations. Angiogenesis 2020; 23:559-566. [PMID: 32506200 PMCID: PMC7524831 DOI: 10.1007/s10456-020-09731-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022]
Abstract
Introduction Endoglin (ENG) forms a receptor complex with ALK1 in endothelial cells (ECs) to promote BMP9/10 signalling. Loss of function mutations in either ENG or ALK1 genes lead to the inherited vascular disorder hereditary haemorrhagic telangiectasia (HHT), characterised by arteriovenous malformations (AVMs). However, the vessel-specific role of ENG and ALK1 proteins in protecting against AVMs is unclear. For example, AVMs have been described to initiate in arterioles, whereas ENG is predominantly expressed in venous ECs. To investigate whether ENG has any arterial involvement in protecting against AVM formation, we specifically depleted the Eng gene in venous and capillary endothelium whilst maintaining arterial expression, and investigated how this affected the incidence and location of AVMs in comparison with pan-endothelial Eng knockdown. Methods Using the mouse neonatal retinal model of angiogenesis, we first established the earliest time point at which Apj-Cre-ERT2 activity was present in venous and capillary ECs but absent from arterial ECs. We then compared the incidence of AVMs following pan-endothelial or venous/capillary-specific ENG knockout. Results Activation of Apj-Cre-ERT2 with tamoxifen from postnatal day (P) 5 ensured preservation of arterial ENG protein expression. Specific loss of ENG expression in ECs of veins and capillaries led to retinal AVMs at a similar frequency to pan-endothelial loss of ENG. AVMs occurred in the proximal as well as the distal part of the retina consistent with a defect in vascular remodelling during maturation of the vasculature. Conclusion Expression of ENG is not required in arterial ECs to protect against AVM formation. Electronic supplementary material The online version of this article (10.1007/s10456-020-09731-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Esha Singh
- Centre for Life, Biosciences Institute, Newcastle University, Newcastle, NE1 3BZ, UK
| | - Rachael E Redgrave
- Centre for Life, Biosciences Institute, Newcastle University, Newcastle, NE1 3BZ, UK
| | - Helen M Phillips
- Centre for Life, Biosciences Institute, Newcastle University, Newcastle, NE1 3BZ, UK
| | - Helen M Arthur
- Centre for Life, Biosciences Institute, Newcastle University, Newcastle, NE1 3BZ, UK.
| |
Collapse
|
50
|
Vitverova B, Najmanova I, Vicen M, Tripska K, Sa ICI, Hyspler R, Pericacho M, Nachtigal P. Long term effects of soluble endoglin and mild hypercholesterolemia in mice hearts. PLoS One 2020; 15:e0233725. [PMID: 32470058 PMCID: PMC7259503 DOI: 10.1371/journal.pone.0233725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/11/2020] [Indexed: 01/15/2023] Open
Abstract
Soluble endoglin (sEng) released into the circulation was suggested to be related to cardiovascular based pathologies. It was demonstrated that a combination of high sEng levels and long-term exposure (six months) to high fat diet (HFD) resulted in aggravation of endothelial dysfunction in the aorta. Thus, in this study, we hypothesized that a similar experimental design would affect the heart morphology, TGFβ signaling, inflammation, fibrosis, oxidative stress and eNOS signaling in myocardium in transgenic mice overexpressing human sEng. Three-month-old female transgenic mice overexpressing human sEng in plasma (Sol-Eng+ high) and their age-matched littermates with low levels of human sEng (Sol-Eng+ low) were fed a high-fat diet containing 1.25% of cholesterol and 40% of fat for six months. A blood analysis was performed, and the heart samples were analyzed by qRT-PCR and Western blot. The results of this study showed no effects of sEng and HFD on myocardial morphology/hypertrophy/fibrosis. However, the expression of pSmad2/3 and p-eNOS was reduced in Sol-Eng+ high mice. On the other hand, sEng and HFD did not significantly affect the expression of selected members of TGFβ signaling (membrane endoglin, TGFβRII, ALK-5, ALK-1, Id-1, PAI-1), inflammation (VCAM-1, ICAM-1), oxidative stress (NQO1, HO-1) and heart remodeling (PDGFβ, COL1A1, β-MHC). In conclusion, the results of this study confirmed that sEng, even combined with a high-fat diet inducing hypercholesterolemia administered for six months, does not affect the structure of the heart with respect to hypertrophy, fibrosis, inflammation and oxidative stress. Interestingly, pSmad2/3/p-eNOS signaling was reduced in both the heart in this study and the aorta in the previous study, suggesting a possible alteration of NO metabolism caused by six months exposure to high sEng levels and HFD. Thus, we might conclude that sEng combined with a high-fat diet might be related to the alteration of NO production due to altered pSmad2/3/p-eNOS signaling in the heart and aorta.
Collapse
Affiliation(s)
- Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Iveta Najmanova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Matej Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Katarina Tripska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ivone Cristina Igreja Sa
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Radek Hyspler
- Centrum for Research and Development, University Hospital, Hradec Kralove, Czech Republic
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, and the Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
- * E-mail:
| |
Collapse
|