201
|
Mechanobiology of TGFβ signaling in the skeleton. Matrix Biol 2016; 52-54:413-425. [PMID: 26877077 DOI: 10.1016/j.matbio.2016.02.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 12/12/2022]
Abstract
Physical and biochemical cues play fundamental roles in the skeleton at both the tissue and cellular levels. The precise coordination of these cues is essential for skeletal development and homeostasis, and disruption of this coordination can drive disease progression. The growth factor TGFβ is involved in both the regulation of and cellular response to the physical microenvironment. It is essential to summarize the current findings regarding the mechanisms by which skeletal cells integrate physical and biochemical cues so that we can identify and address remaining gaps that could ultimately improve skeletal health. In this review, we describe the role of TGFβ in mechanobiological signaling in bone and cartilage at the tissue and cellular levels. We provide detail on how static and dynamic physical cues at the macro-level are transmitted to the micro-level, ultimately leading to regulation at each level of the TGFβ pathway and to cell differentiation. The continued integration of engineering and biological approaches is needed to answer many remaining questions, such as the mechanisms by which cells generate a coordinated response to physical and biochemical cues. We propose one such mechanism, through which the combination of TGFβ and an optimal physical microenvironment leads to synergistic induction of downstream TGFβ signaling.
Collapse
|
202
|
Yan XC, Cao J, Liang L, Wang L, Gao F, Yang ZY, Duan JL, Chang TF, Deng SM, Liu Y, Dou GR, Zhang J, Zheng QJ, Zhang P, Han H. miR-342-5p Is a Notch Downstream Molecule and Regulates Multiple Angiogenic Pathways Including Notch, Vascular Endothelial Growth Factor and Transforming Growth Factor β Signaling. J Am Heart Assoc 2016; 5:JAHA.115.003042. [PMID: 26857067 PMCID: PMC4802463 DOI: 10.1161/jaha.115.003042] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Endothelial cells (ECs) form blood vessels through angiogenesis that is regulated by coordination of vascular endothelial growth factor (VEGF), Notch, transforming growth factor β, and other signals, but the detailed molecular mechanisms remain unclear. Methods and Results Small RNA sequencing initially identified miR‐342‐5p as a novel downstream molecule of Notch signaling in ECs. Reporter assay, quantitative reverse transcription polymerase chain reaction and Western blot analysis indicated that miR‐342‐5p targeted endoglin and modulated transforming growth factor β signaling by repressing SMAD1/5 phosphorylation in ECs. Transfection of miR‐342‐5p inhibited EC proliferation and lumen formation and reduced angiogenesis in vitro and in vivo, as assayed by using a fibrin beads–based sprouting assay, mouse aortic ring culture, and intravitreal injection of miR‐342‐5p agomir in P3 pups. Moreover, miR‐342‐5p promoted the migration of ECs, accompanied by reduced endothelial markers and increased mesenchymal markers, indicative of increased endothelial–mesenchymal transition. Transfection of endoglin at least partially reversed endothelial–mesenchymal transition induced by miR‐342‐5p. The expression of miR‐342‐5p was upregulated by transforming growth factor β, and inhibition of miR‐342‐5p attenuated the inhibitory effects of transforming growth factor β on lumen formation and sprouting by ECs. In addition, VEGF repressed miR‐342‐5p expression, and transfection of miR‐342‐5p repressed VEGFR2 and VEGFR3 expression and VEGF‐triggered Akt phosphorylation in ECs. miR‐342‐5p repressed angiogenesis in a laser‐induced choroidal neovascularization model in mice, highlighting its clinical potential. Conclusions miR‐342‐5p acts as a multifunctional angiogenic repressor mediating the effects and interaction among angiogenic pathways.
Collapse
Affiliation(s)
- Xian-Chun Yan
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Cao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liang Liang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Li Wang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fang Gao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zi-Yan Yang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Juan-Li Duan
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tian-Fang Chang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - San-Ming Deng
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuan Liu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guo-Rui Dou
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jian Zhang
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qi-Jun Zheng
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ping Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
203
|
Gratchev A. TGF-β signalling in tumour associated macrophages. Immunobiology 2016; 222:75-81. [PMID: 26876591 DOI: 10.1016/j.imbio.2015.11.016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/23/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022]
Abstract
Tumour associated macrophages (TAM) represent an important component of tumour stroma. They develop under the influence of tumour microenvironment where transforming growth factor (TGF)β is frequently present. Activities of TAM regulated by TGFβ stimulate proliferation of tumour cells and lead to tumour immune escape. Despite high importance of TGFβ-induction of TAM activities till now our understanding of the mechanism of this induction is limited. We have previously developed a model of type 2 macrophages (M2) resembling certain properties of TAM. We established that in M2 TGFβRII is regulated on the level of subcellular sorting by glucocorticoids. Further studies revealed that in M2 with high levels of TGFβRII on the surface TGFβ activates not only its canonical Smad2/3-mediated signaling, but also Smad1/5-mediated signaling, what is rather typical for bone morphogenetic protein (BMP) stimulation. Complexity of macrophage populations, however, allows assumption that TGFβ signalling may function in different ways depending on the functional state of the cell. To understand the peculiarities of TGFβ signalling in human TAMs experimental systems using primary cells have to be developed and used together with the modern mathematical modelling approaches.
Collapse
Affiliation(s)
- Alexei Gratchev
- Blokhin Cancer Research Center, Moscow, Russia; Laboratory for translational cellular and molecular biomedicine, Tomsk State University, Tomsk, Russia.
| |
Collapse
|
204
|
Morrell NW, Bloch DB, ten Dijke P, Goumans MJTH, Hata A, Smith J, Yu PB, Bloch KD. Targeting BMP signalling in cardiovascular disease and anaemia. Nat Rev Cardiol 2016; 13:106-20. [PMID: 26461965 PMCID: PMC4886232 DOI: 10.1038/nrcardio.2015.156] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone morphogenetic proteins (BMPs) and their receptors, known to be essential regulators of embryonic patterning and organogenesis, are also critical for the regulation of cardiovascular structure and function. In addition to their contributions to syndromic disorders including heart and vascular development, BMP signalling is increasingly recognized for its influence on endocrine-like functions in postnatal cardiovascular and metabolic homeostasis. In this Review, we discuss several critical and novel aspects of BMP signalling in cardiovascular health and disease, which highlight the cell-specific and context-specific nature of BMP signalling. Based on advancing knowledge of the physiological roles and regulation of BMP signalling, we indicate opportunities for therapeutic intervention in a range of cardiovascular conditions including atherosclerosis and pulmonary arterial hypertension, as well as for anaemia of inflammation. Depending on the context and the repertoire of ligands and receptors involved in specific disease processes, the selective inhibition or enhancement of signalling via particular BMP ligands (such as in atherosclerosis and pulmonary arterial hypertension, respectively) might be beneficial. The development of selective small molecule antagonists of BMP receptors, and the identification of ligands selective for BMP receptor complexes expressed in the vasculature provide the most immediate opportunities for new therapies.
Collapse
Affiliation(s)
- Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Donald B Bloch
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Peter ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Marie-Jose T H Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medicine Centre, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Akiko Hata
- Cardiovascular Research Institute, University of California, 500 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jim Smith
- MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Paul B Yu
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Kenneth D Bloch
- Anaesthesia Centre for Critical Care Research, Department of Anaesthesia, Critical Care and Pain Medicine, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
205
|
Activin Receptor-Like Kinase Receptors ALK5 and ALK1 Are Both Required for TGFβ-Induced Chondrogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells. PLoS One 2015; 10:e0146124. [PMID: 26720610 PMCID: PMC4697836 DOI: 10.1371/journal.pone.0146124] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023] Open
Abstract
Introduction Bone marrow-derived mesenchymal stem cells (BMSCs) are promising for cartilage regeneration because BMSCs can differentiate into cartilage tissue-producing chondrocytes. Transforming Growth Factor β (TGFβ) is crucial for inducing chondrogenic differentiation of BMSCs and is known to signal via Activin receptor-Like Kinase (ALK) receptors ALK5 and ALK1. Since the specific role of these two TGFβ receptors in chondrogenesis is unknown, we investigated whether ALK5 and ALK1 are expressed in BMSCs and whether both receptors are required for chondrogenic differentiation of BMSCs. Materials & Methods ALK5 and ALK1 gene expression in human BMSCs was determined with RT-qPCR. To induce chondrogenesis, human BMSCs were pellet-cultured in serum-free chondrogenic medium containing TGFβ1. Chondrogenesis was evaluated by aggrecan and collagen type IIα1 RT-qPCR analysis, and histological stainings of proteoglycans and collagen type II. To overexpress constitutively active (ca) receptors, BMSCs were transduced either with caALK5 or caALK1. Expression of ALK5 and ALK1 was downregulated by transducing BMSCs with shRNA against ALK5 or ALK1. Results ALK5 and ALK1 were expressed in in vitro-expanded as well as in pellet-cultured BMSCs from five donors, but mRNA levels of both TGFβ receptors did not clearly associate with chondrogenic induction. TGFβ increased ALK5 and decreased ALK1 gene expression in chondrogenically differentiating BMSC pellets. Neither caALK5 nor caALK1 overexpression induced cartilage matrix formation as efficient as that induced by TGFβ. Moreover, short hairpin-mediated downregulation of either ALK5 or ALK1 resulted in a strong inhibition of TGFβ-induced chondrogenesis. Conclusion ALK5 as well as ALK1 are required for TGFβ-induced chondrogenic differentiation of BMSCs, and TGFβ not only directly induces chondrogenesis, but also modulates ALK5 and ALK1 receptor signaling in BMSCs. These results imply that optimizing cartilage formation by mesenchymal stem cells will depend on activation of both receptors.
Collapse
|
206
|
Kienast Y, Jucknischke U, Scheiblich S, Thier M, de Wouters M, Haas A, Lehmann C, Brand V, Bernicke D, Honold K, Lorenz S. Rapid Activation of Bone Morphogenic Protein 9 by Receptor-mediated Displacement of Pro-domains. J Biol Chem 2015; 291:3395-410. [PMID: 26677222 DOI: 10.1074/jbc.m115.680009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Indexed: 12/21/2022] Open
Abstract
By non-covalent association after proteolytic cleavage, the pro-domains modulate the activities of the mature growth factor domains across the transforming growth factor-β family. In the case of bone morphogenic protein 9 (BMP9), however, the pro-domains do not inhibit the bioactivity of the growth factor, and the BMP9·pro-domain complexes have equivalent biological activities as the BMP9 mature ligand dimers. By using real-time surface plasmon resonance, we could demonstrate that either binding of pro-domain-complexed BMP9 to type I receptor activin receptor-like kinase 1 (ALK1), type II receptors, co-receptor endoglin, or to mature BMP9 domain targeting antibodies leads to immediate and complete displacement of the pro-domains from the complex. Vice versa, pro-domain binding by an anti-pro-domain antibody results in release of the mature BMP9 growth factor. Based on these findings, we adjusted ELISA assays to measure the protein levels of different BMP9 variants. Although mature BMP9 and inactive precursor BMP9 protein were directly detectable by ELISA, BMP9·pro-domain complex could only be measured indirectly as dissociated fragments due to displacement of mature growth factor and pro-domains after antibody binding. Our studies provide a model in which BMP9 can be readily activated upon getting into contact with its receptors. This increases the understanding of the underlying biology of BMP9 activation and also provides guidance for ELISA development for the detection of circulating BMP9 variants.
Collapse
Affiliation(s)
- Yvonne Kienast
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany,
| | - Ute Jucknischke
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Stefan Scheiblich
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Martina Thier
- From the Roche Pharma Research and Early Development (pRED), Translational Technologies and Bioinformatics, Roche Innovation Center, Basel, 4070 Basel, Switzerland, and
| | - Mariana de Wouters
- From the Roche Pharma Research and Early Development (pRED), Translational Technologies and Bioinformatics, Roche Innovation Center, Basel, 4070 Basel, Switzerland, and
| | - Alexander Haas
- From the Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Penzberg, 82377 Penzberg Germany
| | - Christian Lehmann
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Verena Brand
- From the Roche Pharma Research and Early Development (pRED)
| | - Dirk Bernicke
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Konrad Honold
- From the Roche Pharma Research and Early Development (pRED), Discovery Oncology, Roche Innovation Center Penzberg, 82377 Penzberg, Germany
| | - Stefan Lorenz
- From the Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Penzberg, 82377 Penzberg Germany
| |
Collapse
|
207
|
García de Vinuesa A, Abdelilah-Seyfried S, Knaus P, Zwijsen A, Bailly S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev 2015; 27:65-79. [PMID: 26823333 DOI: 10.1016/j.cytogfr.2015.12.005] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vascular system is critical for developmental growth, tissue homeostasis and repair but also for tumor development. Bone morphogenetic protein (BMP) signaling has recently emerged as a fundamental pathway of the endothelium by regulating cardiovascular and lymphatic development and by being causative for several vascular dysfunctions. Two vascular disorders have been directly linked to impaired BMP signaling: pulmonary arterial hypertension and hereditary hemorrhagic telangiectasia. Endothelial BMP signaling critically depends on the cellular context, which includes among others vascular heterogeneity, exposure to flow, and the intertwining with other signaling cascades (Notch, WNT, Hippo and hypoxia). The purpose of this review is to highlight the most recent findings illustrating the clear need for reconsidering the role of BMPs in vascular biology.
Collapse
Affiliation(s)
- Amaya García de Vinuesa
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - An Zwijsen
- VIB Center for the Biology of Disease, Leuven, Belgium; KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Médicale (INSERM, U1036), Grenoble F-38000, France; Commissariat à l'Énergie Atomique et aux Energies Alternatives, Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire Biologie du Cancer et de l'Infection, Grenoble F-38000, France; Université Grenoble-Alpes, Grenoble F-38000, France.
| |
Collapse
|
208
|
Hazzaa HHA, El-Wakeel NM, Attia EAS, Abo Hager EA. ALK1 expression in oral lichen planus: a possible relation to microvessel density. J Oral Pathol Med 2015; 45:373-80. [PMID: 26662187 DOI: 10.1111/jop.12396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2015] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To assess the expression of activin receptor-like kinase 1 (ALK1) and investigate its possible relationship with microvessel density (MVD) in different forms of oral lichen planus (OLP) compared to controls' biopsies. METHODS Biopsies from 20 reticular/papular OLP (R/PLP), 20 atrophic/erosive OLP (A/ELP) patients, and 20 healthy subjects were immunohistochemically analyzed and statistically compared and correlated for ALK1 expression and MVD as assessed by CD34 expression. RESULTS All OLP specimens revealed the presence of positive cytoplasmic CD34 immunostaining in endothelial cells, with statistically high significant MVD in each of R/PLP (Median; M = 4.40) and A/ELP (M = 7.69) compared to controls (M = 1.16) (P < 0.001). Statistically significant MVD was found in A/ELP compared to R/PLP (P < 0.001). All control specimens revealed negative ALK1 immunostaining of the few inflammatory cells found, while 85% of A/ELP cases and 70% of R/PLP cases showed positively immunostained sections for ALK-1, with statistically significant higher ALK1 expression In A/ELP (M = 1.95) compared to R/PLP (M = 0.86) (P = 0.005). No significant correlation between CD34 and ALK1 was detected in R/PLP (r = 0.081), while a barely moderate positive correlation was found in A/ELP (r = 0.396). CONCLUSIONS ALK1 expression and MVD are increased in OLP, particularly in A/ELP type.
Collapse
Affiliation(s)
- Hala H A Hazzaa
- Oral Medicine, Diagnosis, Periodontology and Radiology, Faculty of Dental Medicine, Al Azhar University (Girls Branch), Cairo, Egypt.,Oral Medicine, Diagnosis, Periodontology and Radiology, Faculty of Dentistry, Nahda University, Beni Swaif, Egypt
| | - Naglaa M El-Wakeel
- Oral Medicine, Diagnosis, Periodontology and Radiology, Faculty of Dental Medicine, Al Azhar University (Girls Branch), Cairo, Egypt
| | - Enas A S Attia
- Dermatology, Venereology and Andrology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman A Abo Hager
- Oral and Dental patholology, Faculty of Dental Medicine, Al Azhar University (Girls Branch), Cairo, Egypt
| |
Collapse
|
209
|
Peacock HM, Caolo V, Jones EAV. Arteriovenous malformations in hereditary haemorrhagic telangiectasia: looking beyond ALK1-NOTCH interactions. Cardiovasc Res 2015; 109:196-203. [PMID: 26645978 DOI: 10.1093/cvr/cvv264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/29/2015] [Indexed: 12/20/2022] Open
Abstract
Hereditary haemorrhagic telangiectasia (HHT) is characterized by the development of arteriovenous malformations--enlarged shunts allowing arterial flow to bypass capillaries and enter directly into veins. HHT is caused by mutations in ALK1 or Endoglin; however, the majority of arteriovenous malformations are idiopathic and arise spontaneously. Idiopathic arteriovenous malformations differ from those due to loss of ALK1 in terms of both location and disease progression. Furthermore, while arteriovenous malformations in HHT and Alk1 knockout models have decreased NOTCH signalling, some idiopathic arteriovenous malformations have increased NOTCH signalling. The pathogenesis of these lesions also differs, with loss of ALK1 causing expansion of the shunt through proliferation, and NOTCH gain of function inducing initial shunt enlargement by cellular hypertrophy. Hence, we propose that idiopathic arteriovenous malformations are distinct from those of HHT. In this review, we explore the role of ALK1-NOTCH interactions in the development of arteriovenous malformations and examine a possible role of two signalling pathways downstream of ALK1, TMEM100 and IDs, in the development of arteriovenous malformations in HHT. A nuanced understanding of the precise molecular mechanisms underlying idiopathic and HHT-associated arteriovenous malformations will allow for development of targeted treatments for these lesions.
Collapse
Affiliation(s)
- Hanna M Peacock
- Department of Cardiovascular Science, Centre for Molecular and Vascular Biology, KU Leuven, UZ Herestraat 49-Box 911, 3000 Leuven, Belgium
| | - Vincenza Caolo
- Department of Cardiovascular Science, Centre for Molecular and Vascular Biology, KU Leuven, UZ Herestraat 49-Box 911, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Science, Centre for Molecular and Vascular Biology, KU Leuven, UZ Herestraat 49-Box 911, 3000 Leuven, Belgium
| |
Collapse
|
210
|
Yadin D, Knaus P, Mueller TD. Structural insights into BMP receptors: Specificity, activation and inhibition. Cytokine Growth Factor Rev 2015; 27:13-34. [PMID: 26690041 DOI: 10.1016/j.cytogfr.2015.11.005] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 11/13/2015] [Indexed: 12/29/2022]
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-β family (TGFβ), which signal through hetero-tetrameric complexes of type I and type II receptors. In humans there are many more TGFβ ligands than receptors, leading to the question of how particular ligands can initiate specific signaling responses. Here we review structural features of the ligands and receptors that contribute to this specificity. Ligand activity is determined by receptor-ligand interactions, growth factor prodomains, extracellular modulator proteins, receptor assembly and phosphorylation of intracellular signaling proteins, including Smad transcription factors. Detailed knowledge about the receptors has enabled the development of BMP-specific type I receptor kinase inhibitors. In future these may help to treat human diseases such as fibrodysplasia ossificans progressiva.
Collapse
Affiliation(s)
- David Yadin
- Institute for Chemistry and Biochemistry, Free University Berlin, Institute of Chemistry and Biochemistry, D-14195 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Campus Virchow Klinikum, Augustenburger Platz 1, D-13351 Berlin, Germany.
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Free University Berlin, Institute of Chemistry and Biochemistry, D-14195 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Campus Virchow Klinikum, Augustenburger Platz 1, D-13351 Berlin, Germany.
| | - Thomas D Mueller
- Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute of the University Wuerzburg, Julius-von-Sachs-Platz 2, D-97082 Wuerzburg, Germany.
| |
Collapse
|
211
|
Ormiston ML, Upton PD, Li W, Morrell NW. The promise of recombinant BMP ligands and other approaches targeting BMPR-II in the treatment of pulmonary arterial hypertension. Glob Cardiol Sci Pract 2015; 2015:47. [PMID: 26779522 PMCID: PMC4710869 DOI: 10.5339/gcsp.2015.47] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 08/27/2015] [Indexed: 12/11/2022] Open
Abstract
Human genetic discoveries offer a powerful method to implicate pathways of major importance to disease pathobiology and hence provide targets for pharmacological intervention. The genetics of pulmonary arterial hypertension (PAH) strongly implicates loss-of-function of the bone morphogenetic protein type II receptor (BMPR-II) signalling pathway and moreover implicates the endothelial cell as a central cell type involved in disease initiation. We and others have described several approaches to restore BMPR-II function in genetic and non-genetic forms of PAH. Of these, supplementation of endothelial BMP9/10 signalling with exogenous recombinant ligand has been shown to hold considerable promise as a novel large molecule biopharmaceutical therapy. Here, we describe the mechanism of action and discuss potential additional effects of BMP ligand therapy.
Collapse
Affiliation(s)
- Mark L Ormiston
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, United Kingdom
| | - Paul D Upton
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, United Kingdom
| | - Wei Li
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, United Kingdom
| | - Nicholas W Morrell
- The Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, United Kingdom
| |
Collapse
|
212
|
Animal Models in Studying Cerebral Arteriovenous Malformation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:178407. [PMID: 26649296 PMCID: PMC4663287 DOI: 10.1155/2015/178407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/11/2015] [Accepted: 10/25/2015] [Indexed: 12/13/2022]
Abstract
Brain arteriovenous malformation (AVM) is an important cause of hemorrhagic stroke. The etiology is largely unknown and the therapeutics are controversial. A review of AVM-associated animal models may be helpful in order to understand the up-to-date knowledge and promote further research about the disease. We searched PubMed till December 31, 2014, with the term “arteriovenous malformation,” limiting results to animals and English language. Publications that described creations of AVM animal models or investigated AVM-related mechanisms and treatments using these models were reviewed. More than 100 articles fulfilling our inclusion criteria were identified, and from them eight different types of the original models were summarized. The backgrounds and procedures of these models, their applications, and research findings were demonstrated. Animal models are useful in studying the pathogenesis of AVM formation, growth, and rupture, as well as in developing and testing new treatments. Creations of preferable models are expected.
Collapse
|
213
|
Fujita M, Sakabe M, Ioka T, Watanabe Y, Kinugasa-Katayama Y, Tsuchihashi T, Utset MF, Yamagishi H, Nakagawa O. Pharyngeal arch artery defects and lethal malformations of the aortic arch and its branches in mice deficient for the Hrt1/Hey1 transcription factor. Mech Dev 2015; 139:65-73. [PMID: 26577899 DOI: 10.1016/j.mod.2015.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/09/2015] [Indexed: 11/26/2022]
Abstract
The aortic arch and major branch arteries are formed from the three pairs of pharyngeal arch arteries (PAAs) during embryonic development. Their morphological defects are clinically observed as isolated diseases, as a part of complicated cardiovascular anomalies or as a manifestation of multi-organ syndromes such as 22q11.2 deletion syndrome. Although numerous genes have been implicated in PAA formation and remodeling, detailed mechanisms remain poorly understood. Here we report that the mice null for Hrt1/Hey1, a gene encoding a downstream transcription factor of Notch and ALK1 signaling pathways, show perinatal lethality on the C57BL/6N, C57BL/6N × C57BL/6J or C57BL/6N × 129X1/SvJ background. Hrt1/Hey1 null embryos display abnormal development of the fourth PAA (PAA4), which results in congenital vascular defects including right-sided aortic arch, interruption of the aortic arch and aberrant origin of the right subclavian artery. Impaired vessel formation occurs randomly in PAA4 of Hrt1/Hey1 null embryos, which likely causes the variability of congenital malformations. Endothelial cells in PAA4 of null embryos differentiate normally but are structurally disorganized at embryonic day 10.5 and 11.5. Vascular smooth muscle cells are nearly absent in the structurally-defective PAA4, despite the appropriate migration of cardiac neural crest cells into the fourth pharyngeal arches. Endothelial expression of Jag1 is down-regulated in the structurally-defective PAA4 of null embryos, which may be one of the mechanisms underlying the suppression of vascular smooth muscle cell differentiation. While the direct downstream phenomena of the Hrt1/Hey1 deficiency remain to be clarified, we suggest that Hrt1/Hey1-dependent transcriptional regulation has an important role in PAA formation during embryonic development.
Collapse
Affiliation(s)
- Masahide Fujita
- Laboratory for Cardiovascular System Research, Nara Medical University Advanced Medical Research Center, Kashihara, Nara, Japan; Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Masahide Sakabe
- Laboratory for Cardiovascular System Research, Nara Medical University Advanced Medical Research Center, Kashihara, Nara, Japan; Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Tomoko Ioka
- Laboratory for Cardiovascular System Research, Nara Medical University Advanced Medical Research Center, Kashihara, Nara, Japan; Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yusuke Watanabe
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yumi Kinugasa-Katayama
- Laboratory for Cardiovascular System Research, Nara Medical University Advanced Medical Research Center, Kashihara, Nara, Japan; Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Takatoshi Tsuchihashi
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Manuel F Utset
- Department of Pathology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Hiroyuki Yamagishi
- Division of Pediatric Cardiology, Department of Pediatrics, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Osamu Nakagawa
- Laboratory for Cardiovascular System Research, Nara Medical University Advanced Medical Research Center, Kashihara, Nara, Japan; Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| |
Collapse
|
214
|
Pappalardo A, Porreca I, Caputi L, De Felice E, Schulte-Merker S, Zannini M, Sordino P. Thyroid development in zebrafish lacking Taz. Mech Dev 2015; 138 Pt 3:268-78. [PMID: 26478012 DOI: 10.1016/j.mod.2015.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 10/22/2022]
Abstract
Taz is a signal-responsive transcriptional coregulator implicated in several biological functions, from chondrogenesis to regulation of organ size. Less well studied, however, is its role in thyroid formation. Here, we explored the in vivo effects on thyroid development of morpholino (MO)-mediated knockdown of wwtr1, the gene encoding zebrafish Taz. The wwtr1 gene is expressed in the thyroid primordium and pharyngeal tissue of developing zebrafish. Compared to mammalian cells, in which Taz promotes expression of thyroid transcription factors and thyroid differentiation genes, wwtr1 MO injection in zebrafish had little or no effect on the expression of thyroid transcription factors, and differentially altered the expression of thyroid differentiation genes. Analysis of wwtr1 morphants at later stages of development revealed that the number and the lumen of thyroid follicles, and the number of thyroid follicle cells, were significantly smaller. In addition, Taz-depleted larvae displayed patterning defects in ventral cranial vessels that correlate with lateral displacement of thyroid follicles. These findings indicate that the zebrafish Taz protein is needed for the normal differentiation of the thyroid and are the first to suggest that Taz confers growth advantage to the endocrine gland.
Collapse
Affiliation(s)
- Andrea Pappalardo
- Institute of Experimental Endocrinology and Oncology 'G. Salvatore' - CNR, 80131 Naples, Italy; IRCCS Fondazione Stella Maris, 56018 Calambrone, Pisa, Italy
| | - Immacolata Porreca
- Stazione Zoologica Anton Dohrn, 80121 Naples, Italy; IRGS, Biogem, 83031 Ariano Irpino, Avellino, Italy
| | - Luigi Caputi
- Stazione Zoologica Anton Dohrn, 80121 Naples, Italy
| | | | | | - Mariastella Zannini
- Institute of Experimental Endocrinology and Oncology 'G. Salvatore' - CNR, 80131 Naples, Italy
| | - Paolo Sordino
- Stazione Zoologica Anton Dohrn, 80121 Naples, Italy.
| |
Collapse
|
215
|
Lin S, Xie J, Gong T, Shi S, Zhang T, Fu N, Ye L, Wang M, Lin Y. TGFβ signalling pathway regulates angiogenesis by endothelial cells, in an adipose-derived stromal cell/endothelial cell co-culture 3D gel model. Cell Prolif 2015; 48:729-37. [PMID: 26487556 DOI: 10.1111/cpr.12222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/28/2015] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the role of the TGFβ signalling pathway in angiogenesis in a three-dimensional (3D) collagen gel model, with co-culture between adipose-derived stromal cells (ASCs) and endothelial cells (ECs). MATERIALS AND METHODS A 3D collagen gel, implanted with green fluorescent protein-labelled mouse ASCs and red fluorescent protein-labelled mouse ECs, was established in vitro. Phenomena of angiogenesis with or without type I TGFβ receptor inhibitor (LY2157299) treatment, were observed 7 days post-implantation, using confocal laser scanning microscopy. To detect expression of angiogenesis-related genes, semi-quantitative PCR and quantitative real-time PCR were conducted. Zymography was performed to explore secretion of matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9) after treatment with LY2157299 of 5, 10, 20 to 50 μm concentrations, for 24 h. RESULTS Angiogenesis was found to be attenuated in co-culture gels after ASC and EC treatment with LY2157299. Genes VEGF-A, VEGF-B, VE-ca, FGF-1, FGF-2, PDGF, HGF, BMP-4 were significantly reduced in the presence of LY2157299 in both mono-cultured and co-cultured ECs. Furthermore, reduction in co-cultured ECs was prominent relative to mono-cultured ECs, while the same results did not occur to ASCs. We further confirmed that gelatinases secreted by ECs were reduced in a dose-dependent manner, after treatment with LY2157299. CONCLUSIONS These results indicate that in ASC/EC co-culture, the TGFβ signalling pathway regulated angiogenesis via EC activity. Co-cultured ECs were regulated more significantly than mono-cultured ECs suggesting that inhibition of TGFβRI may regulate paracrine secretion of ASCs to further modulate EC angiogenesis.
Collapse
Affiliation(s)
- Shiyu Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tao Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Na Fu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Min Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
216
|
Shi N, Chen SY. Smooth Muscle Cell Differentiation: Model Systems, Regulatory Mechanisms, and Vascular Diseases. J Cell Physiol 2015; 231:777-87. [DOI: 10.1002/jcp.25208] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Ning Shi
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| | - Shi-You Chen
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| |
Collapse
|
217
|
Costamagna D, Quattrocelli M, van Tienen F, Umans L, de Coo IFM, Zwijsen A, Huylebroeck D, Sampaolesi M. Smad1/5/8 are myogenic regulators of murine and human mesoangioblasts. J Mol Cell Biol 2015; 8:73-87. [PMID: 26450990 PMCID: PMC4710210 DOI: 10.1093/jmcb/mjv059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 06/29/2015] [Indexed: 01/22/2023] Open
Abstract
Mesoangioblasts (MABs) are vessel-associated stem cells that express pericyte marker genes and participate in skeletal muscle regeneration. Molecular circuits that regulate the myogenic commitment of MABs are still poorly characterized. The critical role of bone morphogenetic protein (BMP) signalling during proliferation and differentiation of adult myogenic precursors, such as satellite cells, has recently been established. We evaluated whether BMP signalling impacts on the myogenic potential of embryonic and adult MABs both in vitro and in vivo. Addition of BMP inhibited MAB myogenic differentiation, whereas interference with the interactions between BMPs and receptor complexes induced differentiation. Similarly, siRNA-mediated knockdown of Smad8 in Smad1/5-null MABs or inhibition of SMAD1/5/8 phosphorylation with Dorsomorphin (DM) also improved myogenic differentiation, demonstrating a novel role of SMAD8. Moreover, using a transgenic mouse model of Smad8 deletion, we demonstrated that the absence of SMAD8 protein improved MAB myogenic differentiation. Furthermore, once injected into α-Sarcoglycan (Sgca)-null muscles, DM-treated MABs were more efficacious to restore α-sarcoglycan (αSG) protein levels and re-establish functional muscle properties. Similarly, in acute muscle damage, DM-treated MABs displayed a better myogenic potential compared with BMP-treated and untreated cells. Finally, SMADs also control the myogenic commitment of human MABs (hMABs). BMP signalling antagonists are therefore novel candidates to improve the therapeutic effects of hMABs.
Collapse
Affiliation(s)
- Domiziana Costamagna
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium Laboratory of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Mattia Quattrocelli
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Florence van Tienen
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lieve Umans
- Laboratory for Developmental Signalling, VIB Center for the Biology of Disease, Department of Human Genetics, KU Leuven, Leuven, Belgium Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Irineus F M de Coo
- Department of Neurology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - An Zwijsen
- Laboratory for Developmental Signalling, VIB Center for the Biology of Disease, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Danny Huylebroeck
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven, Belgium Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium Division of Human Anatomy, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
218
|
González-Núñez M, Riolobos AS, Castellano O, Fuentes-Calvo I, de los Ángeles Sevilla M, Oujo B, Pericacho M, Cruz-Gonzalez I, Pérez-Barriocanal F, ten Dijke P, López-Novoa JM. Heterozygous disruption of activin receptor-like kinase 1 is associated with increased arterial pressure in mice. Dis Model Mech 2015; 8:1427-39. [PMID: 26398936 PMCID: PMC4631783 DOI: 10.1242/dmm.019695] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 08/27/2015] [Indexed: 12/20/2022] Open
Abstract
The activin receptor-like kinase 1 (ALK-1) is a type I cell-surface receptor for the transforming growth factor-β (TGF-β) family of proteins. Hypertension is related to TGF-β1, because increased TGF-β1 expression is correlated with an elevation in arterial pressure (AP) and TGF-β expression is upregulated by the renin-angiotensin-aldosterone system. The purpose of this study was to assess the role of ALK-1 in regulation of AP using Alk1 haploinsufficient mice (Alk1(+/-)). We observed that systolic and diastolic AP were significantly higher in Alk1(+/-) than in Alk1(+/+) mice, and all functional and structural cardiac parameters (echocardiography and electrocardiography) were similar in both groups. Alk1(+/-) mice showed alterations in the circadian rhythm of AP, with higher AP than Alk1(+/+) mice during most of the light period. Higher AP in Alk1(+/-) mice is not a result of a reduction in the NO-dependent vasodilator response or of overactivation of the peripheral renin-angiotensin system. However, intracerebroventricular administration of losartan had a hypotensive effect in Alk1(+/-) and not in Alk1(+/+) mice. Alk1(+/-) mice showed a greater hypotensive response to the β-adrenergic antagonist atenolol and higher concentrations of epinephrine and norepinephrine in plasma than Alk1(+/+) mice. The number of brain cholinergic neurons in the anterior basal forebrain was reduced in Alk1(+/-) mice. Thus, we concluded that the ALK-1 receptor is involved in the control of AP, and the high AP of Alk1(+/-) mice is explained mainly by the sympathetic overactivation shown by these animals, which is probably related to the decreased number of cholinergic neurons.
Collapse
Affiliation(s)
- María González-Núñez
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Adela S Riolobos
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain Instituto de Neurociencias de Castilla y León (INCYL), Salamanca 37008, Spain
| | - Orlando Castellano
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain Instituto de Neurociencias de Castilla y León (INCYL), Salamanca 37008, Spain
| | - Isabel Fuentes-Calvo
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | | | - Bárbara Oujo
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Miguel Pericacho
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Ignacio Cruz-Gonzalez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain Departamento de Cardiología, Hospital Universitario de Salamanca, Salamanca 37007, Spain
| | - Fernando Pérez-Barriocanal
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| | - Peter ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Jose M López-Novoa
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca 37007, Spain Unidad de Fisiopatología Renal y Cardiovascular, Instituto 'Reina Sofía' de Investigación Nefrológica, Salamanca 37007, Spain Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca 37007, Spain
| |
Collapse
|
219
|
Kim J, Kim M, Jeong Y, Lee WB, Park H, Kwon JY, Kim YM, Hwang D, Kwon YG. BMP9 Induces Cord Blood-Derived Endothelial Progenitor Cell Differentiation and Ischemic Neovascularization via ALK1. Arterioscler Thromb Vasc Biol 2015; 35:2020-31. [PMID: 26229139 DOI: 10.1161/atvbaha.115.306142] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/20/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Modulating endothelial progenitor cells (EPCs) is essential for therapeutic angiogenesis, and thus various clinical trials involving EPCs are ongoing. However, the identification of environmental conditions and development of optimal methods are required to accelerate EPC-driven vasculogenesis. APPROACH AND RESULTS We evaluated gene expression profiles of cord blood-derived EPCs and endothelial cells to identify the key factors in EPC→endothelial cell differentiation and to show that transforming growth factor-β family members contribute to EPC differentiation. The expression levels of activin receptor-like kinase 1 (ALK1) and its high-affinity ligand, bone morphogenetic protein 9 (BMP9) were markedly changed in EPC→endothelial cell differentiation. Interestingly, BMP9 induced EPC→endothelial cell differentiation and EPC incorporation into vessel-like structures by acting on ALK1 expressed on EPCs in vitro. BMP9 also induced neovascularization in mice with hindlimb ischemia by increasing vessel formation and the incorporation of EPCs into vessels. Conversely, neovascularization was impaired when ALK1 signaling was blocked. Furthermore, EPCs exposed to either short- or long-term BMP9 stimulation demonstrated these functions in EPC-mediated neovascularization. CONCLUSIONS Collectively, our results indicated that BMP9/ALK1 augmented vasculogenesis and angiogenesis, and thereby enhanced neovascularization. Thus, we suggest that BMP9/ALK1 may improve the efficacy of EPC-based therapies for treating ischemic diseases.
Collapse
Affiliation(s)
- Jihye Kim
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Minhyung Kim
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Yoonjeong Jeong
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Wook-Bin Lee
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Hyojin Park
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Ja-Young Kwon
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Young-Myeong Kim
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Daehee Hwang
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.)
| | - Young-Guen Kwon
- From the Department of Biochemistry, College of Life Science and Biotechnology (J.K., Y.J., W.-b.L., H.P., Y.-G.K.) and Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, College of Medicine (J.-Y.K.), Yonsei University, Seoul, Korea; School of Interdisciplinary Biosciences and Bioengineering, Pohang University of Science and Technology, Pohang, Korea (M.K., D.H.); Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea (M.K.); and Department of new Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea (D.H.).
| |
Collapse
|
220
|
Abstract
Formation of arterial vasculature, here termed arteriogenesis, is a central process in embryonic vascular development as well as in adult tissues. Although the process of capillary formation, angiogenesis, is relatively well understood, much remains to be learned about arteriogenesis. Recent discoveries point to the key role played by vascular endothelial growth factor receptor 2 in control of this process and to newly identified control circuits that dramatically influence its activity. The latter can present particularly attractive targets for a new class of therapeutic agents capable of activation of this signaling cascade in a ligand-independent manner, thereby promoting arteriogenesis in diseased tissues.
Collapse
Affiliation(s)
- Michael Simons
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| | - Anne Eichmann
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
221
|
Abstract
Bone morphogenetic proteins (BMPs), together with the eponymous transforming growth factor (TGF) β and the Activins form the TGFβ superfamily of ligands. This protein family comprises more than 30 structurally highly related proteins, which determine formation, maintenance, and regeneration of tissues and organs. Their importance for the development of multicellular organisms is evident from their existence in all vertebrates as well as nonvertebrate animals. From their highly specific functions in vivo either a strict relation between a particular ligand and its cognate cellular receptor and/or a stringent regulation to define a distinct temperospatial expression pattern for the various ligands and receptor is expected. However, only a limited number of receptors are found to serve a large number of ligands thus implicating highly promiscuous ligand-receptor interactions instead. Since in tissues a multitude of ligands are often found, which signal via a highly overlapping set of receptors, this raises the question how such promiscuous interactions between different ligands and their receptors can generate concerted and highly specific cellular signals required during embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Thomas D Mueller
- Department Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
222
|
Alaa el Din F, Patri S, Thoreau V, Rodriguez-Ballesteros M, Hamade E, Bailly S, Gilbert-Dussardier B, Abou Merhi R, Kitzis A. Functional and splicing defect analysis of 23 ACVRL1 mutations in a cohort of patients affected by Hereditary Hemorrhagic Telangiectasia. PLoS One 2015; 10:e0132111. [PMID: 26176610 PMCID: PMC4503601 DOI: 10.1371/journal.pone.0132111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/10/2015] [Indexed: 11/26/2022] Open
Abstract
Hereditary Hemorrhagic Telangiectasia syndrome (HHT) or Rendu-Osler-Weber (ROW) syndrome is an autosomal dominant vascular disorder. Two most common forms of HHT, HHT1 and HHT2, have been linked to mutations in the endoglin (ENG) and activin receptor-like kinase 1 (ACVRL1or ALK1) genes respectively. This work was designed to examine the pathogenicity of 23 nucleotide variations in ACVRL1 gene detected in more than 400 patients. Among them, 14 missense mutations and one intronic variant were novels, and 8 missense mutations were previously identified with questionable implication in HHT2. The functionality of missense mutations was analyzed in response to BMP9 (specific ligand of ALK1), the maturation of the protein products and their localization were analyzed by western blot and fluorescence microscopy. The splicing impairment of the intronic and of two missense mutations was examined by minigene assay. Functional analysis showed that 18 out of 22 missense mutations were defective. Splicing analysis revealed that one missense mutation (c.733A>G, p.Ile245Val) affects the splicing of the harboring exon 6. Similarly, the intronic mutation outside the consensus splicing sites (c.1048+5G>A in intron 7) was seen pathogenic by splicing study. Both mutations induce a frame shift creating a premature stop codon likely resulting in mRNA degradation by NMD surveillance mechanism. Our results confirm the haploinsufficiency model proposed for HHT2. The affected allele of ACVRL1 induces mRNA degradation or the synthesis of a protein lacking the receptor activity. Furthermore, our data demonstrate that functional and splicing analyses together, represent two robust diagnostic tools to be used by geneticists confronted with novel or conflicted ACVRL1 mutations.
Collapse
Affiliation(s)
- Ferdos Alaa el Din
- Genetics of rare diseases, University of Poitiers, Poitiers, France
- Lebanese University Campus Hariri, Faculty of Science / EDST, Hadath, Lebanon
| | - Sylvie Patri
- Genetics of rare diseases, University of Poitiers, Poitiers, France
- Department of Genetics, University Hospital of Poitiers, Poitiers, France
- * E-mail: (SP); (RAM)
| | - Vincent Thoreau
- Genetics of rare diseases, University of Poitiers, Poitiers, France
| | - Montserrat Rodriguez-Ballesteros
- Genetics of rare diseases, University of Poitiers, Poitiers, France
- Department of Genetics, University Hospital of Poitiers, Poitiers, France
| | - Eva Hamade
- Lebanese University Campus Hariri, Faculty of Science / EDST, Hadath, Lebanon
| | | | - Brigitte Gilbert-Dussardier
- Genetics of rare diseases, University of Poitiers, Poitiers, France
- Department of Genetics, University Hospital of Poitiers, Poitiers, France
- Competence Centre of Rendu-Osler, University Hospital of Poitiers, Poitiers, France
| | - Raghida Abou Merhi
- Lebanese University Campus Hariri, Faculty of Science / EDST, Hadath, Lebanon
- * E-mail: (SP); (RAM)
| | - Alain Kitzis
- Genetics of rare diseases, University of Poitiers, Poitiers, France
- Department of Genetics, University Hospital of Poitiers, Poitiers, France
| |
Collapse
|
223
|
Pomeraniec L, Hector-Greene M, Ehrlich M, Blobe GC, Henis YI. Regulation of TGF-β receptor hetero-oligomerization and signaling by endoglin. Mol Biol Cell 2015; 26:3117-27. [PMID: 26157163 PMCID: PMC4551323 DOI: 10.1091/mbc.e15-02-0069] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 06/30/2015] [Indexed: 11/23/2022] Open
Abstract
Endoglin is a modulator of TGF-β signaling in endothelial cells. We show that it forms stable homodimers serving as a scaffold for binding TβRII, ALK5, and ALK1. ALK1 and ALK5 bind endoglin differentially, with TβRII recruiting ALK5. Signaling data indicate a role for this receptor complex in balancing TGF-β signaling between Smad1/5/8 and Smad2/3. Complex formation among transforming growth factor-β (TGF-β) receptors and its modulation by coreceptors represent an important level of regulation for TGF-β signaling. Oligomerization of ALK5 and the type II TGF-β receptor (TβRII) has been thoroughly investigated, both in vitro and in intact cells. However, such studies, especially in live cells, are missing for the endothelial cell coreceptor endoglin and for the ALK1 type I receptor, which enables endothelial cells to respond to TGF-β by activation of both Smad2/3 and Smad1/5/8. Here we combined immunoglobulin G–mediated immobilization of one cell-surface receptor with lateral mobility studies of a coexpressed receptor by fluorescence recovery after photobleaching (FRAP) to demonstrate that endoglin forms stable homodimers that function as a scaffold for binding TβRII, ALK5, and ALK1. ALK1 and ALK5 bind to endoglin with differential dependence on TβRII, which plays a major role in recruiting ALK5 to the complex. Signaling data indicate a role for the quaternary receptor complex in regulating the balance between TGF-β signaling to Smad1/5/8 and to Smad2/3.
Collapse
Affiliation(s)
- Leslie Pomeraniec
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC 27708
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
224
|
The high affinity ALK1-ligand BMP9 induces a hypertrophy-like state in chondrocytes that is antagonized by TGFβ1. Osteoarthritis Cartilage 2015; 23:985-95. [PMID: 25681563 DOI: 10.1016/j.joca.2015.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 01/26/2015] [Accepted: 02/03/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In osteoarthritic cartilage, expression of the receptor ALK1 correlates with markers of deleterious chondrocyte hypertrophy. Recently, bone morphogenetic protein 9 (BMP9) was identified as a high affinity ligand for ALK1. Therefore, we studied if BMP9 signaling results in expression of hypertrophy markers in chondrocytes. Furthermore, because transforming growth factorß1 (TGFβ1) is a well known anti-hypertrophic factor, the interaction between BMP9 and TGFβ1 signaling was also studied. DESIGN Primary chondrocytes were isolated from bovine cartilage and stimulated with BMP9 and/or TGFβ1 to measure intracellular signaling via pSmads with the use of Western blot. Expression of Smad-responsive genes or hypertrophy-marker genes was measured using qPCR. To confirm observations on TGFβ/Smad3 responsive genes, a Smad3-dependent CAGA12-luc transcriptional reporter assay was performed in the chondrocyte G6 cell line. RESULTS In primary chondrocytes, BMP9 potently induced phosphorylation of Smad1/5 and Smad2 to a lesser extent. BMP9-induced Smad1/5 phosphorylation was rapidly (2 h) reflected in gene expression, whereas Smad2 phosphorylation was not. Remarkably, BMP9 and TGFβ1 dose-dependently synergized on Smad2 phosphorylation, and showed an additive effect on expression of Smad3-dependent genes like bSerpine1 after 24 h. The activation of the TGFβ/Smad3 signaling cascade was confirmed using the CAGA12-luc transcriptional reporter. BMP9 selectively induced bAlpl and bColX expression, which are considered early markers of cellular hypertrophy, but this was potently antagonized by addition of a low dose of TGFβ1. CONCLUSIONS This study shows that in vitro in chondrocytes, BMP9 potently induces pSmad1/5 and a chondrocyte hypertrophy-like state, which is potently blocked by TGFβ1. This observation underlines the importance of TGFβ1 in maintenance of chondrocyte phenotype.
Collapse
|
225
|
Fan Y, Li X, Xiao W, Fu J, Harris RC, Lindenmeyer M, Cohen CD, Guillot N, Baron MH, Wang N, Lee K, He JC, Schlondorff D, Chuang PY. BAMBI elimination enhances alternative TGF-β signaling and glomerular dysfunction in diabetic mice. Diabetes 2015; 64:2220-33. [PMID: 25576053 PMCID: PMC4439561 DOI: 10.2337/db14-1397] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/02/2015] [Indexed: 01/11/2023]
Abstract
BMP, activin, membrane-bound inhibitor (BAMBI) acts as a pseudo-receptor for the transforming growth factor (TGF)-β type I receptor family and a negative modulator of TGF-β kinase signaling, and BAMBI(-/-) mice show mild endothelial dysfunction. Because diabetic glomerular disease is associated with TGF-β overexpression and microvascular alterations, we examined the effect of diabetes on glomerular BAMBI mRNA levels. In isolated glomeruli from biopsies of patients with diabetic nephropathy and in glomeruli from mice with type 2 diabetes, BAMBI was downregulated. We then examined the effects of BAMBI deletion on streptozotocin-induced diabetic glomerulopathy in mice. BAMBI(-/-) mice developed more albuminuria, with a widening of foot processes, than BAMBI(+/+) mice, along with increased activation of alternative TGF-β pathways such as extracellular signal-related kinase (ERK)1/2 and Smad1/5 in glomeruli and cortices of BAMBI(-/-) mice. Vegfr2 and Angpt1, genes controlling glomerular endothelial stability, were downmodulated in glomeruli from BAMBI(-/-) mice with diabetes. Incubation of glomeruli from nondiabetic BAMBI(+/+) or BAMBI(-/-) mice with TGF-β resulted in the downregulation of Vegfr2 and Angpt1, effects that were more pronounced in BAMBI(-/-) mice and were prevented by a MEK inhibitor. The downregulation of Vegfr2 in diabetes was localized to glomerular endothelial cells using a histone yellow reporter under the Vegfr2 promoter. Thus, BAMBI modulates the effects of diabetes on glomerular permselectivity in association with altered ERK1/2 and Smad1/5 signaling. Future therapeutic interventions with inhibitors of alternative TGF-β signaling may therefore be of interest in diabetic nephropathy.
Collapse
Affiliation(s)
- Ying Fan
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xuezhu Li
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Wenzhen Xiao
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jia Fu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ray C Harris
- Vanderbilt University School of Medicine, Nashville, TN
| | - Maja Lindenmeyer
- Division of Nephrology and Institute of Physiology with Center of Integrative Human Physiology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Clemens D Cohen
- Division of Nephrology and Institute of Physiology with Center of Integrative Human Physiology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Nicolas Guillot
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Margaret H Baron
- Tisch Cancer Institute and Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Kyung Lee
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY Renal Section, James J. Peter Veterans Administration Medical Center, Bronx, NY
| | - Detlef Schlondorff
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Peter Y Chuang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
226
|
Jerkic M, Letarte M. Increased endothelial cell permeability in endoglin-deficient cells. FASEB J 2015; 29:3678-88. [PMID: 25972355 DOI: 10.1096/fj.14-269258] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 05/04/2015] [Indexed: 01/12/2023]
Abstract
Endoglin (ENG) is a TGF-β superfamily coreceptor essential for vascular endothelium integrity. ENG mutations lead to a vascular dysplasia associated with frequent hemorrhages in multiple organs, whereas ENG null mouse embryos die at midgestation with impaired heart development and leaky vasculature. ENG interacts with several proteins involved in cell adhesion, and we postulated that it regulates vascular permeability. The current study assessed the permeability of ENG homozygous null (Eng(-/-)), heterozygous (Eng(+/-)), and normal (Eng(+/+)) mouse embryonic endothelial cell (EC) lines. Permeability, measured by passage of fluorescent dextran through EC monolayers, was increased 2.9- and 1.7-fold for Eng(-/-) and Eng(+/-) ECs, respectively, compared to control ECs and was not increased by TGF-β1 or VEGF. Prolonged starvation increased Eng(-/-) EC permeability by 3.7-fold with no effect on control ECs; neutrophils transmigrated faster through Eng(-/-) than Eng(+/+) monolayers. Using a pull-down assay, we demonstrate that Ras homolog gene family (Rho) A is constitutively active in Eng(-/-) and Eng(+/-) ECs. We show that the endothelial barrier destabilizing factor thrombospondin-1 and its receptor-like protein tyrosine phosphatase are increased, whereas stabilizing factors VEGF receptor 2, vascular endothelial-cadherin, p21-activated kinase, and Ras-related C3 botulinum toxin substrate 2 are decreased in Eng(-/-) cells. Our findings indicate that ENG deficiency leads to EC hyperpermeability through constitutive activation of RhoA and destabilization of endothelial barrier function.
Collapse
Affiliation(s)
- Mirjana Jerkic
- *Molecular Structure and Function Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and Department of Immunology and Keenan Research Centre for Biomedical Science, Anesthesia Research, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Michelle Letarte
- *Molecular Structure and Function Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and Department of Immunology and Keenan Research Centre for Biomedical Science, Anesthesia Research, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
227
|
Makker V, Filiaci VL, Chen LM, Darus CJ, Kendrick JE, Sutton G, Moxley K, Aghajanian C. Phase II evaluation of dalantercept, a soluble recombinant activin receptor-like kinase 1 (ALK1) receptor fusion protein, for the treatment of recurrent or persistent endometrial cancer: an NRG Oncology/Gynecologic Oncology Group Study 0229N. Gynecol Oncol 2015; 138:24-9. [PMID: 25888978 DOI: 10.1016/j.ygyno.2015.04.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/08/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE This two-stage phase II study assessed activity of single agent dalantercept in patients with recurrent/persistent endometrial carcinoma (EMC). METHODS Eligible patients had persistent/recurrent EMC after 1-2 prior cytotoxic regimens, measurable disease (RECIST 1.1), and GOG performance≤2. Dalantercept 1.2mg/kg subcutaneous was administered once every 3weeks until disease progression (PD)/development of prohibitory toxicity. Primary objectives were to estimate the proportion of patients with persistent/recurrent EMC, who survive progression-free without receiving non-protocol therapy (TPFS) for at least 6months and to estimate the proportion having objective tumor response. RESULTS All 28 enrolled patients were eligible and evaluable. Median age: 62years. Most common histologies: 32% Grade 1/2 endometrioid and 54% serous tumors. Prior treatment: 1 or 2 regimens in 82% and 18% of patients, respectively. Eighteen patients received prior radiation therapy. Patients received 1-12 cycles of dalantercept, and 46% of patients received ≤2cycles. The most common adverse events (AE) were fatigue, anemia, constipation and peripheral edema. Grade 3/4 AEs occurred in 39% and 4% of patients. One grade 5 gastric hemorrhage in a patient with a history of radiation fibrosis/small bowel obstruction was deemed possibly dalantercept-related. All patients are off study: 86% for PD. No ORs were observed; 57% had stable disease and 11% had TPFS>6 mos. Median progression-free and overall survival: 2.1months (90% CI: 1.4-3.2) and 14.5months (90% CI: 7.0-17.5), respectively. CONCLUSIONS Dalantercept has insufficient single agent activity in recurrent EMC to warrant further investigation at this dose level and schedule.
Collapse
Affiliation(s)
- Vicky Makker
- Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| | - Virginia L Filiaci
- NRG Oncology Statistics and Data Management Center, Roswell Park Cancer Institute, Buffalo, NY 14263, United States
| | - Lee-May Chen
- Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA 94115, United States
| | - Christopher J Darus
- Gynecologic Oncology, Maine Medical Center, Scarborough, ME 04074, United States
| | - James E Kendrick
- Gynecologic Oncology, Florida Hospital Cancer Institute Orlando, FL 32804, United States
| | - Gregory Sutton
- Gynecologic Oncology, St. Vincent Hospitals and Health Services, Indianapolis, IN 46269, United States
| | - Katherine Moxley
- Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Carol Aghajanian
- Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| |
Collapse
|
228
|
Lin S, Leonard D, Co MAM, Mukhopadhyay D, Giri B, Perger L, Beeram MR, Kuehl TJ, Uddin MN. Pre-eclampsia has an adverse impact on maternal and fetal health. Transl Res 2015; 165:449-63. [PMID: 25468481 DOI: 10.1016/j.trsl.2014.10.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/08/2014] [Accepted: 10/09/2014] [Indexed: 12/15/2022]
Abstract
Pre-eclampsia (preE) is a multifaceted complication found uniquely in the pregnant patient and one that has puzzled scientists for years. PreE is not a single disorder, but a complex syndrome that is produced by various pathophysiological triggers and mechanisms affecting about 5% of obstetrical patients. PreE is a major cause of premature delivery and maternal and fetal morbidity and mortality. PreE is characterized by de novo development of hypertension and proteinuria after 20 weeks of gestation and affects nearly every organ system, with the most severe consequences being eclampsia, pulmonary edema, intrauterine growth restriction, and thrombocytopenia. PreE alters the intrauterine environment by modulating the pattern of hormonal signals and activating the detrimental cellular signaling that has been transported to the fetus. The fetus has to adapt to this intrauterine environment with detrimental signals. The adaptive changes increase the risk of disease later in life. This review defines the predisposition and causes of preE and the cellular signaling detrimental to maternal health during preE. Moreover, the risk factors for diseases that are transmitted to the offspring have been addressed in this review. The detrimental signaling molecules that have been overexpressed in preE patients raises the possibility that those signals could be therapeutically blocked one day.
Collapse
Affiliation(s)
- Saunders Lin
- Texas A&M University College of Medicine, Temple, Tex
| | | | - Mary A M Co
- Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Dhriti Mukhopadhyay
- Department of Surgery, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Badri Giri
- Department of Pulmonary Critical Care, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Lena Perger
- Department of Surgery, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Madhava R Beeram
- Texas A&M University College of Medicine, Temple, Tex; Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Thomas J Kuehl
- Texas A&M University College of Medicine, Temple, Tex; Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex; Department of Obstetrics and Gynecology, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex
| | - Mohammad N Uddin
- Texas A&M University College of Medicine, Temple, Tex; Prehealth Studies, Baylor University, Waco, Tex; Department of Pediatrics, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex; Department of Obstetrics and Gynecology, Scott & White Healthcare and Texas A&M Health Science Center College of Medicine, Temple, Tex.
| |
Collapse
|
229
|
Herrer I, Roselló-Lletí E, Ortega A, Tarazón E, Molina-Navarro MM, Triviño JC, Martínez-Dolz L, Almenar L, Lago F, Sánchez-Lázaro I, González-Juanatey JR, Salvador A, Portolés M, Rivera M. Gene expression network analysis reveals new transcriptional regulators as novel factors in human ischemic cardiomyopathy. BMC Med Genomics 2015; 8:14. [PMID: 25884818 PMCID: PMC4386080 DOI: 10.1186/s12920-015-0088-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 03/10/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Ischemic cardiomyopathy (ICM) is characterized by transcriptomic changes that alter cellular processes leading to decreased cardiac output. Because the molecular network of ICM is largely unknown, the aim of this study was to characterize the role of new transcriptional regulators in the molecular mechanisms underlying the responses to ischemia. METHODS Myocardial tissue explants from ICM patients and control (CNT) subjects were analyzed by RNA-Sequencing (RNA-Seq) and quantitative Real-Time PCR. RESULTS Enrichment analysis of the ICM transcriptomic profile allowed the characterization of novel master regulators. We found that the expression of the transcriptional regulators SP100 (-1.5-fold, p < 0.05), CITED2 (-3.8-fold, p < 0.05), CEBPD (-4.9-fold, p < 0.05) and BCL3 (-3.3-fold, p < 0.05) were lower in ICM than in CNT. To gain insights into the molecular network defined by the transcription factors, we identified CEBPD, BCL3, and HIF1A target genes in the RNA-Seq datasets. We further characterized the biological processes of the target genes by gene ontology annotation. Our results suggest that CEBPD-inducible genes with roles in the inhibition of apoptosis are downregulated and that BCL3-repressible genes are involved in the regulation of cellular metabolism in ICM. Moreover, our results suggest that CITED2 downregulation causes increased expression of HIF1A target genes. Functional analysis of HIF1A target genes revealed that hypoxic and stress response genes are activated in ICM. Finally, we found a significant correlation between the mRNA levels of BCL3 and the mRNA levels of both CEBPD (r = 0.73, p < 0.001) and CITED2 (r = 0.56, p < 0.05). Interestingly, CITED2 mRNA levels are directly related to ejection fraction (EF) (r = 0.54, p < 0.05). CONCLUSIONS Our data indicate that changes in the expression of SP100, CITED2, CEBPD, and BCL3 affect their transcription regulatory networks, which subsequently alter a number of biological processes in ICM patients. The relationship between CITED2 mRNA levels and EF emphasizes the importance of this transcription factor in ICM. Moreover, our findings identify new mechanisms used to interpret gene expression changes in ICM and provide valuable resources for further investigation of the molecular basis of human cardiac ischemic response.
Collapse
Affiliation(s)
- Isabel Herrer
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Esther Roselló-Lletí
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Ana Ortega
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Estefanía Tarazón
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - María Micaela Molina-Navarro
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | | | - Luis Martínez-Dolz
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - Luis Almenar
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - Francisca Lago
- Cellular and Molecular Cardiology Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago Compostela, Spain.
| | - Ignacio Sánchez-Lázaro
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago Compostela, Spain.
| | - Antonio Salvador
- Heart Failure and Transplantation Unit, Cardiology Department, La Fe University Hospital, Valencia, Spain.
| | - Manuel Portolés
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute of La Fe University Hospital, Avd de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| |
Collapse
|
230
|
Wöltje K, Jabs M, Fischer A. Serum induces transcription of Hey1 and Hey2 genes by Alk1 but not Notch signaling in endothelial cells. PLoS One 2015; 10:e0120547. [PMID: 25799559 PMCID: PMC4370690 DOI: 10.1371/journal.pone.0120547] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 01/23/2015] [Indexed: 01/10/2023] Open
Abstract
The transcriptional repressors Hey1 and Hey2 are primary target genes of Notch signaling in the cardiovascular system and induction of Hey gene expression is often interpreted as activation of Notch signaling. Here we report that treatment of primary human endothelial cells with serum or fresh growth medium led to a strong wave of Hey1 and Hey2 transcription lasting for approximately three hours. Transcription of other Notch target genes (Hes1, Hes5, ephrinB2, Dll4) was however not induced by serum in endothelial cells. Gamma secretase inhibition or expression of dominant-negative MAML1 did not prevent the induction of Hey genes indicating that canonical Notch signaling is dispensable. Pretreatment with soluble BMP receptor Alk1, but not Alk3, abolished Hey gene induction by serum. Consequently, the Alk1 ligand BMP9 stimulated Hey gene induction in endothelial cells. Several other cell types however did not show such a strong BMP signaling and consequently only a very mild induction of Hey genes. Taken together, the experiments revealed that bone morphogenic proteins within the serum of cell culture medium are potent inducers of endothelial Hey1 and Hey2 gene expression within the first few hours after medium change.
Collapse
Affiliation(s)
- Kerstin Wöltje
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ-ZMBH Alliance), D-69120, Heidelberg, Germany
| | - Markus Jabs
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ-ZMBH Alliance), D-69120, Heidelberg, Germany
| | - Andreas Fischer
- Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ-ZMBH Alliance), D-69120, Heidelberg, Germany
- Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, D-68167, Mannheim, Germany
- Department of Medicine I and Clinical Chemistry, Heidelberg University, D-69120, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
231
|
Dingenouts CKE, Goumans MJ, Bakker W. Mononuclear cells and vascular repair in HHT. Front Genet 2015; 6:114. [PMID: 25852751 PMCID: PMC4369645 DOI: 10.3389/fgene.2015.00114] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/05/2015] [Indexed: 12/31/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) or Rendu–Osler–Weber disease is a rare genetic vascular disorder known for its endothelial dysplasia causing arteriovenous malformations and severe bleedings. HHT-1 and HHT-2 are the most prevalent variants and are caused by heterozygous mutations in endoglin and activin receptor-like kinase 1, respectively. An undervalued aspect of the disease is that HHT patients experience persistent inflammation. Although endothelial and mural cells have been the main research focus trying to unravel the mechanism behind the disease, wound healing is a process with a delicate balance between inflammatory and vascular cells. Inflammatory cells are part of the mononuclear cells (MNCs) fraction, and can, next to eliciting an immune response, also have angiogenic potential. This biphasic effect of MNC can hold a promising mechanism to further elucidate treatment strategies for HHT patients. Before MNC are able to contribute to repair, they need to home to and retain in ischemic and damaged tissue. Directed migration (homing) of MNCs following tissue damage is regulated by the stromal cell derived factor 1 (SDF1). MNCs that express the C-X-C chemokine receptor 4 (CXCR4) migrate toward the tightly regulated gradient of SDF1. This directed migration of monocytes and lymphocytes can be inhibited by dipeptidyl peptidase 4 (DPP4). Interestingly, MNC of HHT patients express elevated levels of DPP4 and show impaired homing toward damaged tissue. Impaired homing capacity of the MNCs might therefore contribute to the impaired angiogenesis and tissue repair observed in HHT patients. This review summarizes recent studies regarding the role of MNCs in the etiology of HHT and vascular repair, and evaluates the efficacy of DPP4 inhibition in tissue integrity and repair.
Collapse
Affiliation(s)
- Calinda K E Dingenouts
- Department of Molecular Cell Biology, Leiden University Medical Center Leiden, Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center Leiden, Netherlands
| | - Wineke Bakker
- Department of Molecular Cell Biology, Leiden University Medical Center Leiden, Netherlands
| |
Collapse
|
232
|
P7170: A Novel Molecule with Unique Profile of mTORC1/C2 and Activin Receptor-like Kinase 1 Inhibition Leading to Antitumor and Antiangiogenic Activity. Mol Cancer Ther 2015; 14:1095-106. [DOI: 10.1158/1535-7163.mct-14-0486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 01/18/2015] [Indexed: 11/16/2022]
|
233
|
Tual-Chalot S, Oh SP, Arthur HM. Mouse models of hereditary hemorrhagic telangiectasia: recent advances and future challenges. Front Genet 2015; 6:25. [PMID: 25741358 PMCID: PMC4332371 DOI: 10.3389/fgene.2015.00025] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/19/2015] [Indexed: 12/15/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a genetic disorder characterized by a multi-systemic vascular dysplasia and hemorrhage. The precise factors leading to these vascular malformations are not yet understood and robust animal models of HHT are essential to gain a detailed understanding of the molecular and cellular events that lead to clinical symptoms, as well as to test new therapeutic modalities. Most cases of HHT are caused by mutations in either endoglin (ENG) or activin receptor-like kinase 1 (ACVRL1, also known as ALK1). Both genes are associated with TGFβ/BMP signaling, and loss of function mutations in the co-receptor ENG are causal in HHT1, while HHT2 is associated with mutations in the signaling receptor ACVRL1. Significant advances in mouse genetics have provided powerful ways to study the function of Eng and Acvrl1 in vivo, and to generate mouse models of HHT disease. Mice that are null for either Acvrl1 or Eng genes show embryonic lethality due to major defects in angiogenesis and heart development. However mice that are heterozygous for mutations in either of these genes develop to adulthood with no effect on survival. Although these heterozygous mice exhibit selected vascular phenotypes relevant to the clinical pathology of HHT, the phenotypes are variable and generally quite mild. An alternative approach using conditional knockout mice allows us to study the effects of specific inactivation of either Eng or Acvrl1 at different times in development and in different cell types. These conditional knockout mice provide robust and reproducible models of arteriovenous malformations, and they are currently being used to unravel the causal factors in HHT pathologies. In this review, we will summarize the strengths and limitations of current mouse models of HHT, discuss how knowledge obtained from these studies has already informed clinical care and explore the potential of these models for developing improved treatments for HHT patients in the future.
Collapse
Affiliation(s)
| | - S Paul Oh
- Department of Physiology and Functional Genomics, University of Florida , Gainesville, FL, USA
| | - Helen M Arthur
- Institute of Genetic Medicine, Newcastle University , Newcastle, UK
| |
Collapse
|
234
|
Thalgott J, Dos-Santos-Luis D, Lebrin F. Pericytes as targets in hereditary hemorrhagic telangiectasia. Front Genet 2015; 6:37. [PMID: 25763012 PMCID: PMC4327729 DOI: 10.3389/fgene.2015.00037] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/26/2015] [Indexed: 12/04/2022] Open
Abstract
Defective paracrine Transforming Growth Factor-β (TGF-β) signaling between endothelial cells and the neighboring mural cells have been thought to lead to the development of vascular lesions that are characteristic of Hereditary Hemorrhagic Telangiectasia (HHT). This review highlights recent progress in our understanding of TGF-β signaling in mural cell recruitment and vessel stabilization and how perturbed TGF-β signaling might contribute to defective endothelial-mural cell interaction affecting vessel functionalities. Our recent findings have provided exciting insights into the role of thalidomide, a drug that reduces both the frequency and the duration of epistaxis in individuals with HHT by targeting mural cells. These advances provide opportunities for the development of new therapies for vascular malformations.
Collapse
Affiliation(s)
- Jérémy Thalgott
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| | - Damien Dos-Santos-Luis
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| | - Franck Lebrin
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| |
Collapse
|
235
|
Ardelean DS, Letarte M. Anti-angiogenic therapeutic strategies in hereditary hemorrhagic telangiectasia. Front Genet 2015; 6:35. [PMID: 25717337 PMCID: PMC4324154 DOI: 10.3389/fgene.2015.00035] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/26/2015] [Indexed: 01/22/2023] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular dysplastic disorder, characterized by recurrent nosebleeds (epistaxis), multiple telangiectases and arteriovenous malformations (AVMs) in major organs. Mutations in Endoglin (ENG or CD105) and Activin receptor-like kinase 1 (ACVRL1 or ALK1) genes of the TGF-β superfamily receptors are responsible for HHT1 and HHT2 respectively and account for the majority of HHT cases. Haploinsufficiency in ENG and ALK1 is recognized at the underlying cause of HHT. However, the mechanisms responsible for the predisposition to and generation of AVMs, the hallmark of this disease, are poorly understood. Recent data suggest that dysregulated angiogenesis contributes to the pathogenesis of HHT and that the vascular endothelial growth factor, VEGF, may be implicated in this disease, by modulating the angiogenic–angiostatic balance in the affected tissues. Hence, anti-angiogenic therapies that target the abnormal vessels and restore the angiogenic–angiostatic balance are candidates for treatment of HHT. Here we review the experimental evidence for dysregulated angiogenesis in HHT, the anti-angiogenic therapeutic strategies used in animal models and some patients with HHT and the potential benefit of the anti-angiogenic treatment for ameliorating this severe, progressive vascular disease.
Collapse
Affiliation(s)
- Daniela S Ardelean
- Department of Pediatrics, The Hospital for Sick Children Toronto, ON, Canada
| | - Michelle Letarte
- Molecular Structure and Function Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children Toronto, ON, Canada ; Department of Immunology, University of Toronto Toronto, ON, Canada ; Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto Toronto, ON, Canada
| |
Collapse
|
236
|
Houlahan KE, Prokopec SD, Moffat ID, Lindén J, Lensu S, Okey AB, Pohjanvirta R, Boutros PC. Transcriptional profiling of rat hypothalamus response to 2,3,7,8-tetrachlorodibenzo- ρ -dioxin. Toxicology 2015; 328:93-101. [DOI: 10.1016/j.tox.2014.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 12/12/2014] [Accepted: 12/16/2014] [Indexed: 12/21/2022]
|
237
|
Xing C, Gong B, Xue Y, Han Y, Wang Y, Meng A, Jia S. TGFβ1a regulates zebrafish posterior lateral line formation via Smad5 mediated pathway. J Mol Cell Biol 2015; 7:48-61. [PMID: 25603803 DOI: 10.1093/jmcb/mjv004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The zebrafish sensory posterior lateral line (pLL) has become an attractive model for studying collective cell migration and cell morphogenesis. Recent studies have indicated that chemokine, Wnt/β-catenin, Fgf, and Delta-Notch signaling pathways participate in regulating pLL development. However, it remains unclear whether TGFβ signaling pathway is involved in pLL development. Here we report a critical role of TGFβ1 in regulating morphogenesis of the pLL primordium (pLLP). The tgfβ1a gene is abundantly expressed in the lateral line primordium. Knockdown or knockout of tgfβ1a leads to a reduction of neuromast number, an increase of inter-neuromast distance, and a reduced number of hair cells. The aberrant morphogenesis in embryos depleted of tgfβ1a correlates with the reduced expression of atoh1a, deltaA, and n-cadherin/cdh2, which are known important regulators of the pLLP morphogenesis. Like tgfβ1a depletion, knockdown of smad5 that expresses in the pLLP, affects pLLP development whereas overexpression of a constitutive active Smad5 isoform rescues the defects in embryos depleted of tgfβ1a, indicating that Smad5 mediates tgfβ1a function in pLLP development. Therefore, TGFβ/Smad5 signaling plays an important role in the zebrafish lateral line formation.
Collapse
Affiliation(s)
- Cencan Xing
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bo Gong
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yu Xue
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanchao Han
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yixia Wang
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Anming Meng
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shunji Jia
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
238
|
Tillet E, Bailly S. Emerging roles of BMP9 and BMP10 in hereditary hemorrhagic telangiectasia. Front Genet 2015; 5:456. [PMID: 25620979 PMCID: PMC4288046 DOI: 10.3389/fgene.2014.00456] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/12/2014] [Indexed: 12/21/2022] Open
Abstract
Rendu-Osler-Weber syndrome, also known as hereditary hemorrhagic telangiectasia (HHT), is an autosomal dominant vascular disorder. Three genes are causally related to HHT: the ENG gene encoding endoglin, a co-receptor of the TGFβ family (HHT1), the ACVRL1 gene encoding ALK1 (activin receptor-like kinase 1), a type I receptor of the TGFβ family (HHT2), and the SMAD4 gene, encoding a transcription factor critical for this signaling pathway. Bone morphogenetic proteins (BMPs) are growth factors of the TGFβ family. Among them, BMP9 and BMP10 have been shown to bind directly with high affinity to ALK1 and endoglin, and BMP9 mutations have recently been linked to a vascular anomaly syndrome that has phenotypic overlap with HHT. BMP9 and BMP10 are both circulating cytokines in blood, and the current working model is that BMP9 and BMP10 maintain a quiescent endothelial state that is dependent on the level of ALK1/endoglin activation in endothelial cells. In accordance with this model, to explain the etiology of HHT we hypothesize that a deficient BMP9/BMP10/ALK1/endoglin pathway may lead to re-activation of angiogenesis or a greater sensitivity to an angiogenic stimulus. Resulting endothelial hyperproliferation and hypermigration may lead to vasodilatation and generation of an arteriovenous malformation (AVM). HHT would thus result from a defect in the angiogenic balance. This review will focus on the emerging role played by BMP9 and BMP10 in the development of this disease and the therapeutic approaches that this opens.
Collapse
Affiliation(s)
- Emmanuelle Tillet
- Inserm, U1036 , Grenoble, France ; Laboratoire Biologie du Cancer et de l'Infection, Institut de Recherches en Technologies et Sciences pour le Vivant, Commissariat à l'énergie atomique et aux énergies alternatives , Grenoble, France ; Université Grenoble-Alpes , Grenoble, France
| | - Sabine Bailly
- Inserm, U1036 , Grenoble, France ; Laboratoire Biologie du Cancer et de l'Infection, Institut de Recherches en Technologies et Sciences pour le Vivant, Commissariat à l'énergie atomique et aux énergies alternatives , Grenoble, France ; Université Grenoble-Alpes , Grenoble, France
| |
Collapse
|
239
|
|
240
|
Moon EH, Kim YS, Seo J, Lee S, Lee YJ, Oh SP. Essential role for TMEM100 in vascular integrity but limited contributions to the pathogenesis of hereditary haemorrhagic telangiectasia. Cardiovasc Res 2014; 105:353-60. [PMID: 25538155 DOI: 10.1093/cvr/cvu260] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIMS TMEM100 was previously identified as a downstream target of activin receptor-like kinase 1 (ALK1; ACVRL1) signalling. Mutations on ALK1 cause hereditary haemorrhagic telangiectasia (HHT), a vascular disorder characterized by mucocutaneous telangiectases and visceral arteriovenous malformations (AVMs). The aims of this study are to investigate the in vivo role of TMEM100 at various developmental and adult stages and to determine the extent to which TMEM100 contributed to the development of AVMs as a key downstream effector of ALK1. METHODS AND RESULTS Blood vasculature in Tmem100-null embryos and inducible Tmem100-null neonatal and adult mice was examined. We found that TMEM100 deficiency resulted in cardiovascular defects at embryonic stage; dilated vessels, hyperbranching, and increased number of filopodia in the retinal vasculature at neonatal stage; and various vascular abnormalities, including internal haemorrhage, arteriovenous shunts, and weakening of vasculature with abnormal elastin layers at adult stage. However, arteriovenous shunts in adult mutant mice appeared to be underdeveloped without typical tortuosity of vessels associated with AVMs. We uncovered that the expression of genes encoding cell adhesion and extracellular matrix proteins was significantly affected in lungs of adult mutant mice. Especially Mfap4, which is associated with elastin fibre formation, was mostly down-regulated. CONCLUSION These results demonstrate that TMEM100 has essential functions for the maintenance of vascular integrity as well as the formation of blood vessels. Our results also indicate that down-regulation of Tmem100 is not the central mechanism of HHT pathogenesis, but it may contribute to the development of vascular pathology of HHT by weakening vascular integrity.
Collapse
Affiliation(s)
- Eun-Hye Moon
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Yoo Sung Kim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Jiyoung Seo
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Sabin Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Young Jae Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Suk Paul Oh
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea Department of Physiology and Functional Genomics, College of Medicine, University of Florida, 1600 SW Archer Road, Room CG-20B, Gainesville, FL 32610, USA
| |
Collapse
|
241
|
Roman BL, Finegold DN. Genetic and Molecular Basis for Hereditary Hemorrhagic Telangiectasia. CURRENT GENETIC MEDICINE REPORTS 2014. [DOI: 10.1007/s40142-014-0061-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
242
|
Chen L, Hou X, Xiao J, Kuroda J, Ago T, Sadoshima J, Cohen RA, Tong X. Both hydrogen peroxide and transforming growth factor beta 1 contribute to endothelial Nox4 mediated angiogenesis in endothelial Nox4 transgenic mouse lines. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2489-99. [DOI: 10.1016/j.bbadis.2014.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/14/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
|
243
|
Rivas V, Nogués L, Reglero C, Mayor F, Penela P. Role of G protein-coupled receptor kinase 2 in tumoral angiogenesis. Mol Cell Oncol 2014; 1:e969166. [PMID: 27308373 PMCID: PMC4905215 DOI: 10.4161/23723548.2014.969166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/22/2014] [Accepted: 08/23/2014] [Indexed: 12/26/2022]
Abstract
Downregulation of G protein-coupled receptor kinase 2 (GRK2) in endothelial cells has recently been identified as a relevant event in the tumoral angiogenic switch. Based on the effects of altering GRK2 dosage in cell and animal models, this kinase appears to act as a hub in key signaling pathways involved in vascular stabilization and remodeling. Accordingly, decreased GRK2 expression in endothelial cells accelerates tumor growth in mice by impairing the pericytes ensheathing the vessels, thereby promoting hypoxia and macrophage infiltration. These results raise new questions regarding the mechanisms by which transformed cells trigger the decrease in GRK2 observed in human breast cancer vessels and how GRK2 modulates the interactions between different cell types that occur in the tumor microenvironment.
Collapse
Affiliation(s)
- Verónica Rivas
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Laura Nogués
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Clara Reglero
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Petronila Penela
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| |
Collapse
|
244
|
Mizuta K, Sakabe M, Hashimoto A, Ioka T, Sakai C, Okumura K, Hattammaru M, Fujita M, Araki M, Somekawa S, Saito Y, Nakagawa O. Impairment of endothelial-mesenchymal transformation during atrioventricular cushion formation inTmem100null embryos. Dev Dyn 2014; 244:31-42. [DOI: 10.1002/dvdy.24216] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/28/2014] [Accepted: 10/08/2014] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ken Mizuta
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Masahide Sakabe
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Aya Hashimoto
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Tomoko Ioka
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Chihiro Sakai
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Kazuki Okumura
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Miwa Hattammaru
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
- Department of Internal Medicine; Tokyo Women's Medical University Medical Center East; Tokyo Japan
| | - Masahide Fujita
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Mutsumi Araki
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| | - Satoshi Somekawa
- The First Department of Internal Medicine; Nara Medical University; Kashihara Nara Japan
| | - Yoshihiko Saito
- The First Department of Internal Medicine; Nara Medical University; Kashihara Nara Japan
| | - Osamu Nakagawa
- Laboratory for Cardiovascular System Research; Nara Medical University Advanced Medical Research Center; Kashihara Nara Japan
| |
Collapse
|
245
|
ALK5 and ALK1 play antagonistic roles in transforming growth factor β-induced podosome formation in aortic endothelial cells. Mol Cell Biol 2014; 34:4389-403. [PMID: 25266657 DOI: 10.1128/mcb.01026-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Transforming growth factor β (TGF-β) and related cytokines play a central role in the vascular system. In vitro, TGF-β induces aortic endothelial cells to assemble subcellular actin-rich structures specialized for matrix degradation called podosomes. To explore further this TGF-β-specific response and determine in which context podosomes form, ALK5 and ALK1 TGF-β receptor signaling pathways were investigated in bovine aortic endothelial cells. We report that TGF-β drives podosome formation through ALK5 and the downstream effectors Smad2 and Smad3. Concurrent TGF-β-induced ALK1 signaling mitigates ALK5 responses through Smad1. ALK1 signaling induced by BMP9 also antagonizes TGF-β-induced podosome formation, but this occurs through both Smad1 and Smad5. Whereas ALK1 neutralization brings ALK5 signals to full potency for TGF-β-induced podosome formation, ALK1 depletion leads to cell disturbances not compatible with podosome assembly. Thus, ALK1 possesses passive and active modalities. Altogether, our results reveal specific features of ALK1 and ALK5 signaling with potential clinical implications.
Collapse
|
246
|
Bai Y, Liang S, Yu W, Zhao M, Huang L, Zhao M, Li X. Semaphorin 3A blocks the formation of pathologic choroidal neovascularization induced by transforming growth factor beta. Mol Vis 2014; 20:1258-70. [PMID: 25352735 PMCID: PMC4168834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 09/17/2014] [Indexed: 11/01/2022] Open
Abstract
OBJECTIVE Choroidal neovascularization (CNV) is a major cause of vision loss in retinal diseases such as age-related macular degeneration (AMD). Previously, we demonstrated that semaphorin3A (Sema3A), which is a chemorepellent guidance molecule, inhibited the formation of retina neovascularization. In the present study, we investigated the antiangiogenic effects of Sema3A on transforming growth factor beta (TGF-β) in vitro and in vivo. METHODS Enzyme-linked immunosorbent assays (ELISAs) were used to measure the TGF-β levels in the vitreous humor of patients with AMD and controls. Human umbilical vein endothelial cells (HUVECs) were used for the in vitro study, and a laser-induced CNV mouse model was prepared for the in vivo study. The HUVECs were incubated with TGF-β and Sema3A. The proliferation, migration, apoptosis, and tube formation of the cells were then measured using BrdU, Transwell, flow cytometry, and Matrigel assays, respectively, and the SMAD2/3 signaling pathways were analyzed using western blot analysis. The C57BL/6J mouse retina was exposed to a laser to induce choroidal neovascularization (CNV), and Sema3A was injected intravitreously. After 14 days, fundus fluorescein angiography was performed to evaluate the leakage area of the CNV. The vascular endothelial growth factor (VEGF) and TGF-β concentrations in the retina-choroid complex were measured with ELISA. Components of the p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase-1/2 (ERK1/2), c-Jun NH2-terminal kinase (JNK), and SMAD2/3 signaling pathways in the Sema3A-treated groups were analyzed using western blotting. RESULTS In this study, we first verified that the vitreous TGF-β level was higher in patients with neovascular AMD than in the controls. We also showed that Sema3A inhibited TGF-β-induced HUVEC proliferation, migration, and tube formation and inhibited the downstream SMAD2/3 signaling pathway. Sema3A also induced TGF-β-stimulated HUVEC apoptosis and inhibited the response of TGF-β in vitro. In vivo, the TGF-β level was increased in the CNV mouse model. Sema3A not only inhibited laser-induced CNV formation but also inhibited the uptake of VEGF and TGF-β. In the western blot analysis, Sema3A was shown to inhibit the phosphorylation of p38 MAPK, ERK1/2, and JNK and to inhibit the SMAD2/3 signaling pathway after Sema3A treatment in CNV mice. CONCLUSIONS Sema3A can be applied as a useful, adjunctive therapeutic strategy for preventing CNV formation.
Collapse
|
247
|
Wei Z, Salmon RM, Upton PD, Morrell NW, Li W. Regulation of bone morphogenetic protein 9 (BMP9) by redox-dependent proteolysis. J Biol Chem 2014; 289:31150-9. [PMID: 25237187 PMCID: PMC4223318 DOI: 10.1074/jbc.m114.579771] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BMP9, a member of the TGFβ superfamily, is a homodimer that forms a signaling complex with two type I and two type II receptors. Signaling through high-affinity activin receptor-like kinase 1 (ALK1) in endothelial cells, circulating BMP9 acts as a vascular quiescence factor, maintaining endothelial homeostasis. BMP9 is also the most potent BMP for inducing osteogenic signaling in mesenchymal stem cells in vitro and promoting bone formation in vivo. This activity requires ALK1, the lower affinity type I receptor ALK2, and higher concentrations of BMP9. In adults, BMP9 is constitutively expressed in hepatocytes and secreted into the circulation. Optimum concentrations of BMP9 are essential to maintain the highly specific endothelial-protective function. Factors regulating BMP9 stability and activity remain unknown. Here, we showed by chromatography and a 1.9 Å crystal structure that stable BMP9 dimers could form either with (D-form) or without (M-form) an intermolecular disulfide bond. Although both forms of BMP9 were capable of binding to the prodomain and ALK1, the M-form demonstrated less sustained induction of Smad1/5/8 phosphorylation. The two forms could be converted into each other by changing the redox potential, and this redox switch caused a major alteration in BMP9 stability. The M-form displayed greater susceptibility to redox-dependent cleavage by proteases present in serum. This study provides a mechanism for the regulation of circulating BMP9 concentrations and may provide new rationales for approaches to modify BMP9 levels for therapeutic purposes.
Collapse
Affiliation(s)
- Zhenquan Wei
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Richard M Salmon
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Paul D Upton
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Nicholas W Morrell
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Wei Li
- From the Department of Medicine, University of Cambridge, School of Clinical Medicine, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
248
|
Pál G, Lovas G, Dobolyi A. Induction of transforming growth factor beta receptors following focal ischemia in the rat brain. PLoS One 2014; 9:e106544. [PMID: 25192322 PMCID: PMC4156357 DOI: 10.1371/journal.pone.0106544] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/07/2014] [Indexed: 01/02/2023] Open
Abstract
Transforming growth factor-βs (TGF-βs) regulate cellular proliferation, differentiation, and survival. TGF-βs bind to type I (TGF-βRI) and II receptors (TGF-βRII), which are transmembrane kinase receptors, and an accessory type III receptor (TGF-βRIII). TGF-β may utilize another type I receptor, activin-like kinase receptor (Alk1). TGF-β is neuroprotective in the middle cerebral artery occlusion (MCAO) model of stroke. Recently, we reported the expression pattern of TGF-β1-3 after MCAO. To establish how TGF-βs exert their actions following MCAO, the present study describes the induction of TGF-βRI, RII, RIII and Alk1 at 24 h, 72 h and 1 mo after transient 1 h MCAO as well as following 24 h permanent MCAO using in situ hybridization histochemistry. In intact brain, only TGF-βRI had significant expression: neurons in cortical layer IV contained TGF-βRI. At 24 h after the occlusion, no TGF-β receptors showed induction. At 72 h following MCAO, all four types of TGF-β receptors were induced in the infarct area, while TGF-βRI and RII also appeared in the penumbra. Most cells with elevated TGF-βRI mRNA levels were microglia. TGF-βRII co-localized with both microglial and endothelial markers while TGF-βRIII and Alk1 were present predominantly in endothels. All four TGF-β receptors were induced within the lesion 1 mo after the occlusion. In particular, TGF-βRIII was further induced as compared to 72 h after MCAO. At this time point, TGF-βRIII signal was predominantly not associated with blood vessels suggesting its microglial location. These data suggest that TGF-β receptors are induced after MCAO in a timely and spatially regulated fashion. TGF-β receptor expression is preceded by increased TGF-β expression. TGF-βRI and RII are likely to be co-expressed in microglial cells while Alk1, TGF-βRII, and RIII in endothels within the infarct where TGF-β1 may be their ligand. At later time points, TGF-βRIII may also appear in glial cells to potentially affect signal transduction via TGF-βRI and RII.
Collapse
Affiliation(s)
- Gabriella Pál
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gábor Lovas
- Department of Neurology, Semmelweis University, Budapest, Hungary
- Department of Neurology, Jahn Ferenc Teaching Hospital, Budapest, Hungary
| | - Arpád Dobolyi
- Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
- Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
249
|
Garcia MD, Larina IV. Vascular development and hemodynamic force in the mouse yolk sac. Front Physiol 2014; 5:308. [PMID: 25191274 PMCID: PMC4138559 DOI: 10.3389/fphys.2014.00308] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/29/2014] [Indexed: 11/13/2022] Open
Abstract
Vascular remodeling of the mouse embryonic yolk sac is a highly dynamic process dependent on multiple genetic signaling pathways as well as biomechanical factors regulating proliferation, differentiation, migration, cell-cell, and cell-matrix interactions. During this early developmental window, the initial primitive vascular network of the yolk sac undergoes a dynamic remodeling process concurrent with the onset of blood flow, in which endothelial cells establish a branched, hierarchical structure of large vessels and smaller capillary beds. In this review, we will describe the molecular and biomechanical regulators which guide vascular remodeling in the mouse embryonic yolk sac, as well as live imaging methods for characterizing endothelial cell and hemodynamic function in cultured embryos.
Collapse
Affiliation(s)
- Monica D Garcia
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine Houston, TX, USA
| | - Irina V Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine Houston, TX, USA
| |
Collapse
|
250
|
Lupo G, Motta C, Salmeri M, Spina-Purrello V, Alberghina M, Anfuso CD. An in vitro retinoblastoma human triple culture model of angiogenesis: a modulatory effect of TGF-β. Cancer Lett 2014; 354:181-8. [PMID: 25128651 DOI: 10.1016/j.canlet.2014.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/05/2014] [Accepted: 08/05/2014] [Indexed: 01/15/2023]
Abstract
Retinoblastoma is the most common intraocular tumour in children. In view of understanding the molecular mechanisms through which angiogenic switch on happens in the early phases of reciprocal interaction between tumour and cells constituting retinal microvessel, Transwell co-cultures constituted by human retinal endothelial cells (HREC), pericytes (HRPC), and human retinoblastoma cell line Y-79 were performed. Y-79 enhanced HREC proliferation, reduced by the introduction of HRPC in triple culture. In HREC/HRPC cultures, TGF-β in media increased, decreasing in triple cultures. High VEGF levels in triple cultures witnessed the establishment of a strongly in vitro angiogenic environment. Y-79 induced in HREC an increase in c- and iPLA2, phospho-cPLA2, inducible COX-2 protein expressions, PLA2 activities and prostaglandin E2 (PGE2) release. These effects were attenuated when HRPC were introduced in triple culture. Moreover, antibody silencing of TGF-β demonstrated a strong correlation between the signalling pathway triggered by TGF-β of pericytal origin and the phospholipase activation and the modulation of PGE2 release. Inhibiting VEGFA effect, the HRPC loss in triple culture decreased, showing its modulatory effect on their survival. Relying on the data here presented, sustaining the pericytal survival in a tumour retinal environment could ensure the integrity of microvessels and the TGF-β supply, essential for controlling aberrant endothelial pruning and angiogenesis.
Collapse
Affiliation(s)
- Gabriella Lupo
- Dipartimento di Biomedicina Clinica e Molecolare, Università di Catania, Italy
| | - Carla Motta
- Dipartimento di Biomedicina Clinica e Molecolare, Università di Catania, Italy
| | - Mario Salmeri
- Dipartimento di Scienze Bio-Mediche, Università di Catania, Italy
| | | | - Mario Alberghina
- Dipartimento di Biomedicina Clinica e Molecolare, Università di Catania, Italy
| | | |
Collapse
|