1
|
Hu T, Pang M, Sun Q, Gou Y, Liu J, Wang X, Ma Y, Chen W, Wei C, Liu M, Ding Y, Zhang Y, Liu D, Wu W, Wang P, Zhu H, Li Q, Yang F. Sema3A relieves neuropathic pain by reducing eIF2α phosphorylation via suppressing PI3K/Akt/mTOR pathway. THE JOURNAL OF PAIN 2025; 30:105374. [PMID: 40107588 DOI: 10.1016/j.jpain.2025.105374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
Primary sensory neurons serve as a critical link between the peripheral nervous system (PNS) and the central nervous system (CNS). They represent the initial neural tissue responsible for transmitting sensations and pain. In case where peripheral nerves are injured, nerve fiber regeneration can lead to severe pain. Semaphorin3A (Sema3A), an axon guidance molecule that can be secreted by Schwann cells, has been shown to effectively inhibit the regeneration of embryonic and adult dorsal root ganglion (DRG). However, its role in neuropathic pain and the underlying mechanisms remain unexplored. This study employed a chronic constriction injury (CCI) model of neuropathic pain in mice. We observed that increased expression of Sema3A could alleviate both mechanical and heat nociceptive behaviors in model mice. By overexpressing Sema3A in ipsilateral DRG neurons via DRG injection, we found that the phosphorylation of the PI3K/Akt/mTOR signaling pathway and eukaryotic initiation factor 2α (eIF2α) was inhibited, thereby inhibiting pain. eIF2α is a translation initiation factor and its phosphorylation can regulate global translation. The inhibition of eIF2α phosphorylation through PKR and PERK inhibitors also reduced the expression of ion channels and ultimately alleviated neuropathic pain. We found that Sema3A could suppress the phosphorylation of eIF2α by inhibiting the PI3K/AKT/mTOR pathway, thus affecting pain perception. These findings suggested that alterations in Sema3A expression and eIF2α phosphorylation were involved in the development of neuropathic pain, providing potential new targets for clinical pain-relief drug development. PERSPECTIVE: The expression of Sema3A in DRG neurons was decreased following peripheral nerve injury. Elevating Sema3A levels alleviated neuropathic pain by inhibiting the PI3K/Akt/mTOR pathway and eIF2α phosphorylation, thus affecting ion channel expression in DRG of neuropathic pain model animals. This highlighted Sema3A as potential therapeutic targets for pain relief.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin 300222, China
| | - Miaoyi Pang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Qingyu Sun
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yu Gou
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin University, Tianjin 300299, China
| | - Jing Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaotong Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yiran Ma
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wen Chen
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Chao Wei
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Meng Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yumeng Ding
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yurui Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Dianxin Liu
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Weihua Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Peipei Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hongwei Zhu
- Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Qian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
2
|
Lu YP, Luo YL, Wu ZY, Han C, Jin YZ, Han JM, Chen SY, Teng F, Han F, Liu XX, Lu YM. Semaphorin 3s signaling in the central nervous system: Mechanisms and therapeutic implication for brain diseases. Pharmacol Ther 2025; 267:108800. [PMID: 39855276 DOI: 10.1016/j.pharmthera.2025.108800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/16/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Class 3 semaphorins (Sema3s), identified as secreted soluble proteins, present many therapeutic potentials. Recent evidence has suggested that Sema3s as molecular cue participate in neuroregulation, angiogenesis, and microenvironment homeostasis of the central nervous system. Moreover, Sema3s signaling pathways may be targeted for enhancing neural network connectivity, promoting neural regeneration and repair, and inhibiting pathological angiogenesis. Due to the complex co-expression patterns and crosstalk among Sema3s, new drugs targeting Sema3s-related signaling pathways are expected to be discovered to counter brain diseases. This review summarizes the specific roles of Sema3s in pathological processes of various brain diseases, and provides potential targeted strategies for the prevention and treatment.
Collapse
Affiliation(s)
- Ya-Ping Lu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases of Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 210019, China
| | - Yi-Ling Luo
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Zhou-Yue Wu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases of Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chao Han
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Yin-Zhi Jin
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Jun-Ming Han
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Shu-Yang Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases of Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Fei Teng
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases of Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 210019, China; The affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian 223300, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiu-Xiu Liu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases of Ministry of Education, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| | - Ying-Mei Lu
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China; Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
3
|
Thomas L, Low S, Hansen G, Bakker RA, Zippel N. BI-Y, an Neuropilin-1 Antagonist, Enhances Revascularization and Prevents Vascular Endothelial Growth Factor-A Induced Retinal Hyperpermeability in Rodent Models of Retinopathies. J Pharmacol Exp Ther 2023; 385:214-221. [PMID: 36997325 DOI: 10.1124/jpet.122.001473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 04/01/2023] Open
Abstract
Diabetic retinopathy (DR) is a leading cause of vision loss in working-age adults. Despite an established standard of care for advanced forms of DR, some patients continue to lose vision after treatment. This may be due to the development of diabetic macular ischemia (DMI), which has no approved treatment. Neuropilin-1 (Nrp-1) is a coreceptor with two ligand-binding domains, with semaphorin-3A (Sema3A) binding to the A-domain and vascular endothelial growth factor-A (VEGF-A) binding to the B-domain. Sema3A directs a subset of neuronal growth cones as well as blood vessel growth by repulsion; when bound to Nrp-1, VEGF-A mediates vascular permeability and angiogenesis. Modulating Nrp-1 could therefore address multiple complications arising from DR, such as diabetic macular edema (DME) and DMI. BI-Y is a monoclonal antibody that binds to the Nrp-1 A-domain, antagonizing the effects of the ligand Sema3A and inhibiting VEGF-A-induced vascular permeability. This series of in vitro and in vivo studies examined the binding kinetics of BI-Y to Nrp-1 with and without VEGF-A165, the effect of BI-Y on Sema3A-induced cytoskeletal collapse, the effect of BI-Y on VEGF- A165-induced angiogenesis, neovascularization, cell integrity loss and permeability, and retinal revascularization. The data show that BI-Y binds to Nrp-1 and inhibits Sema3A-induced cytoskeletal collapse in vitro, may enhance revascularization of ischemic areas in an oxygen-induced retinopathy mouse model, and prevents VEGF-A-induced retinal hyperpermeability in rats. However, BI-Y does not interfere with VEGF-A-dependent choroidal neovascularization. These results support further investigation of BI-Y as a potential treatment for DMI and DME. SIGNIFICANCE STATEMENT: Diabetic macular ischemia (DMI) is a complication of diabetic retinopathy (DR) with no approved pharmacological treatment. Diabetic macular edema (DME) commonly co-occurs with DMI in patients with DR. This series of preclinical studies in mouse and rat models shows that neuropilin-1 antagonist BI-Y may enhance the revascularization of ischemic areas and prevents vascular endothelial growth factor-A (VEGF-A)-induced retinal hyperpermeability without affecting VEGF-A-dependent choroidal neovascularization; thus, BI-Y may be of interest as a potential treatment for patients with DR.
Collapse
Affiliation(s)
- Leo Thomas
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (L.T., R.A.B., N.Z.) and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (S.L., G.H.)
| | - Sarah Low
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (L.T., R.A.B., N.Z.) and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (S.L., G.H.)
| | - Gale Hansen
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (L.T., R.A.B., N.Z.) and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (S.L., G.H.)
| | - Remko A Bakker
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (L.T., R.A.B., N.Z.) and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (S.L., G.H.)
| | - Nina Zippel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany (L.T., R.A.B., N.Z.) and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (S.L., G.H.)
| |
Collapse
|
4
|
Laws KM, Bashaw GJ. Diverse roles for axon guidance pathways in adult tissue architecture and function. NATURAL SCIENCES (WEINHEIM, GERMANY) 2022; 2:e20220021. [PMID: 37456985 PMCID: PMC10346896 DOI: 10.1002/ntls.20220021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Classical axon guidance ligands and their neuronal receptors were first identified due to their fundamental roles in regulating connectivity in the developing nervous system. Since their initial discovery, it has become clear that these signaling molecules play important roles in the development of a broad array of tissue and organ systems across phylogeny. In addition to these diverse developmental roles, there is a growing appreciation that guidance signaling pathways have important functions in adult organisms, including the regulation of tissue integrity and homeostasis. These roles in adult organisms include both tissue-intrinsic activities of guidance molecules, as well as systemic effects on tissue maintenance and function mediated by the nervous and vascular systems. While many of these adult functions depend on mechanisms that mirror developmental activities, such as regulating adhesion and cell motility, there are also examples of adult roles that may reflect signaling activities that are distinct from known developmental mechanisms, including the contributions of guidance signaling pathways to lineage commitment in the intestinal epithelium and bone remodeling in vertebrates. In this review, we highlight studies of guidance receptors and their ligands in adult tissues outside of the nervous system, focusing on in vivo experimental contexts. Together, these studies lay the groundwork for future investigation into the conserved and tissue-specific mechanisms of guidance receptor signaling in adult tissues.
Collapse
Affiliation(s)
- Kaitlin M. Laws
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Current address: Department of Biology, Randolph-Macon College, Ashland, VA 23005, USA
| | - Greg J. Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Moon JE, Lawrence JB. Chromosome silencing in vitro reveals trisomy 21 causes cell-autonomous deficits in angiogenesis and early dysregulation in Notch signaling. Cell Rep 2022; 40:111174. [PMID: 35947952 PMCID: PMC9505374 DOI: 10.1016/j.celrep.2022.111174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/24/2021] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Despite the prevalence of Down syndrome (DS), little is known regarding the specific cell pathologies that underlie this multi-system disorder. To understand which cell types and pathways are more directly affected by trisomy 21 (T21), we used an inducible-XIST system to silence one chromosome 21 in vitro. T21 caused the dysregulation of Notch signaling in iPSCs, potentially affecting cell-type programming. Further analyses identified dysregulation of pathways important for two cell types: neurogenesis and angiogenesis. Angiogenesis is essential to many bodily systems, yet is understudied in DS; therefore, we focused next on whether T21 affects endothelial cells. An in vitro assay for microvasculature formation revealed a cellular pathology involving delayed tube formation in response to angiogenic signals. Parallel transcriptomic analysis of endothelia further showed deficits in angiogenesis regulators. Results indicate a direct cell-autonomous impact of T21 on endothelial function, highlighting the importance of angiogenesis, with wide-reaching implications for development and disease progression. Moon and Lawrence examine the immediate effects of trisomy 21 silencing and find angiogenesis and neurogenesis pathways, including Notch signaling, affected as early as pluripotency. In endothelial cells, functional analyses show that trisomy delays the angiogenic response for microvessel formation and transcriptomics show a parallel impact on angiogenic regulators and signal-response and cytoskeleton processes.
Collapse
Affiliation(s)
- Jennifer E Moon
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jeanne B Lawrence
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
6
|
Lai YJ, Tsai FC, Chang GJ, Chang SH, Huang CC, Chen WJ, Yeh YH. miR-181b targets semaphorin 3A to mediate TGF-β-induced endothelial-mesenchymal transition related to atrial fibrillation. J Clin Invest 2022; 132:142548. [PMID: 35775491 PMCID: PMC9246393 DOI: 10.1172/jci142548] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Atrial fibrosis is an essential contributor to atrial fibrillation (AF). It remains unclear whether atrial endocardial endothelial cells (AEECs) that undergo endothelial-mesenchymal transition (EndMT) are among the sources of atrial fibroblasts. We studied human atria, TGF-β-treated human AEECs, cardiac-specific TGF-β-transgenic mice, and heart failure rabbits to identify the underlying mechanism of EndMT in atrial fibrosis. Using isolated AEECs, we found that miR-181b was induced in TGF-β-treated AEECs, which decreased semaphorin 3A (Sema3A) and increased EndMT markers, and these effects could be reversed by a miR-181b antagomir. Experiments in which Sema3A was increased by a peptide or decreased by a siRNA in AEECs revealed a mechanistic link between Sema3A and LIM-kinase 1/phosphorylated cofilin (LIMK/p-cofilin) signaling and suggested that Sema3A is upstream of LIMK in regulating actin remodeling through p-cofilin. Administration of the miR-181b antagomir or recombinant Sema3A to TGF-β-transgenic mice evoked increased Sema3A, reduced EndMT markers, and significantly decreased atrial fibrosis and AF vulnerability. Our study provides a mechanistic link between the induction of EndMT by TGF-β via miR-181b/Sema3A/LIMK/p-cofilin signaling to atrial fibrosis. Blocking miR-181b and increasing Sema3A are potential strategies for AF therapeutic intervention.
Collapse
Affiliation(s)
- Ying-Ju Lai
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chia Yi, Taiwan
| | - Feng-Chun Tsai
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Gwo-Jyh Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Tao Yuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Chung-Chi Huang
- Department of Respiratory Therapy, Chang Gung University College of Medicine, Tao Yuan, Taiwan.,Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Tao Yuan, Taiwan
| | - Wei-Jan Chen
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang Gung Memorial Hospital, Tao Yuan, Taiwan.,Department of Medicine and
| |
Collapse
|
7
|
Zippel N, Kenny CH, Wu H, Garneau M, Kroe-Barrett R, Gupta P, Low S, Bakker RA, Thomas L. Sema3A Antibody BI-X Prevents Cell Permeability and Cytoskeletal Collapse in HRMECs and Increases Tip Cell Density in Mouse Oxygen-Induced Retinopathy. Transl Vis Sci Technol 2022; 11:17. [PMID: 35727188 PMCID: PMC9233289 DOI: 10.1167/tvst.11.6.17] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose Semaphorin 3A (Sema3A) is an axonal guidance molecule that inhibits angiogenesis by vasorepulsion and blocks revascularization in the ischemic retina. BI-X is an intravitreal anti-Sema3A agent under clinical investigation in patients with proliferative diabetic retinopathy (PDR) and diabetic macular ischemia (DMI). Methods Surface plasmon resonance was used to determine binding affinity of BI-X to human and murine Sema3A. In vitro, human retinal microvascular endothelial cells (HRMECs) were used to assess effects of BI-X on cell permeability and cytoskeletal collapse induced by Sema3A. In vivo, intravitreal BI-X or an anti-trinitrophenol control antibody was administered in both eyes in mice with oxygen-induced retinopathy (OIR). Retinal flat mounts were prepared, and avascular area and tip cell density were determined using confocal laser-scanning microscopy. Results Dissociation constants for BI-X binding to human and murine Sema3A were 29 pM and 27 pM, respectively. In vitro, BI-X prevented HRMEC permeability and cytoskeletal collapse induced by Sema3A. In vivo, BI-X increased tip cell density by 33% (P < 0.001) and reduced avascular area by 12% (not significant). A significant negative correlation was evident between avascular area and tip cell density (r2 = 0.4205, P < 0.0001). Conclusions BI-X binds to human Sema3A with picomolar affinity and prevents cell permeability and cytoskeletal collapse in HRMECs. BI-X also enhances revascularization in mice with OIR. Translational Relevance BI-X is a potent inhibitor of human Sema3A that improves revascularization in a murine model of OIR; BI-X is currently being investigated in patients with laser-treated PDR and DMI.
Collapse
Affiliation(s)
- Nina Zippel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Helen Wu
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Michel Garneau
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Priyanka Gupta
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Sarah Low
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Remko A Bakker
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Leo Thomas
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
8
|
Watterston C, Halabi R, McFarlane S, Childs SJ. Endothelial Semaphorin 3fb regulates Vegf pathway-mediated angiogenic sprouting. PLoS Genet 2021; 17:e1009769. [PMID: 34424892 PMCID: PMC8412281 DOI: 10.1371/journal.pgen.1009769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 09/02/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Vessel growth integrates diverse extrinsic signals with intrinsic signaling cascades to coordinate cell migration and sprouting morphogenesis. The pro-angiogenic effects of Vascular Endothelial Growth Factor (VEGF) are carefully controlled during sprouting to generate an efficiently patterned vascular network. We identify crosstalk between VEGF signaling and that of the secreted ligand Semaphorin 3fb (Sema3fb), one of two zebrafish paralogs of mammalian Sema3F. The sema3fb gene is expressed by endothelial cells in actively sprouting vessels. Loss of sema3fb results in abnormally wide and stunted intersegmental vessel artery sprouts. Although the sprouts initiate at the correct developmental time, they have a reduced migration speed. These sprouts have persistent filopodia and abnormally spaced nuclei suggesting dysregulated control of actin assembly. sema3fb mutants show simultaneously higher expression of pro-angiogenic (VEGF receptor 2 (vegfr2) and delta-like 4 (dll4)) and anti-angiogenic (soluble VEGF receptor 1 (svegfr1)/ soluble Fms Related Receptor Tyrosine Kinase 1 (sflt1)) pathway components. We show increased phospho-ERK staining in migrating angioblasts, consistent with enhanced Vegf activity. Reducing Vegfr2 kinase activity in sema3fb mutants rescues angiogenic sprouting. Our data suggest that Sema3fb plays a critical role in promoting endothelial sprouting through modulating the VEGF signaling pathway, acting as an autocrine cue that modulates intrinsic growth factor signaling.
Collapse
Affiliation(s)
- Charlene Watterston
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Rami Halabi
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sarah McFarlane
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Canada
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
9
|
Semaphorin3A increases M1-like microglia and retinal ganglion cell apoptosis after optic nerve injury. Cell Biosci 2021; 11:97. [PMID: 34039431 PMCID: PMC8157735 DOI: 10.1186/s13578-021-00603-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/04/2021] [Indexed: 11/10/2022] Open
Abstract
Background The mechanisms leading to retinal ganglion cell (RGC) death after optic nerve injury have not been fully elucidated. Current evidence indicates that microglial activation and M1- and M2-like dynamics may be an important factor in RGC apoptosis after optic nerve crush (ONC). Semaphorin3A (Sema3A) is a classic axonal guidance protein,which has been found to have a role in neuroinflammation processes. In this study, we investigated the contribution of microglial-derived Sema3A to progressive RGC apoptosis through regulating paradigm of M1- and M2-like microglia after ONC. Method
A mouse ONC model and a primary microglial-RGC co-culture system were used in the present study. The expression of M1- and M2-like microglial activation markers were assessed by real-time polymerase chain reaction (RT-qPCR). Histological and Western blot (WB) analyses were used to investigate the polarization patterns of microglia transitions and the levels of Sema3A. RGC apoptosis was investigated by TUNEL staining and caspase-3 detection. Results Levels of Sema3A in the mouse retina increased after ONC. Treatment of mice with the stimulating factor 1 receptor antagonist PLX3397 resulted in a decrease of retinal microglia. The levels of CD16/32 (M1) were up-regulated at days 3 and 7 post-ONC. However, CD206 (M2) declined on day 7 after ONC. Exposure to anti-Sema3A antibodies (anti-Sema3A) resulted in a decrease in the number of M1-like microglia, an increase in the number of M2-like microglia, and the amelioration of RGC apoptosis. Conclusions An increase in microglia-derived Sema3A in the retina after ONC partially leads to a continuous increase of M1-like microglia and plays an important role in RGC apoptosis. Inhibition of Sema3A activity may be a novel approach to the prevention of RGC apoptosis after optic nerve injury. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00603-7.
Collapse
|
10
|
Taberner L, Bañón A, Alsina B. Sensory Neuroblast Quiescence Depends on Vascular Cytoneme Contacts and Sensory Neuronal Differentiation Requires Initiation of Blood Flow. Cell Rep 2021; 32:107903. [PMID: 32668260 DOI: 10.1016/j.celrep.2020.107903] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 04/02/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023] Open
Abstract
In many organs, stem cell function depends on communication with their niche partners. Cranial sensory neurons develop in close proximity to blood vessels; however, whether vasculature is an integral component of their niches is yet unknown. Here, two separate roles for vasculature in cranial sensory neurogenesis in zebrafish are uncovered. The first involves precise spatiotemporal endothelial-neuroblast cytoneme contacts and Dll4-Notch signaling to restrain neuroblast proliferation. The second, instead, requires blood flow to trigger a transcriptional response that modifies neuroblast metabolic status and induces sensory neuron differentiation. In contrast, no role of sensory neurogenesis in vascular development is found, suggesting unidirectional signaling from vasculature to sensory neuroblasts. Altogether, we demonstrate that the cranial vasculature constitutes a niche component of the sensory ganglia that regulates the pace of their growth and differentiation dynamics.
Collapse
Affiliation(s)
- Laura Taberner
- Developmental Biology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Aitor Bañón
- Developmental Biology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Berta Alsina
- Developmental Biology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
11
|
Liu L, Xing L, Chen R, Zhang J, Huang Y, Huang L, Xie B, Ren X, Wang S, Kuang H, Lin X, Kumar A, Kim JK, Lee C, Li X. Mitogen-Inducible Gene 6 Inhibits Angiogenesis by Binding to SHC1 and Suppressing Its Phosphorylation. Front Cell Dev Biol 2021; 9:634242. [PMID: 33693003 PMCID: PMC7937727 DOI: 10.3389/fcell.2021.634242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/05/2021] [Indexed: 12/17/2022] Open
Abstract
The mitogen-inducible gene 6 (MIG6) is an adaptor protein widely expressed in vascular endothelial cells. However, it remains unknown thus far whether it plays a role in angiogenesis. Here, using comprehensive in vitro and in vivo model systems, we unveil a potent anti-angiogenic effect of MIG6 in retinal development and neovascularization and the underlying molecular and cellular mechanisms. Loss of function assays using genetic deletion of Mig6 or siRNA knockdown increased angiogenesis in vivo and in vitro, while MIG6 overexpression suppressed pathological angiogenesis. Moreover, we identified the cellular target of MIG6 by revealing its direct inhibitory effect on vascular endothelial cells (ECs). Mechanistically, we found that the anti-angiogenic effect of MIG6 is fulfilled by binding to SHC1 and inhibiting its phosphorylation. Indeed, SHC1 knockdown markedly diminished the effect of MIG6 on ECs. Thus, our findings show that MIG6 is a potent endogenous inhibitor of angiogenesis that may have therapeutic value in anti-angiogenic therapy.
Collapse
Affiliation(s)
- Lixian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Liying Xing
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Rongyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuye Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bingbing Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiangrong Ren
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shasha Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Haiqing Kuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xianchai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Anil Kumar
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jong Kyong Kim
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
12
|
Noueihed B, Rivera JC, Dabouz R, Abram P, Omri S, Lahaie I, Chemtob S. Mesenchymal Stromal Cells Promote Retinal Vascular Repair by Modulating Sema3E and IL-17A in a Model of Ischemic Retinopathy. Front Cell Dev Biol 2021; 9:630645. [PMID: 33553187 PMCID: PMC7859341 DOI: 10.3389/fcell.2021.630645] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic retinopathies (IRs), such as retinopathy of prematurity and diabetic retinopathy, are characterized by an initial phase of microvascular degeneration that results in retinal ischemia, followed by exaggerated pathologic neovascularization (NV). Mesenchymal stromal cells (MSCs) have potent pro-angiogenic and anti-inflammatory properties associated with tissue repair and regeneration, and in this regard exert protection to neurons in ischemic and degenerative conditions; however, the exact mechanisms underlying these functions remain largely unknown. Class III Semaphorins (A–G) are particularly implicated in regulating neural blood supply (as well as neurogenesis) by suppressing angiogenesis and affecting myeloid cell function; this is the case for distinct neuropillin-activating Sema3A as well as PlexinD1-activating Sema3E; but during IR the former Sema3A increases while Sema3E decreases. We investigated whether retinal vascular repair actions of MSCs are exerted by normalizing Semaphorin and downstream cytokines in IR. Intravitreal administration of MSCs or their secretome (MSCs-conditioned media [MSCs-CM]) significantly curtailed vasoobliteration as well as aberrant preretinal NV in a model of oxygen-induced retinopathy (OIR). The vascular repair effects of MSCs-CM in the ischemic retina were associated with restored levels of Sema3E. Vascular benefits of MSCs-CM were reversed by anti-Sema3E; while intravitreal injection of anti-angiogenic recombinant Sema3E (rSema3E) in OIR-subjected mice reproduced effects of MSCs-CM by inhibiting as expected preretinal NV but also by decreasing vasoobliteration. To explain these opposing vascular effects of Sema3E we found in OIR high retinal levels, respectively, of the pro- and anti-angiogenic IL-17A and Sema3A-regulating IL-1β; IL-17A positively affected expression of IL-1β. rSema3E decreased concentrations of these myeloid cell-derived pro-inflammatory cytokines in vitro and in vivo. Importantly, IL-17A suppression by MSCs-CM was abrogated by anti-Sema3E neutralizing antibody. Collectively, our findings provide novel evidence by which MSCs inhibit aberrant NV and diminish vasoobliteration (promoting revascularization) in retinopathy by restoring (at least in part) neuronal Sema3E levels that reduce pathological levels of IL-17A (and in turn other proinflammatory factors) in myeloid cells. The ability of MSCs to generate a microenvironment permissive for vascular regeneration by controlling the production of neuronal factors involved in immunomodulatory activities is a promising opportunity for stem cell therapy in ocular degenerative diseases.
Collapse
Affiliation(s)
- Baraa Noueihed
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - José Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Rabah Dabouz
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Pénélope Abram
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Samy Omri
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Isabelle Lahaie
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Sylvain Chemtob
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| |
Collapse
|
13
|
Hypoxia-induced downregulation of Sema3a and CXCL12/CXCR4 regulate the formation of the coronary artery stem at the proper site. J Mol Cell Cardiol 2020; 147:62-73. [PMID: 32777295 DOI: 10.1016/j.yjmcc.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/18/2020] [Accepted: 08/03/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND During the formation of the coronary artery stem, endothelial strands from the endothelial progenitor pool surrounding the conotruncus penetrate into the aortic wall. Vascular endothelial growth factors (VEGFs) as well as CXCL12/CXCR4 signaling are thought to play a role in the formation of the coronary stem. However, the mechanisms regulating how endothelial strands exclusively invade into the aorta remain unknown. METHODS AND RESULTS Immunohistochemistry showed that before the formation of endothelial strands, Sema3a was highly expressed in endothelial progenitors surrounding the great arteries. At the onset of/during invasion of endothelial strands into the aorta, Sema3a was downregulated and CXCR4 was upregulated in the endothelial strands. In situ hybridization showed that Cxcl12 was highly expressed in the aortic wall compared with in the pulmonary artery. Using avian embryonic hearts, we established two types of endothelial penetration assay, in which coronary endothelial strands preferentially invaded into the aorta in culture. Sema3a blocking peptide induced an excess number of endothelial strands penetrating into the pulmonary artery, whereas recombinant Sema3a inhibited the formation of endothelial strands. In cultured coronary endothelial progenitors, recombinant VEGF protein induced CXCR4-positive endothelial strands, which were capable of being attracted by CXCL12-impregnated beads. Monoazo rhodamine detected that hypoxia was predominant in aortic/subaortic region in ovo and hypoxic condition downregulated the expression of Sema3a in culture. CONCLUSION Results suggested that hypoxia in the aortic region downregulates the expression of Sema3a, thereby enhancing VEGF activity to induce the formation of CXCR4-positive endothelial strands, which are subsequently attracted into the Cxcl12-positive aortic wall to connect the aortic lumen.
Collapse
|
14
|
Ding MH, Lozoya EG, Rico RN, Chew SA. The Role of Angiogenesis-Inducing microRNAs in Vascular Tissue Engineering. Tissue Eng Part A 2020; 26:1283-1302. [PMID: 32762306 DOI: 10.1089/ten.tea.2020.0170] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is an important process in tissue repair and regeneration as blood vessels are integral to supply nutrients to a functioning tissue. In this review, the application of microRNAs (miRNAs) or anti-miRNAs that can induce angiogenesis to aid in blood vessel formation for vascular tissue engineering in ischemic diseases such as peripheral arterial disease and stroke, cardiac diseases, and skin and bone tissue engineering is discussed. Endothelial cells (ECs) form the endothelium of the blood vessel and are recognized as the primary cell type that drives angiogenesis and studied in the applications that were reviewed. Besides ECs, mesenchymal stem cells can also play a pivotal role in these applications, specifically, by secreting growth factors or cytokines for paracrine signaling and/or as constituent cells in the new blood vessel formed. In addition to delivering miRNAs or cells transfected/transduced with miRNAs for angiogenesis and vascular tissue engineering, the utilization of extracellular vesicles (EVs), such as exosomes, microvesicles, and EVs collectively, has been more recently explored. Proangiogenic miRNAs and anti-miRNAs contribute to angiogenesis by targeting the 3'-untranslated region of targets to upregulate proangiogenic factors such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor, and hypoxia-inducible factor-1 and increase the transduction of VEGF signaling through the PI3K/AKT and Ras/Raf/MEK/ERK signaling pathways such as phosphatase and tensin homolog or regulating the signaling of other pathways important for angiogenesis such as the Notch signaling pathway and the pathway to produce nitric oxide. In conclusion, angiogenesis-inducing miRNAs and anti-miRNAs are promising tools for vascular tissue engineering for several applications; however, future work should emphasize optimizing the delivery and usage of these therapies as miRNAs can also be associated with the negative implications of cancer.
Collapse
Affiliation(s)
- May-Hui Ding
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eloy G Lozoya
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Rene N Rico
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Sue Anne Chew
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
15
|
The role of semaphorins in small vessels of the eye and brain. Pharmacol Res 2020; 160:105044. [PMID: 32590102 DOI: 10.1016/j.phrs.2020.105044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
Small vessel diseases, such as ischemic retinopathy and cerebral small vessel disease (CSVD), are increasingly recognized in patients with diabetes, dementia and cerebrovascular disease. The mechanisms of small vessel diseases are poorly understood, but the latest studies suggest a role for semaphorins. Initially identified as axon guidance cues, semaphorins are mainly studied in neuronal morphogenesis, neural circuit assembly, and synapse assembly and refinement. In recent years, semaphorins have been found to play important roles in regulating vascular growth and development and in many pathophysiological processes, including atherosclerosis, angiogenesis after stroke and retinopathy. Growing evidence indicates that semaphorins affect the occurrence, perfusion and regression of both the macrovasculature and microvasculature by regulating the proliferation, apoptosis, migration, barrier function and inflammatory response of endothelial cells, vascular smooth muscle cells (VSMCs) and pericytes. In this review, we concentrate on the regulatory effects of semaphorins on the cell components of the vessel wall and their potential roles in microvascular diseases, especially in the retina and cerebral small vessel. Finally, we discuss potential molecular approaches in targeting semaphorins as therapies for microvascular disorders in the eye and brain.
Collapse
|
16
|
Li J, Zhang JS, Zhao JY, Han GG. Role of Smad4 from ocular surface ectoderm in retinal vasculature development. Int J Ophthalmol 2020; 13:231-238. [PMID: 32090031 DOI: 10.18240/ijo.2020.02.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 12/09/2019] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate how signals from lens regulate retinal vascular development and neovascularization. METHODS Le-Cre transgenic mouse line was employed to inactivate Smad4 in the surface ectoderm selectively. Standard histological and whole-mount retina staining were employed to reveal morphological changes of retinal vasculature in Smad4 defective eye. cDNA microarray and subsequent analyses were conducted to investigate the molecular mechanism underlying the vascular phenotype. Quantitative polymerase chain reaction (qPCR) was carried out to verify the microarrays results. RESULTS We found that inactivation of Smad4 specifically on surface ectoderm leads to a variety of retinal vasculature anomalies. Microarray analyses and qPCR revealed that Sema3c, Sema3e, Nrp1, Tie1, Sox7, Sox17, and Sox18 are significantly affected in the knockout retinas at different developmental stages, suggesting that ocular surface ectoderm-derived Smad4 can signal to the retina and regulates various angiogenic signaling in the retina. CONCLUSION Our data suggest that the cross-talk between ocular surface ectoderm and retina is important for retinal vasculature development, and Smad4 regulates various signaling associated with sprouting angiogenesis, vascular remodeling and maturation in the retina of mice.
Collapse
Affiliation(s)
- Jing Li
- Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Nankai University, Tianjin 300000, China
| | - Jin-Song Zhang
- Shenyang Aier Excellence Eye Hospital, Shenyang 110000, Liaoning Province, China
| | - Jiang-Yue Zhao
- Department of Ophthalmology, the Fourth Affiliated Hospital of China Medical University, Eye Hospital of China Medical University, Key Lens Research Laboratory, Shenyang 110000, Liaoning Province, China
| | - Guo-Ge Han
- Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Nankai University, Tianjin 300000, China
| |
Collapse
|
17
|
Fu Z, Sun Y, Cakir B, Tomita Y, Huang S, Wang Z, Liu CH, S. Cho S, Britton W, S. Kern T, Antonetti DA, Hellström A, E.H. Smith L. Targeting Neurovascular Interaction in Retinal Disorders. Int J Mol Sci 2020; 21:E1503. [PMID: 32098361 PMCID: PMC7073081 DOI: 10.3390/ijms21041503] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
The tightly structured neural retina has a unique vascular network comprised of three interconnected plexuses in the inner retina (and choroid for outer retina), which provide oxygen and nutrients to neurons to maintain normal function. Clinical and experimental evidence suggests that neuronal metabolic needs control both normal retinal vascular development and pathological aberrant vascular growth. Particularly, photoreceptors, with the highest density of mitochondria in the body, regulate retinal vascular development by modulating angiogenic and inflammatory factors. Photoreceptor metabolic dysfunction, oxidative stress, and inflammation may cause adaptive but ultimately pathological retinal vascular responses, leading to blindness. Here we focus on the factors involved in neurovascular interactions, which are potential therapeutic targets to decrease energy demand and/or to increase energy production for neovascular retinal disorders.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Bertan Cakir
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Shuo Huang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Zhongxiao Wang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Steve S. Cho
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - William Britton
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Timothy S. Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, Irvine, CA 92697, USA;
| | - David A. Antonetti
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Göteborg, Sweden;
| | - Lois E.H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| |
Collapse
|
18
|
Yanagida K, Engelbrecht E, Niaudet C, Jung B, Gaengel K, Holton K, Swendeman S, Liu CH, Levesque MV, Kuo A, Fu Z, Smith LEH, Betsholtz C, Hla T. Sphingosine 1-Phosphate Receptor Signaling Establishes AP-1 Gradients to Allow for Retinal Endothelial Cell Specialization. Dev Cell 2020; 52:779-793.e7. [PMID: 32059774 DOI: 10.1016/j.devcel.2020.01.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 12/09/2019] [Accepted: 01/16/2020] [Indexed: 12/17/2022]
Abstract
Transcriptional mechanisms that drive angiogenesis and organotypic vascular endothelial cell specialization are poorly understood. Here, we show that retinal endothelial sphingosine 1-phosphate receptors (S1PRs), which restrain vascular endothelial growth factor (VEGF)-induced angiogenesis, spatially restrict expression of JunB, a member of the activator protein 1 (AP-1) family of transcription factors (TFs). Mechanistically, VEGF induces JunB expression at the sprouting vascular front while S1PR-dependent vascular endothelial (VE)-cadherin assembly suppresses JunB expression in the nascent vascular network, thus creating a gradient of this TF. Endothelial-specific JunB knockout mice showed diminished expression of neurovascular guidance genes and attenuated retinal vascular network progression. In addition, endothelial S1PR signaling is required for normal expression of β-catenin-dependent genes such as TCF/LEF1 and ZIC3 TFs, transporters, and junctional proteins. These results show that S1PR signaling restricts JunB function to the expanding vascular front, thus creating an AP-1 gradient and enabling organotypic endothelial cell specialization of the vascular network.
Collapse
Affiliation(s)
- Keisuke Yanagida
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Eric Engelbrecht
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Colin Niaudet
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Bongnam Jung
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Steven Swendeman
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Catherine H Liu
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; ICMC (Integrated Cardio Metabolic Centre), Karolinska Institutet, Novum, Huddinge, Sweden
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
19
|
Li JY, Wang TT, Li C, Wang ZF, Li S, Ma L, Zheng LL. Semaphorin 3A-hypoxia inducible factor 1 subunit alpha co-overexpression enhances the osteogenic differentiation of induced pluripotent stem cells-derived mesenchymal stem cells in vitro. Chin Med J (Engl) 2020; 133:301-309. [PMID: 31929360 PMCID: PMC7004611 DOI: 10.1097/cm9.0000000000000612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Background: Mesenchymal stem or stromal cells (MSCs) derived from the induced pluripotent stem cells (iPSCs) have uniform biological activity, which makes the clinical application of MSCs in bone repair possible. Culturing the iPSC-MSCs onto osteoconductive materials is a promising tissue engineering-based strategy in bone regeneration. The aim of this work was to evaluate the effects of semaphorin 3A (Sema3A) and hypoxia inducible factor 1 subunit alpha (HIF1α) co-overexpression on the survival and osteogenic differentiation of iPSC-MSCs. Methods: Sema3A and HIF1α were linked together with the three (GGGGS; G, glycine; S, serine) peptide fragment, and their co-expression in iPSC-MSCs was mediated by a lentiviral vector. The fusion protein retained the immune reactivity for both Sema3A and HIF1α as determined with Western blotting. iPSC-MSCs were infected with overexpression lentivirus (oeLenti) as negative control, oeLenti-Sema3A, oeLenti-HIF1α or oeLenti-Sema3A-HIF1α lentiviruses. Results: Sema3A overexpression alone promoted the osteogenic differentiation of iPSC-MSCs (the activity and/or expression of osteoblast markers, such as alkaline phosphatase, osteopontin, and osteocalcin, were upregulated), and suppressed cell survival. The Sema3A-HIF1α fusion protein showed a comparable osteoconductive effect to that of Sema3A without reducing cell survival. We further seeded iPSC-MSCs modified by SemaA-HIF1α overexpression onto hydroxyapatite (HA) scaffolds, and evaluated their growth and differentiation on this three-dimensional material. Additional data indicated that, as compared to iPSC-MSCs cultured in ordinary two-dimensional dishes, cells cultured in HA scaffolds grew (blank vs. HA scaffolds: 0.83 vs. 1.39 for survival) and differentiated better (blank vs. HA scaffolds: 11.29 vs. 16.62 for alkaline phosphatase activity). Conclusion: Modifying iPSC-MSCs with pro-osteogenic (Sema3A) and pro-survival (HIF1α) factors may represent a promising strategy to optimize tissue engineering-based strategy in bone repair.
Collapse
Affiliation(s)
- Jing-Yi Li
- Department of Plastic Surgery, Capital Medical University Affiliated Beijing Tiantan Hospital, Beijing 100050, China
| | - Ting-Ting Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chong Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Zhi-Fang Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Shan Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Li Ma
- Department of Plastic Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Li-Li Zheng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| |
Collapse
|
20
|
Mangialardi G, Ferland-McCollough D, Maselli D, Santopaolo M, Cordaro A, Spinetti G, Sambataro M, Sullivan N, Blom A, Madeddu P. Bone marrow pericyte dysfunction in individuals with type 2 diabetes. Diabetologia 2019; 62:1275-1290. [PMID: 31001672 PMCID: PMC6560025 DOI: 10.1007/s00125-019-4865-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Previous studies have shown that diabetes mellitus destabilises the integrity of the microvasculature in different organs by damaging the interaction between pericytes and endothelial cells. In bone marrow, pericytes exert trophic functions on endothelial cells and haematopoietic cells through paracrine mechanisms. However, whether bone marrow pericytes are a target of diabetes-induced damage remains unknown. Here, we investigated whether type 2 diabetes can affect the abundance and function of bone marrow pericytes. METHODS We conducted an observational clinical study comparing the abundance and molecular/functional characteristics of CD146+ pericytes isolated from the bone marrow of 25 individuals without diabetes and 14 individuals with uncomplicated type 2 diabetes, referring to our Musculoskeletal Research Unit for hip reconstructive surgery. RESULTS Immunohistochemistry revealed that diabetes causes capillary rarefaction and compression of arteriole size in bone marrow, without changing CD146+ pericyte counts. These data were confirmed by flow cytometry on freshly isolated bone marrow cells. We then performed an extensive functional and molecular characterisation of immunosorted CD146+ pericytes. Type 2 diabetes caused a reduction in pericyte proliferation, viability, migration and capacity to support in vitro angiogenesis, while inducing apoptosis. AKT is a key regulator of the above functions and its phosphorylation state is reportedly reduced in the bone marrow endothelium of individuals with diabetes. Surprisingly, we could not find a difference in AKT phosphorylation (at either Ser473 or Thr308) in bone marrow pericytes from individuals with and without diabetes. Nonetheless, the angiocrine signalling reportedly associated with AKT was found to be significantly downregulated, with lower levels of fibroblast growth factor-2 (FGF2) and C-X-C motif chemokine ligand 12 (CXCL12), and activation of the angiogenesis inhibitor angiopoietin 2 (ANGPT2). Transfection with the adenoviral vector carrying the coding sequence for constitutively active myristoylated AKT rescued functional defects and angiocrine signalling in bone marrow pericytes from diabetic individuals. Furthermore, an ANGPT2 blocking antibody restored the capacity of pericytes to promote endothelial networking. CONCLUSIONS/INTERPRETATION This is the first demonstration of pericyte dysfunction in bone marrow of people with type 2 diabetes. An altered angiocrine signalling from pericytes may participate in bone marrow microvascular remodelling in individuals with diabetes.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - David Ferland-McCollough
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Davide Maselli
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
- IRCCS Multimedica, Milan, Italy
- Department of Biochemistry, University of Sassari, Sassari, Italy
| | - Marianna Santopaolo
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Andrea Cordaro
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | | | - Maria Sambataro
- Department of Specialized Medicines, Endocrine, Metabolic and Nutrition Diseases Unit, Santa Maria of Ca' Foncello Hospital, Treviso, Italy
| | - Niall Sullivan
- Muscloskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Ashley Blom
- Muscloskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Level 7, Upper Maudlin Street, Bristol, BS2 8HW, UK.
| |
Collapse
|
21
|
Guo S, Ren J, Li Z, Fan X, Qin L, Li J. Aqueous semaphorin 3A level correlates with retinal macular oedema and ganglion cell degeneration in patients with retinal vein occlusion. Acta Ophthalmol 2019; 97:273-278. [PMID: 30803163 DOI: 10.1111/aos.14079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 02/02/2019] [Indexed: 11/30/2022]
Abstract
PURPOSE To investigate the semophorin 3A (SEMA3A) level in aqueous humor of patients with retinal vein occlusion (RVO) and explore the correlation of SEMA3A with macular oedema and ganglion cell degeneration in RVO. METHODS This comparative study prospectively included 41 consecutive patients (41 eyes) with RVO who had intravitreal anti-VEGF injections from March 2014 to March 2015 for cystoid macular oedema (CME) or neovascular glaucoma (NVG). The patients were divided into three groups according to the fluorescein angiograghy (FFA): central retinal vein occlusion (CRVO) group (n = 15), branch retinal vein occlusion (BRVO) group (n = 15) and NVG group (secondary to CRVO, n = 11). The patients who had undergone cataract surgery (n = 16) during the same period served as controls. The SEMA3A concentration in aqueous humor collected before the initial anti-VEGF injection was determined by enzyme-linked immunosorbent assay (ELISA). Central retinal thickness (CRT), cube volume (CV) and ganglion cell-inner plexiform layer (GC-IPL) thickness was analysed by spectral-domain optical coherence tomography (SD-OCT). RESULTS Semaphorin 3A level in CRVO group (1.52 ± 1.23 ng/ml) and NVG group (1.67 ± 0.98 ng/ml) were significantly higher than the control group (0.66 ± 0.58 ng/ml; both p < 0.05). Moreover, SEMA3A level in CRVO group was higher than BRVO group (1.52 ± 1.23 ng/ml versus 0.53 ± 0.37 ng/ml; p < 0.05). SEMA3A level was positively correlated with CRT and CV in both BRVO group (CRTr = 0.6535, p = 0.0082; CVr = 0.5190, p = 0.0474) and CRVO group (CRTr = 0.6270, p = 0.0124; CVr = 0.6898, p = 0.0044). In RVO patients, the GC-IPL thickness of affected eyes were significantly reduced compared with the normal follow eyes (CRVOt = 4.55, p = 0.006; BRVOt = 4.54, p = 0.004). Meanwhile, negative correlation of SEMA3A level with GC-IPL thickness was found in both BRVO group (r = -0.5906, p = 0.0205) and CRVO group (r = -0.6100, p = 0.0157). CONCLUSION Semaphorin 3A level is increased in aqueous humor of RVO patients. Positive correlation of CRT as well as negative correlation of GC-IPL thickness with SEMA3A may suggest a pathological role of SEMA3A in macular oedema and ganglion cell degeneration during RVO.
Collapse
Affiliation(s)
- Shengxiang Guo
- Department of Ophthalmology First Affiliated Hospital of Xi'an Jiaotong University Xi'an China
- Department of Ophthalmology Xi'an Third Hospital Xi'an China
| | - Jiawei Ren
- Department of Ophthalmology First Affiliated Hospital of Xi'an Jiaotong University Xi'an China
| | - Zhengli Li
- Department of Clinical Laboratory Xi'an Third Hospital Xi'an China
| | | | - Li Qin
- Department of Ophthalmology First Affiliated Hospital of Xi'an Jiaotong University Xi'an China
| | - Jingming Li
- Department of Ophthalmology First Affiliated Hospital of Xi'an Jiaotong University Xi'an China
| |
Collapse
|
22
|
Montoya-Zegarra JA, Russo E, Runge P, Jadhav M, Willrodt AH, Stoma S, Nørrelykke SF, Detmar M, Halin C. AutoTube: a novel software for the automated morphometric analysis of vascular networks in tissues. Angiogenesis 2018; 22:223-236. [PMID: 30370470 PMCID: PMC6475513 DOI: 10.1007/s10456-018-9652-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/19/2018] [Indexed: 12/17/2022]
Abstract
Due to their involvement in many physiologic and pathologic processes, there is a great interest in identifying new molecular pathways that mediate the formation and function of blood and lymphatic vessels. Vascular research increasingly involves the image-based analysis and quantification of vessel networks in tissue whole-mounts or of tube-like structures formed by cultured endothelial cells in vitro. While both types of experiments deliver important mechanistic insights into (lymph)angiogenic processes, the manual analysis and quantification of such experiments are typically labour-intensive and affected by inter-experimenter variability. To bypass these problems, we developed AutoTube, a new software that quantifies parameters like the area covered by vessels, vessel width, skeleton length and branching or crossing points of vascular networks in tissues and in in vitro assays. AutoTube is freely downloadable, comprises an intuitive graphical user interface and helps to perform otherwise highly time-consuming image analyses in a rapid, automated and reproducible manner. By analysing lymphatic and blood vascular networks in whole-mounts prepared from different tissues or from gene-targeted mice with known vascular abnormalities, we demonstrate the ability of AutoTube to determine vascular parameters in close agreement to the manual analyses and to identify statistically significant differences in vascular morphology in tissues and in vascular networks formed in in vitro assays.
Collapse
Affiliation(s)
- Javier A Montoya-Zegarra
- Scientific Center for Optical and Electron Microscopy (ScopeM), ETH Zürich, Wolfgang-Pauli-Str. 14, 8093, Zurich, Switzerland
| | - Erica Russo
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Peter Runge
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Maria Jadhav
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Ann-Helen Willrodt
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Szymon Stoma
- Scientific Center for Optical and Electron Microscopy (ScopeM), ETH Zürich, Wolfgang-Pauli-Str. 14, 8093, Zurich, Switzerland
| | - Simon F Nørrelykke
- Scientific Center for Optical and Electron Microscopy (ScopeM), ETH Zürich, Wolfgang-Pauli-Str. 14, 8093, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland.
| |
Collapse
|
23
|
Iyer D, Zhao Q, Wirka R, Naravane A, Nguyen T, Liu B, Nagao M, Cheng P, Miller CL, Kim JB, Pjanic M, Quertermous T. Coronary artery disease genes SMAD3 and TCF21 promote opposing interactive genetic programs that regulate smooth muscle cell differentiation and disease risk. PLoS Genet 2018; 14:e1007681. [PMID: 30307970 PMCID: PMC6198989 DOI: 10.1371/journal.pgen.1007681] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/23/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Although numerous genetic loci have been associated with coronary artery disease (CAD) with genome wide association studies, efforts are needed to identify the causal genes in these loci and link them into fundamental signaling pathways. Recent studies have investigated the disease mechanism of CAD associated gene SMAD3, a central transcription factor (TF) in the TGFβ pathway, investigating its role in smooth muscle biology. In vitro studies in human coronary artery smooth muscle cells (HCASMC) revealed that SMAD3 modulates cellular phenotype, promoting expression of differentiation marker genes while inhibiting proliferation. RNA sequencing and chromatin immunoprecipitation sequencing studies in HCASMC identified downstream genes that reside in pathways which mediate vascular development and atherosclerosis processes in this cell type. HCASMC phenotype, and gene expression patterns promoted by SMAD3 were noted to have opposing direction of effect compared to another CAD associated TF, TCF21. At sites of SMAD3 and TCF21 colocalization on DNA, SMAD3 binding was inversely correlated with TCF21 binding, due in part to TCF21 locally blocking chromatin accessibility at the SMAD3 binding site. Further, TCF21 was able to directly inhibit SMAD3 activation of gene expression in transfection reporter gene studies. In contrast to TCF21 which is protective toward CAD, SMAD3 expression in HCASMC was shown to be directly correlated with disease risk. We propose that the pro-differentiation action of SMAD3 inhibits dedifferentiation that is required for HCASMC to expand and stabilize disease plaque as they respond to vascular stresses, counteracting the protective dedifferentiating activity of TCF21 and promoting disease risk.
Collapse
Affiliation(s)
- Dharini Iyer
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Quanyi Zhao
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Robert Wirka
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Ameay Naravane
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Trieu Nguyen
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Boxiang Liu
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Manabu Nagao
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Paul Cheng
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Clint L. Miller
- Departments of Public Health Sciences, Biochemistry and Genetics, and Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
| | - Juyong Brian Kim
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Milos Pjanic
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Thomas Quertermous
- Department of Medicine and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States of America
| |
Collapse
|
24
|
Zhang H, Vreeken D, Bruikman CS, van Zonneveld AJ, van Gils JM. Understanding netrins and semaphorins in mature endothelial cell biology. Pharmacol Res 2018; 137:1-10. [PMID: 30240825 DOI: 10.1016/j.phrs.2018.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/15/2018] [Accepted: 09/15/2018] [Indexed: 02/07/2023]
Abstract
Netrins and semaphorins are known as neuronal guidance molecules that are important to the facilitate patterning of the nervous system in embryonic development. In recent years, their function has been broadened to guide development in other systems, including the vascular system, where netrins and semaphorins critically contribute to the development of the vascular system. Evidence is accumulating that these guidance cues are also of critical importance in the biology of the mature endothelium by regulating the maintenance of endothelial quiescence. Here we review our current insights into the roles of netrins and semaphorins in endothelial cell survival, self-renewing, barrier function, response to wall shear stress, and control of the vascular tone. We also provide suggestions for future research into the functions of netrins and semaphorins in mature endothelial cell biology.
Collapse
Affiliation(s)
- Huayu Zhang
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Dianne Vreeken
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Caroline S Bruikman
- Amsterdam UMC, University of Amsterdam, Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Janine M van Gils
- Department of Internal Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| |
Collapse
|
25
|
Retinal vasculature development in health and disease. Prog Retin Eye Res 2017; 63:1-19. [PMID: 29129724 DOI: 10.1016/j.preteyeres.2017.11.001] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/17/2022]
Abstract
Development of the retinal vasculature is based on highly coordinated signalling between different cell types of the retina, integrating internal metabolic requirements with external influences such as the supply of oxygen and nutrients. The developing mouse retinal vasculature is a useful model system to study these interactions because it is experimentally accessible for intra ocular injections and genetic manipulations, can be easily imaged and develops in a similar fashion to that of humans. Research using this model has provided insights about general principles of angiogenesis as well as pathologies that affect the developing retinal vasculature. In this review, we discuss recent advances in our understanding of the molecular and cellular mechanisms that govern the interactions between neurons, glial and vascular cells in the developing retina. This includes a review of mechanisms that shape the retinal vasculature, such as sprouting angiogenesis, vascular network remodelling and vessel maturation. We also explore how the disruption of these processes in mice can lead to pathology - such as oxygen induced retinopathy - and how this translates to human retinopathy of prematurity.
Collapse
|
26
|
History and conceptual developments in vascular biology and angiogenesis research: a personal view. Angiogenesis 2017; 20:463-478. [DOI: 10.1007/s10456-017-9569-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/18/2017] [Indexed: 01/05/2023]
|
27
|
Sun Y, Liu CH, Wang Z, Meng SS, Burnim SB, SanGiovanni JP, Kamenecka TM, Solt LA, Chen J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis. FASEB J 2017. [PMID: 28646017 DOI: 10.1096/fj.201700172r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pathological proliferation of retinal blood vessels commonly causes vision impairment in proliferative retinopathies, including retinopathy of prematurity. Dysregulated crosstalk between the vasculature and retinal neurons is increasingly recognized as a major factor contributing to the pathogenesis of vascular diseases. Class 3 semaphorins (SEMA3s), a group of neuron-secreted axonal and vascular guidance factors, suppress pathological vascular growth in retinopathy. However, the upstream transcriptional regulators that mediate the function of SEMA3s in vascular growth are poorly understood. Here we showed that retinoic acid receptor-related orphan receptor α (RORα), a nuclear receptor and transcription factor, is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in a mouse model of oxygen-induced proliferative retinopathy. We found that genetic deficiency of RORα substantially induced Sema3e expression in retinopathy. Both RORα and SEMA3E were expressed in retinal ganglion cells. RORα directly bound to a specific ROR response element on the promoter of Sema3e and negatively regulated Sema3e promoter-driven luciferase expression. Suppression of Sema3e using adeno-associated virus 2 carrying short hairpin RNA targeting Sema3e promoted disoriented pathological neovascularization and partially abolished the inhibitory vascular effects of RORα deficiency in retinopathy. Our findings suggest that RORα is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in pathological retinal angiogenesis.-Sun, Y., Liu, C.-H., Wang, Z., Meng, S. S., Burnim, S. B., SanGiovanni, J. P., Kamenecka, T. M., Solt, L. A., Chen, J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Zhongxiao Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Steven S Meng
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Samuel B Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - John Paul SanGiovanni
- Section of Nutritional Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown School of Medicine, Washington, D.C., USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - Laura A Solt
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA;
| |
Collapse
|
28
|
Tai-Nagara I, Yoshikawa Y, Numata N, Ando T, Okabe K, Sugiura Y, Ieda M, Takakura N, Nakagawa O, Zhou B, Okabayashi K, Suematsu M, Kitagawa Y, Bastmeyer M, Sato K, Klein R, Navankasattusas S, Li DY, Yamagishi S, Kubota Y. Placental labyrinth formation in mice requires endothelial FLRT2/UNC5B signaling. Development 2017; 144:2392-2401. [PMID: 28576770 DOI: 10.1242/dev.149757] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/26/2017] [Indexed: 12/27/2022]
Abstract
The placental labyrinth is the interface for gas and nutrient exchange between the embryo and the mother; hence its proper development is essential for embryogenesis. However, the molecular mechanism underlying development of the placental labyrinth, particularly in terms of its endothelial organization, is not well understood. Here, we determined that fibronectin leucine-rich transmembrane protein 2 (FLRT2), a repulsive ligand of the UNC5 receptor family for neurons, is unexpectedly expressed in endothelial cells specifically in the placental labyrinth. Mice lacking FLRT2 in endothelial cells exhibited embryonic lethality at mid-gestation, with systemic congestion and hypoxia. Although they lacked apparent deformities in the embryonic vasculature and heart, the placental labyrinths of these embryos exhibited aberrant alignment of endothelial cells, which disturbed the feto-maternal circulation. Interestingly, this vascular deformity was related to endothelial repulsion through binding to the UNC5B receptor. Our results suggest that the proper organization of the placental labyrinth depends on coordinated inter-endothelial repulsion, which prevents uncontrolled layering of the endothelium.
Collapse
Affiliation(s)
- Ikue Tai-Nagara
- Department of Vascular Biology, The Sakaguchi Laboratory, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yusuke Yoshikawa
- Department of Vascular Biology, The Sakaguchi Laboratory, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Department of Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Naoko Numata
- Department of Vascular Biology, The Sakaguchi Laboratory, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tomofumi Ando
- Department of Vascular Biology, The Sakaguchi Laboratory, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Department of Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Keisuke Okabe
- Department of Vascular Biology, The Sakaguchi Laboratory, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Department of Plastic Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuki Sugiura
- Department of Biochemistry, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masaki Ieda
- Department of Cardiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Albert Einstein College of Medicine of Yeshiva University, 1301 Morris Park Avenue, Price Center 420, Bronx, NY 10461, USA
| | - Koji Okabayashi
- Department of Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Makoto Suematsu
- Department of Biochemistry, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Martin Bastmeyer
- Karlsruhe Institute of Technology (KIT), Zoological Institute, Cell- and Neurobiology, Fritz-Haber-Weg 4, Karlsruhe 76131, Germany
| | - Kohji Sato
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Rüdiger Klein
- Department Molecules - Signaling - Development, Max Planck Institute of Neurobiology, Am Klopferspitz 18, Martinsried 82152, Germany.,Munich Cluster for Systems Neurology (Synergy), Munich 80336, Germany
| | - Sutip Navankasattusas
- Department of Medicine, Program in Molecular Medicine, VA Salt Lake City Health Care System, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA
| | - Dean Y Li
- Department of Medicine, Program in Molecular Medicine, VA Salt Lake City Health Care System, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA.,Department of Oncological Sciences, VA Salt Lake City Health Care System, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA.,Department of Human Genetics, VA Salt Lake City Health Care System, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA.,ARUP Laboratories, VA Salt Lake City Health Care System, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA.,Division of Cardiovascular Medicine, Department of Medicine, VA Salt Lake City Health Care System, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA.,Department of Cardiology, VA Salt Lake City Health Care System, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA.,Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Satoru Yamagishi
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1, Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yoshiaki Kubota
- Department of Vascular Biology, The Sakaguchi Laboratory, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
29
|
Nantie L, Sun X. An Ephrin-Eph Tug and Push in Left-Right Organ Placement. Dev Cell 2016; 39:282-283. [PMID: 27825437 DOI: 10.1016/j.devcel.2016.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
During development, the placement of internal organs asymmetrically along the left-right axis is critical for their proper adult function. Reporting in Developmental Cell, Cayuso et al. (2016) demonstrate an active role of the endoderm in this process, challenging the prior view that the endoderm is passively pushed by the mesoderm.
Collapse
Affiliation(s)
- Leah Nantie
- Laboratory of Genetics, Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xin Sun
- Laboratory of Genetics, Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Pediatrics, University of California, San Diego, CA 92093, USA.
| |
Collapse
|
30
|
Neufeld G, Mumblat Y, Smolkin T, Toledano S, Nir-Zvi I, Ziv K, Kessler O. The semaphorins and their receptors as modulators of tumor progression. Drug Resist Updat 2016; 29:1-12. [DOI: 10.1016/j.drup.2016.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/31/2016] [Accepted: 08/23/2016] [Indexed: 12/16/2022]
|
31
|
Abstract
Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are uniquely required to balance the formation of new blood vessels with the maintenance and remodelling of existing ones, during development and in adult tissues. Recent advances have greatly expanded our understanding of the tight and multi-level regulation of VEGFR2 signalling, which is the primary focus of this Review. Important insights have been gained into the regulatory roles of VEGFR-interacting proteins (such as neuropilins, proteoglycans, integrins and protein tyrosine phosphatases); the dynamics of VEGFR2 endocytosis, trafficking and signalling; and the crosstalk between VEGF-induced signalling and other endothelial signalling cascades. A clear understanding of this multifaceted signalling web is key to successful therapeutic suppression or stimulation of vascular growth.
Collapse
|
32
|
Abstract
Secreted class 3 semaphorins (Sema3), which signal through holoreceptor complexes that are formed by different subunits, such as neuropilins (Nrps), proteoglycans, and plexins, were initially characterized as fundamental regulators of axon guidance during embryogenesis. Subsequently, Sema3A, Sema3C, Sema3D, and Sema3E were discovered to play crucial roles in cardiovascular development, mainly acting through Nrp1 and Plexin D1, which funnels the signal of multiple Sema3 in vascular endothelial cells. Mechanistically, Sema3 proteins control cardiovascular patterning through the enzymatic GTPase-activating-protein activity of the cytodomain of Plexin D1, which negatively regulates the function of Rap1, a small GTPase that is well-known for its ability to drive vascular morphogenesis and to elicit the conformational activation of integrin adhesion receptors.
Collapse
Affiliation(s)
- Donatella Valdembri
- a Department of Oncology , University of Torino School of Medicine , Candiolo, Torino , Italy.,b Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) , Candiolo, Torino , Italy
| | - Donatella Regano
- c Laboratory of Transgenic Mouse Models, Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) , Candiolo, Torino , Italy.,d Department of Science and Drug Technology , University of Torino , Candiolo, Torino , Italy
| | - Federica Maione
- c Laboratory of Transgenic Mouse Models, Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) , Candiolo, Torino , Italy.,d Department of Science and Drug Technology , University of Torino , Candiolo, Torino , Italy
| | - Enrico Giraudo
- c Laboratory of Transgenic Mouse Models, Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) , Candiolo, Torino , Italy.,d Department of Science and Drug Technology , University of Torino , Candiolo, Torino , Italy
| | - Guido Serini
- a Department of Oncology , University of Torino School of Medicine , Candiolo, Torino , Italy.,b Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) , Candiolo, Torino , Italy
| |
Collapse
|
33
|
Ochsenbein AM, Karaman S, Proulx ST, Berchtold M, Jurisic G, Stoeckli ET, Detmar M. Endothelial cell-derived semaphorin 3A inhibits filopodia formation by blood vascular tip cells. J Cell Sci 2016. [DOI: 10.1242/jcs.188300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|