151
|
Miller CA, Dahiya S, Li T, Fulton RS, Smyth MD, Dunn GP, Rubin JB, Mardis ER. Resistance-promoting effects of ependymoma treatment revealed through genomic analysis of multiple recurrences in a single patient. Cold Spring Harb Mol Case Stud 2018; 4:mcs.a002444. [PMID: 29440180 PMCID: PMC5880262 DOI: 10.1101/mcs.a002444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/26/2017] [Indexed: 12/11/2022] Open
Abstract
As in other brain tumors, multiple recurrences after complete resection and irradiation of supratentorial ependymoma are common and frequently result in patient death. This standard-of-care treatment was established in the pregenomic era without the ability to evaluate the effect that mutagenic therapies may exert on tumor evolution and in promoting resistance, recurrence, and death. We seized a rare opportunity to characterize treatment effects and the evolution of a single patient's ependymoma across four recurrences after different therapies. A combination of high-depth whole-genome and exome-based DNA sequencing of germline and tumor specimens, RNA sequencing of tumor specimens, and advanced computational analyses were used. Treatment with radiation and chemotherapies resulted in a substantial increase in mutational burden and diversification of the tumor subclonal architecture without eradication of the founding clone. Notable somatic alterations included a MEN1 driver, several epigenetic modifiers, and therapy-induced mutations that impacted multiple other cancer-relevant pathways and altered the neoantigen landscape. These genomic data provided new mechanistic insights into the genesis of ependymoma and pathways of resistance. They also revealed that radiation and chemotherapy were significant forces in shaping the increased subclonal complexity of each tumor recurrence while also failing to eradicate the founding clone. This raises the question of whether standard-of-care treatments have similar consequences in other patients with ependymoma and other types of brain tumors. If so, the perspective obtained by real-time genomic characterization of a tumor may be essential for making effective patient-specific and adaptive clinical decisions.
Collapse
Affiliation(s)
- Christopher A Miller
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.,McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Tiandao Li
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Matthew D Smyth
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Elaine R Mardis
- Institute for Genomic Medicine, Nationwide Children's Hospital, and The Ohio State University College of Medicine, Columbus, Ohio 43205, USA
| |
Collapse
|
152
|
Cross-talk between blood vessels and neural progenitors in the developing brain. Neuronal Signal 2018; 2:NS20170139. [PMID: 32714582 PMCID: PMC7371013 DOI: 10.1042/ns20170139] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 01/26/2023] Open
Abstract
The formation of the central nervous system (CNS) involves multiple cellular and molecular interactions between neural progenitor cells (NPCs) and blood vessels to establish extensive and complex neural networks and attract a vascular supply that support their function. In this review, we discuss studies that have performed genetic manipulations of chick, fish and mouse embryos to define the spatiotemporal roles of molecules that mediate the reciprocal regulation of NPCs and blood vessels. These experiments have highlighted core functions of NPC-expressed ligands in initiating vascular growth into and within the neural tube as well as establishing the blood-brain barrier. More recent findings have also revealed indispensable roles of blood vessels in regulating NPC expansion and eventual differentiation, and specific regional differences in the effect of angiocrine signals. Accordingly, NPCs initially stimulate blood vessel growth and maturation to nourish the brain, but blood vessels subsequently also regulate NPC behaviour to promote the formation of a sufficient number and diversity of neural cells. A greater understanding of the molecular cross-talk between NPCs and blood vessels will improve our knowledge of how the vertebrate nervous system forms and likely help in the design of novel therapies aimed at regenerating neurons and neural vasculature following CNS disease or injury.
Collapse
|
153
|
Pitter B, Werner AC, Montanez E. Parvins Are Required for Endothelial Cell-Cell Junctions and Cell Polarity During Embryonic Blood Vessel Formation. Arterioscler Thromb Vasc Biol 2018; 38:1147-1158. [PMID: 29567677 DOI: 10.1161/atvbaha.118.310840] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/05/2018] [Indexed: 02/01/2023]
Abstract
OBJECTIVE During vascular development, integrin-mediated signaling regulates the formation and stabilization of cell-cell junctions, which are required for endothelial cell (EC) apical-basal polarity and proper deposition of the vascular basement membrane. Parvins are actin-binding proteins that facilitate the interaction of integrins with the actin cytoskeleton. The endothelium expresses 2 parvin isoforms: α-pv (α-parvin) and β-pv (β-parvin). Recently, we have shown that α-pv is critical for vessel growth and vessel stability at late embryonic developmental stages. The role of parvins during early embryonic development is unknown. APPROACH AND RESULTS To investigate the role of endothelial parvins in the developing vasculature, we generated mice with ECs lacking both parvin isoforms by deleting α-pv in ECs in global β-pv-/- mice (α-pvΔEC;β-pv-/- mice). Here, we show that α-pvΔEC;β-pv-/- mice die around embryonic day 11.5 and exhibit hemorrhages, immature capillary beds, and severe vascular defects in the central nervous system, including reduced vessel branching, increased vessel diameter, and balloon-like hemorrhagic clusters of ECs. Vessels in α-pvΔEC;β-pv-/- embryos display disorganized cell-cell junctions, impaired endothelial apical-basal polarity, and discontinuous basement membranes. These vascular defects are accompanied by defective pericyte-vessel interaction. CONCLUSIONS Our results show that parvins are critical for the organization of endothelial cell-cell junctions, the establishment of endothelial apical-basal polarity, and the integrity of the basement membrane.
Collapse
Affiliation(s)
- Bettina Pitter
- From the Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig Maximilian University of Munich, Germany
| | - Ann-Cathrin Werner
- From the Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig Maximilian University of Munich, Germany
| | - Eloi Montanez
- From the Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig Maximilian University of Munich, Germany.
| |
Collapse
|
154
|
Scholz N. Cancer Cell Mechanics: Adhesion G Protein-coupled Receptors in Action? Front Oncol 2018; 8:59. [PMID: 29594040 PMCID: PMC5859372 DOI: 10.3389/fonc.2018.00059] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/21/2018] [Indexed: 12/11/2022] Open
Abstract
In mammals, numerous organ systems are equipped with adhesion G protein-coupled receptors (aGPCRs) to shape cellular processes including migration, adhesion, polarity and guidance. All of these cell biological aspects are closely associated with tumor cell biology. Consistently, aberrant expression or malfunction of aGPCRs has been associated with dysplasia and tumorigenesis. Mounting evidence indicates that cancer cells comprise viscoelastic properties that are different from that of their non-tumorigenic counterparts, a feature that is believed to contribute to the increased motility and invasiveness of metastatic cancer cells. This is particularly interesting in light of the recent identification of the mechanosensitive facility of aGPCRs. aGPCRs are signified by large extracellular domains (ECDs) with adhesive properties, which promote the engagement with insoluble ligands. This configuration may enable reliable force transmission to the ECDs and may constitute a molecular switch, vital for mechano-dependent aGPCR signaling. The investigation of aGPCR function in mechanosensation is still in its infancy and has been largely restricted to physiological contexts. It remains to be elucidated if and how aGPCR function affects the mechanoregulation of tumor cells, how this may shape the mechanical signature and ultimately determines the pathological features of a cancer cell. This article aims to view known aGPCR functions from a biomechanical perspective and to delineate how this might impinge on the mechanobiology of cancer cells.
Collapse
Affiliation(s)
- Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Faculty of Medicine, University Leipzig, Leipzig, Germany
| |
Collapse
|
155
|
Liebner S, Dijkhuizen RM, Reiss Y, Plate KH, Agalliu D, Constantin G. Functional morphology of the blood-brain barrier in health and disease. Acta Neuropathol 2018; 135:311-336. [PMID: 29411111 PMCID: PMC6781630 DOI: 10.1007/s00401-018-1815-1] [Citation(s) in RCA: 595] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/24/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
The adult quiescent blood-brain barrier (BBB), a structure organised by endothelial cells through interactions with pericytes, astrocytes, neurons and microglia in the neurovascular unit, is highly regulated but fragile at the same time. In the past decade, there has been considerable progress in understanding not only the molecular pathways involved in BBB development, but also BBB breakdown in neurological diseases. Specifically, the Wnt/β-catenin, retinoic acid and sonic hedgehog pathways moved into the focus of BBB research. Moreover, angiopoietin/Tie2 signalling that is linked to angiogenic processes has gained attention in the BBB field. Blood vessels play an essential role in initiation and progression of many diseases, including inflammation outside the central nervous system (CNS). Therefore, the potential influence of CNS blood vessels in neurological diseases associated with BBB alterations or neuroinflammation has become a major focus of current research to understand their contribution to pathogenesis. Moreover, the BBB remains a major obstacle to pharmaceutical intervention in the CNS. The complications may either be expressed by inadequate therapeutic delivery like in brain tumours, or by poor delivery of the drug across the BBB and ineffective bioavailability. In this review, we initially describe the cellular and molecular components that contribute to the steady state of the healthy BBB. We then discuss BBB alterations in ischaemic stroke, primary and metastatic brain tumour, chronic inflammation and Alzheimer's disease. Throughout the review, we highlight common mechanisms of BBB abnormalities among these diseases, in particular the contribution of neuroinflammation to BBB dysfunction and disease progression, and emphasise unique aspects of BBB alteration in certain diseases such as brain tumours. Moreover, this review highlights novel strategies to monitor BBB function by non-invasive imaging techniques focussing on ischaemic stroke, as well as novel ways to modulate BBB permeability and function to promote treatment of brain tumours, inflammation and Alzheimer's disease. In conclusion, a deep understanding of signals that maintain the healthy BBB and promote fluctuations in BBB permeability in disease states will be key to elucidate disease mechanisms and to identify potential targets for diagnostics and therapeutic modulation of the BBB.
Collapse
Affiliation(s)
- Stefan Liebner
- Institute of Neurology, Goethe University Clinic, Frankfurt am Main, Germany.
- Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner site Frankfurt, Frankfurt am Main, Germany.
- German Center for Cardiovascular Research (DZHK), Partner site Frankfurt/Mainz, Frankfurt am Main, Germany.
| | - Rick M Dijkhuizen
- Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Yvonne Reiss
- Institute of Neurology, Goethe University Clinic, Frankfurt am Main, Germany
- Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner site Frankfurt, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl H Plate
- Institute of Neurology, Goethe University Clinic, Frankfurt am Main, Germany
- Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Partner site Frankfurt, Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dritan Agalliu
- Departments of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
- Departments of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
- Departments of Pharmacology, Columbia University Medical Center, New York, NY, 10032, USA
- Departments of Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, 10032, USA
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| |
Collapse
|
156
|
Expression of the adhesion G protein-coupled receptor A2 (adgra2) during Xenopus laevis development. Gene Expr Patterns 2018; 28:54-61. [PMID: 29462671 DOI: 10.1016/j.gep.2018.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/18/2017] [Accepted: 02/14/2018] [Indexed: 11/21/2022]
Abstract
The adhesion G protein-coupled receptor A2 (Adgra2) is a seven transmembrane receptor that has been described to be a regulator for angiogenesis in mice. Furthermore, the zebrafish ouchless mutant is unable to develop dorsal root ganglia through a disrupted trafficking of Adgra2. Besides RNA sequencing data, nothing is reported about Adgra2 in the south African crawled frog Xenopus laevis. In this study, we investigated for the first time the spatio-temporal expression of adgra2 during early Xenopus embryogenesis in detail. In silico approaches showed that the genomic adgra2 region as well as the Adgra2 protein sequence is highly conserved among different species including Xenopus. RT-PCR experiments confirmed that embryonic adgra2 expression is primarily detected at the beginning of neurulation and is then present throughout the whole Xenopus embryogenesis until stage 42. Whole mount in situ hybridization approaches visualized adgra2 expression in many tissues during Xenopus embryogenesis such as the cardiovascular system including the heart, the migrating neural crest cells and the developing eye including the periocular mesenchyme. Our results indicate a role of Adgra2 for embryogenesis and are a good starting point for further functional studies during early vertebrate development.
Collapse
|
157
|
Fang M, Mei X, Yao H, Zhang T, Zhang T, Lu N, Liu Y, Xu W, Wan C. β-elemene enhances anticancer and anti-metastatic effects of osteosarcoma of ligustrazine in vitro and in vivo. Oncol Lett 2018; 15:3957-3964. [PMID: 29467906 DOI: 10.3892/ol.2018.7788] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 09/27/2017] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to determine the anticancer effects of the combination of β-elemene and ligustrazine in vitro as well as in in vivo. Following evaluation using an MTT assay, β-elemene, ligustrazine and the β-elemene-ligustrazine combination treatments all exhibited the capacity to inhibit the growth of OS-732 cells, with inhibitory rates of 43.3, 54.4, and 75.0%, respectively. Using a flow cytometry assay, it was determined that the β-elemene-ligustrazine combination possessed the highest apoptotic rate (30.6%). Furthermore, β-elemene-ligustrazine combination treatment resulted in the highest downregulation of G protein-coupled receptor 124, vascular endothelial growth factor, matrix metallopeptidase (MMP)-2 and MMP-9 mRNA, and protein expression levels. In addition, the combined treatment led to an increase in the mRNA and protein expression of endostatin, TIMP metallopeptidase inhibitor (TIMP)-1 and TIMP-2 in OS-732 cells. Additionally, β-elemene-ligustrazine caused a decrease in nuclear factor-κB, interleukin-8, C-X-C motif chemokine receptor 4 and urokinase-type plasminogen activator mRNA expression, as well as an increase in caspase-3, caspase-8, and caspase-9 mRNA expression. In vivo, the β-elemene-ligustrazine combination was able to reduce the weight and the bulk of the tumor in BALB/c-nu/nu nude mice compared with any other group. All the results described above regarding changes to mRNA and protein expression were further confirmed in vivo in the tumor tissue of mice. The results of the present study have suggested that the combination of β-elemene-ligustrazine exhibits greater anticancer effects compared with β-elemene- or ligustrazine-alone treatment.
Collapse
Affiliation(s)
- Min Fang
- Department of Trauma, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Xiaolong Mei
- Department of Trauma, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Hui Yao
- Department of Trauma, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Tao Zhang
- Department of Trauma, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Tao Zhang
- Department of Trauma, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Na Lu
- Department of Trauma, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Yanshi Liu
- Department of Clinical Medicine, Tianjin Medical University, Tianjin 300270, P.R. China
| | - Wenyue Xu
- Department of Ultrasonography, Tianjin Liulin Hospital, Tianjin 300222, P.R. China
| | - Chunyou Wan
- Department of Trauma, Tianjin Hospital, Tianjin 300211, P.R. China
| |
Collapse
|
158
|
Driehuis E, Clevers H. WNT signalling events near the cell membrane and their pharmacological targeting for the treatment of cancer. Br J Pharmacol 2017; 174:4547-4563. [PMID: 28244067 PMCID: PMC5727251 DOI: 10.1111/bph.13758] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 12/16/2022] Open
Abstract
WNT signalling is an essential signalling pathway for all multicellular animals. Although first described more than 30 years ago, new components and regulators of the pathway are still being discovered. Considering its importance in both embryonic development and adult homeostasis, it is not surprising that this pathway is often deregulated in human diseases such as cancer. Recently, it became clear that in addition to cytoplasmic components such as β-catenin, other, membrane-bound or extracellular, components of the WNT pathway are also altered in cancer. This review gives an overview of the recent discoveries on WNT signalling events near the cell membrane. Furthermore, membrane-associated components of the WNT pathway, which are more accessible for therapeutic intervention, as well therapeutic approaches that already target those components will be discussed. In this way, we hope to stimulate the development of effective anti-cancer therapies that target this fascinating pathway. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Else Driehuis
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW)UtrechtThe Netherlands
- University medical center (UMC)UtrechtThe Netherlands
| | - Hans Clevers
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW)UtrechtThe Netherlands
- University medical center (UMC)UtrechtThe Netherlands
- Princess Maxime Center (PMC)UtrechtThe Netherlands
| |
Collapse
|
159
|
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: 225] [Impact Index Per Article: 28.1] [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
|
160
|
Olsen JJ, Pohl SÖG, Deshmukh A, Visweswaran M, Ward NC, Arfuso F, Agostino M, Dharmarajan A. The Role of Wnt Signalling in Angiogenesis. Clin Biochem Rev 2017; 38:131-142. [PMID: 29332977 PMCID: PMC5759160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Angiogenesis is a normal biological process wherein new blood vessels form from the growth of pre-existing blood vessels. Preventing angiogenesis in solid tumours by targeting pro-angiogenic factors including vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1), basic fibroblast growth factor (bFGF), hepatocyte growth factor, and platelet-derived growth factor (PDGF) is currently under investigation for cancer treatment. Concurrently targeting the cell signalling pathways involved in the transcriptional and post-translational regulation of these factors may provide positive therapeutic results. One such pathway is the Wnt signalling pathway. Wnt was first discovered in mice infected with mouse mammary tumour virus, and has been crucial in improving our understanding of oncogenesis and development. In this review, we summarise molecular and cellular aspects of the importance of Wnt signalling to angiogenesis, including β-catenin-dependent mechanisms of angiogenic promotion, as well as the study of Wnt antagonists, such as the secreted frizzled-related protein family (SFRPs) which have been shown to inhibit angiogenesis. The growing understanding of the underlying complexity of the biochemical pathways mediating angiogenesis is critical to the identification of new molecular targets for therapeutic applications.
Collapse
Affiliation(s)
- Jun Jun Olsen
- The School of Human Sciences, The University of Western Australia, Nedlands, WA
| | - Sebastian Öther-Gee Pohl
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley, WA
| | - Abhijeet Deshmukh
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley, WA
| | - Malini Visweswaran
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley, WA
| | - Natalie C Ward
- School of Biomedical Sciences & Curtin Health Innovation Research Institute, Curtin University, Bentley, WA
- Medical School, University of Western Australia, Crawley, WA
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley, WA
| | - Mark Agostino
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley, WA
- Curtin Institute for Computation, Curtin University, Bentley, WA, Australia
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Bentley, WA
| |
Collapse
|
161
|
Abstract
CNS angiogenesis and blood-brain barrier integrity are controlled by the canonical Wnt pathway. In this issue of Neuron, Cho et al. (2017) use advanced mouse genetics and biochemical experiments to unravel the ligand-specific association of membrane proteins GPR124 and RECK with Wnt receptor complexes.
Collapse
Affiliation(s)
- Harald J Junge
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
162
|
McCord M, Mukouyama YS, Gilbert MR, Jackson S. Targeting WNT Signaling for Multifaceted Glioblastoma Therapy. Front Cell Neurosci 2017; 11:318. [PMID: 29081735 PMCID: PMC5645527 DOI: 10.3389/fncel.2017.00318] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/26/2017] [Indexed: 01/17/2023] Open
Abstract
The WNT signaling pathway has been of great interest to developmental biologists for decades and has more recently become a central topic for study in cancer biology. It is vital for cell growth and regulation of embryogenesis in many organ systems, particularly the CNS and its associated vasculature. We summarize the role of WNT in CNS development and describe how WNT signaling makes key contributions to malignant glioma stemness, invasiveness, therapeutic resistance, and angiogenesis. The role of WNT in these mechanisms, along with creation and maintainance of the blood-brain barrier (BBB), points to the potential of WNT as a multi-faceted target in malignant glioma therapy.
Collapse
Affiliation(s)
- Matthew McCord
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, MD, United States
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetic and Developmental Biology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, MD, United States
| | - Sadhana Jackson
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
163
|
Wallace J, Lutgen V, Avasarala S, St Croix B, Winn RA, Al-Harthi L. Wnt7a induces a unique phenotype of monocyte-derived macrophages with lower phagocytic capacity and differential expression of pro- and anti-inflammatory cytokines. Immunology 2017; 153:203-213. [PMID: 28872671 DOI: 10.1111/imm.12830] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/24/2017] [Accepted: 08/26/2017] [Indexed: 12/25/2022] Open
Abstract
The variation of macrophage functions suggests the involvement of multiple signalling pathways in fine tuning their differentiation. Macrophages that originate from monocytes in the blood migrate to tissue in response to homeostatic or 'danger' signals and undergo substantial morphological and functional modifications to meet the needs of the dominant signals in the microenvironment. Wnts are secreted glycoproteins that play a significant role in organ and cell differentiation, yet their impact on monocyte differentiation is not clear. In this study, we assessed the role of Wnt1 and Wnt7a on the differentiation of monocytes and the subsequent phenotype and function of monocyte-derived macrophages (MDMs). We show that Wnt7a decreased the expression of CD14, CD11b, CD163 and CD206, whereas Wnt1 had no effect. The Wnt7a effect on CD11b was also observed in the brain and spleen of Wnt7a-/- adult brain mouse tissue and in embryonic Wnt7a-/- tissue. Wnt7a reduced the phagocytic capacity of M-MDMs, decreased interleukin-10 (IL-10) and IL-12 secretion and increased IL-6 secretion. Collectively, these findings demonstrate that Wnt7a generates an MDM phenotype with both pro-inflammatory and alternative MDM cytokine profiles and reduced phagocytic capacity. As such, Wnt7a can have a significant impact on macrophage responses in health and disease.
Collapse
Affiliation(s)
- Jennillee Wallace
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL, USA
| | - Victoria Lutgen
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL, USA
| | - Sreedevi Avasarala
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Brad St Croix
- Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, MD, USA
| | - Robert A Winn
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Lena Al-Harthi
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
164
|
Abstract
The adhesion G protein-coupled receptors (aGPCRs) are an evolutionarily ancient family of receptors that play key roles in many different physiological processes. These receptors are notable for their exceptionally long ectodomains, which span several hundred to several thousand amino acids and contain various adhesion-related domains, as well as a GPCR autoproteolysis-inducing (GAIN) domain. The GAIN domain is conserved throughout almost the entire family and undergoes autoproteolysis to cleave the receptors into two noncovalently-associated protomers. Recent studies have revealed that the signaling activity of aGPCRs is largely determined by changes in the interactions among these protomers. We review recent advances in understanding aGPCR activation mechanisms and discuss the physiological roles and pharmacological properties of aGPCRs, with an eye toward the potential utility of these receptors as drug targets.
Collapse
Affiliation(s)
- Ryan H Purcell
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, 30322, USA;
| | - Randy A Hall
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, 30322, USA;
| |
Collapse
|
165
|
Ma S, Santhosh D, Kumar T P, Huang Z. A Brain-Region-Specific Neural Pathway Regulating Germinal Matrix Angiogenesis. Dev Cell 2017; 41:366-381.e4. [PMID: 28535372 DOI: 10.1016/j.devcel.2017.04.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 02/28/2017] [Accepted: 04/20/2017] [Indexed: 01/05/2023]
Abstract
Intimate communication between neural and vascular cells is critical for normal brain development and function. Germinal matrix (GM), a key primordium for the brain reward circuitry, is unique among brain regions for its distinct pace of angiogenesis and selective vulnerability to hemorrhage during development. A major neonatal condition, GM hemorrhage can lead to cerebral palsy, hydrocephalus, and mental retardation. Here we identify a brain-region-specific neural progenitor-based signaling pathway dedicated to regulating GM vessel development. This pathway consists of cell-surface sphingosine-1-phosphate receptors, an intracellular cascade including Gα co-factor Ric8a and p38 MAPK, and target gene integrin β8, which in turn regulates vascular TGF-β signaling. These findings provide insights into region-specific specialization of neurovascular communication, with special implications for deciphering potent early-life endocrine, as well as potential gut microbiota impacts on brain reward circuitry. They also identify tissue-specific molecular targets for GM hemorrhage intervention.
Collapse
Affiliation(s)
- Shang Ma
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, 53705, USA; Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI53706, USA
| | - Devi Santhosh
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, 53705, USA; Program in Genetics and Medical Genetics, University of Wisconsin-Madison, Madison, WI53706, USA
| | - Peeyush Kumar T
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, 53705, USA; Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI53706, USA; Program in Genetics and Medical Genetics, University of Wisconsin-Madison, Madison, WI53706, USA.
| |
Collapse
|
166
|
Cho C, Smallwood PM, Nathans J. Reck and Gpr124 Are Essential Receptor Cofactors for Wnt7a/Wnt7b-Specific Signaling in Mammalian CNS Angiogenesis and Blood-Brain Barrier Regulation. Neuron 2017; 95:1056-1073.e5. [PMID: 28803732 PMCID: PMC5586543 DOI: 10.1016/j.neuron.2017.07.031] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/14/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022]
Abstract
Reck, a GPI-anchored membrane protein, and Gpr124, an orphan GPCR, have been implicated in Wnt7a/Wnt7b signaling in the CNS vasculature. We show here that vascular endothelial cell (EC)-specific reduction in Reck impairs CNS angiogenesis and that EC-specific postnatal loss of Reck, combined with loss of Norrin, impairs blood-brain barrier (BBB) maintenance. The most N-terminal domain of Reck binds to the leucine-rich repeat (LRR) and immunoglobulin (Ig) domains of Gpr124, and weakening this interaction by targeted mutagenesis reduces Reck/Gpr124 stimulation of Wnt7a signaling in cell culture and impairs CNS angiogenesis. Finally, a soluble Gpr124(LRR-Ig) probe binds to cells expressing Frizzled, Wnt7a or Wnt7b, and Reck, and a soluble Reck(CC1-5) probe binds to cells expressing Frizzled, Wnt7a or Wnt7b, and Gpr124. These experiments indicate that Reck and Gpr124 are part of the cell surface protein complex that transduces Wnt7a- and Wnt7b-specific signals in mammalian CNS ECs to promote angiogenesis and regulate the BBB.
Collapse
Affiliation(s)
- Chris Cho
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Philip M Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
167
|
Augustin HG, Koh GY. Organotypic vasculature: From descriptive heterogeneity to functional pathophysiology. Science 2017; 357:science.aal2379. [DOI: 10.1126/science.aal2379] [Citation(s) in RCA: 351] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
168
|
Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017; 169:985-999. [PMID: 28575679 DOI: 10.1016/j.cell.2017.05.016] [Citation(s) in RCA: 3052] [Impact Index Per Article: 381.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/07/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022]
Abstract
The WNT signal transduction cascade is a main regulator of development throughout the animal kingdom. Wnts are also key drivers of most types of tissue stem cells in adult mammals. Unsurprisingly, mutated Wnt pathway components are causative to multiple growth-related pathologies and to cancer. Here, we describe the core Wnt/β-catenin signaling pathway, how it controls stem cells, and contributes to disease. Finally, we discuss strategies for Wnt-based therapies.
Collapse
|
169
|
Norrin-induced Frizzled4 endocytosis and endo-lysosomal trafficking control retinal angiogenesis and barrier function. Nat Commun 2017; 8:16050. [PMID: 28675177 PMCID: PMC5500887 DOI: 10.1038/ncomms16050] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 05/17/2017] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis and blood–brain barrier formation are required for normal central nervous system (CNS) function. Both processes are controlled by Wnt or Norrin (NDP) ligands, Frizzled (FZD) receptors, and β-catenin-dependent signalling in vascular endothelial cells. In the retina, FZD4 and the ligand NDP are critical mediators of signalling and are mutated in familial exudative vitreoretinopathy. Here, we report that NDP is a potent trigger of FZD4 ubiquitination and induces internalization of the NDP receptor complex into the endo-lysosomal compartment. Inhibition of ubiquitinated cargo transport through the multivesicular body (MVB) pathway using a dominant negative ESCRT (endosomal sorting complexes required for transport) component VPS4 EQ strongly impairs NDP/FZD4 signalling in vitro and recapitulates CNS angiogenesis and blood-CNS-barrier defects caused by impaired vascular β-catenin signalling in mice. These findings provide evidence for an important role of FZD4 endocytosis in NDP/FZD4 signalling and in CNS vascular biology and disease. Multiple mechanisms regulate Wnt/ß-catenin signalling. Zhang et al. describe a novel regulatory pathway and show that the activator of canonical Wnt signalling, Norrin, triggers endocytosis of its receptor Frizzled4 by promoting Frizzled4 ubiquitination.
Collapse
|
170
|
Díaz-Coránguez M, Ramos C, Antonetti DA. The inner blood-retinal barrier: Cellular basis and development. Vision Res 2017; 139:123-137. [PMID: 28619516 DOI: 10.1016/j.visres.2017.05.009] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 01/01/2023]
Abstract
The blood-retinal barrier (BRB) regulates transport across retinal capillaries maintaining proper neural homeostasis and protecting the neural tissue from potential blood borne toxicity. Loss of the BRB contributes to the pathophysiology of a number of blinding retinal diseases including diabetic retinopathy. In this review, we address the basis of the BRB, including the molecular mechanisms that regulate flux across the retinal vascular bed. The routes of transcellular and paracellular flux are described as well as alterations in these pathways in response to permeabilizing agents in diabetes. Finally, we provide information on exciting new studies that help to elucidate the process of BRB development or barriergenesis and how understanding this process may lead to new opportunities for barrier restoration in diabetic retinopathy.
Collapse
Affiliation(s)
- Mónica Díaz-Coránguez
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - Carla Ramos
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
171
|
Hernández-Vásquez MN, Adame-García SR, Hamoud N, Chidiac R, Reyes-Cruz G, Gratton JP, Côté JF, Vázquez-Prado J. Cell adhesion controlled by adhesion G protein-coupled receptor GPR124/ADGRA2 is mediated by a protein complex comprising intersectins and Elmo-Dock. J Biol Chem 2017; 292:12178-12191. [PMID: 28600358 DOI: 10.1074/jbc.m117.780304] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/30/2017] [Indexed: 11/06/2022] Open
Abstract
Developmental angiogenesis and the maintenance of the blood-brain barrier involve endothelial cell adhesion, which is linked to cytoskeletal dynamics. GPR124 (also known as TEM5/ADGRA2) is an adhesion G protein-coupled receptor family member that plays a pivotal role in brain angiogenesis and in ensuring a tight blood-brain barrier. However, the signaling properties of GPR124 remain poorly defined. Here, we show that ectopic expression of GPR124 promotes cell adhesion, additive to extracellular matrix-dependent effect, coupled with filopodia and lamellipodia formation and an enrichment of a pool of the G protein-coupled receptor at actin-rich cellular protrusions containing VASP, a filopodial marker. Accordingly, GPR124-expressing cells also displayed increased activation of both Rac and Cdc42 GTPases. Mechanistically, we uncover novel direct interactions between endogenous GPR124 and the Rho guanine nucleotide exchange factors Elmo/Dock and intersectin (ITSN). Small fragments of either Elmo or ITSN1 that bind GPR124 blocked GPR124-induced cell adhesion. In addition, Gβγ interacts with the C-terminal tail of GPR124 and promotes the formation of a GPR124-Elmo complex. Furthermore, GPR124 also promotes the activation of the Elmo-Dock complex, as measured by Elmo phosphorylation on a conserved C-terminal tyrosine residue. Interestingly, Elmo and ITSN1 also interact with each other independently of their GPR124-recognition regions. Moreover, endogenous phospho-Elmo and ITSN1 co-localize with GPR124 at lamellipodia of adhering endothelial cells, where GPR124 expression contributes to polarity acquisition during wound healing. Collectively, our results indicate that GPR124 promotes cell adhesion via Elmo-Dock and ITSN. This constitutes a previously unrecognized complex formed of atypical and conventional Rho guanine nucleotide exchange factors for Rac and Cdc42 that is putatively involved in GPR124-dependent angiogenic responses.
Collapse
Affiliation(s)
- Magda Nohemí Hernández-Vásquez
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico
| | - Sendi Rafael Adame-García
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico
| | - Noumeira Hamoud
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Rony Chidiac
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico
| | - Jean Philippe Gratton
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - José Vázquez-Prado
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico.
| |
Collapse
|
172
|
Bonney S, Siegenthaler JA. Differential Effects of Retinoic Acid Concentrations in Regulating Blood-Brain Barrier Properties. eNeuro 2017; 4:ENEURO.0378-16.2017. [PMID: 28560318 PMCID: PMC5446490 DOI: 10.1523/eneuro.0378-16.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 11/21/2022] Open
Abstract
The blood-brain barrier (BBB) is a multifaceted property of the brain vasculature that protects the brain and maintains homeostasis by tightly regulating the flux of ions, molecules, and cells across the vasculature. Blood vessels in the brain are formed by endothelial cells that acquire barrier properties, such as tight and adherens junctions, soon after the brain vasculature is formed. Endothelial WNT signaling is crucial to induce these BBB properties by regulating their expression and stabilization. Recent studies have implicated retinoic acid (RA) signaling in BBB development and shown that pharmacological concentrations of RA (≥5 µm) can induce BBB properties in cultured brain endothelial cells. However, a recent study demonstrated that RA inhibits endothelial WNT signaling during brain development, suggesting that RA does not promote BBB properties. We therefore investigated whether RA plays a physiological role in BBB development. We found that BBB function and junctional protein expression was unaffected in mouse mutants that have a reduced capacity to synthesize RA (Rdh10 mutants). Furthermore, embryos exposed to a RA-enriched diet did not enhance BBB protein expression. Together, our data indicate that RA is not capable of inducing, nor is it required for, BBB protein expression in vivo. Like other studies, we found that pharmacological concentrations of RA induce BBB genes in cultured murine brain endothelial cells, and this may involve activation of the LXR/RXR signaling pathway. Our data do not support a role for RA in BBB development, but confirm reports that pharmacological RA is a robust tool to induce BBB properties in culture.
Collapse
Affiliation(s)
- Stephanie Bonney
- Department of Pediatrics, Section of Developmental Biology School of Medicine Aurora, University of Colorado, CO 80045
| | - Julie A Siegenthaler
- Department of Pediatrics, Section of Developmental Biology School of Medicine Aurora, University of Colorado, CO 80045
| |
Collapse
|
173
|
Haddad-Tóvolli R, Dragano NRV, Ramalho AFS, Velloso LA. Development and Function of the Blood-Brain Barrier in the Context of Metabolic Control. Front Neurosci 2017; 11:224. [PMID: 28484368 PMCID: PMC5399017 DOI: 10.3389/fnins.2017.00224] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/04/2017] [Indexed: 12/21/2022] Open
Abstract
Under physiological conditions, the brain consumes over 20% of the whole body energy supply. The blood-brain barrier (BBB) allows dynamic interactions between blood capillaries and the neuronal network in order to provide an adequate control of molecules that are transported in and out of the brain. Alterations in the BBB structure and function affecting brain accessibility to nutrients and exit of toxins are found in a number of diseases, which in turn may disturb brain function and nutrient signaling. In this review we explore the major advances obtained in the understanding of the BBB development and how its structure impacts on function. Furthermore, we focus on the particularities of the barrier permeability in the hypothalamus, its role in metabolic control and the potential impact of hypothalamic BBB abnormities in metabolic related diseases.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, Faculty of Medical Sciences, University of CampinasCampinas, Brazil
| | | | | | - Licio A. Velloso
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, Faculty of Medical Sciences, University of CampinasCampinas, Brazil
| |
Collapse
|
174
|
Umans RA, Henson HE, Mu F, Parupalli C, Ju B, Peters JL, Lanham KA, Plavicki JS, Taylor MR. CNS angiogenesis and barriergenesis occur simultaneously. Dev Biol 2017; 425:101-108. [PMID: 28365243 DOI: 10.1016/j.ydbio.2017.03.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/24/2017] [Accepted: 03/18/2017] [Indexed: 01/04/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in the central nervous system (CNS). A comprehensive understanding of BBB development has been hampered by difficulties in observing the differentiation of brain endothelial cells (BECs) in real-time. Here, we generated two transgenic zebrafish line, Tg(glut1b:mCherry) and Tg(plvap:EGFP), to serve as in vivo reporters of BBB development. We showed that barriergenesis (i.e. the induction of BEC differentiation) occurs immediately as endothelial tips cells migrate into the brain parenchyma. Using the Tg(glut1b:mCherry) transgenic line, we performed a genetic screen and identified a zebrafish mutant with a nonsense mutation in gpr124, a gene known to play a role in CNS angiogenesis and BBB development. We also showed that our transgenic plvap:EGFP line, a reporter of immature brain endothelium, is initially expressed in newly formed brain endothelial cells, but subsides during BBB maturation. Our results demonstrate the ability to visualize the in vivo differentiation of brain endothelial cells into the BBB phenotype and establish that CNS angiogenesis and barriergenesis occur simultaneously.
Collapse
Affiliation(s)
- Robyn A Umans
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA; Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hannah E Henson
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA; Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Fangzhou Mu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Chaithanyarani Parupalli
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bensheng Ju
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jennifer L Peters
- Cell and Tissue Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kevin A Lanham
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Jessica S Plavicki
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Michael R Taylor
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA; Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
175
|
Gpr124 is essential for blood-brain barrier integrity in central nervous system disease. Nat Med 2017; 23:450-460. [PMID: 28288111 DOI: 10.1038/nm.4309] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 02/21/2017] [Indexed: 12/14/2022]
Abstract
Although blood-brain barrier (BBB) compromise is central to the etiology of diverse central nervous system (CNS) disorders, endothelial receptor proteins that control BBB function are poorly defined. The endothelial G-protein-coupled receptor (GPCR) Gpr124 has been reported to be required for normal forebrain angiogenesis and BBB function in mouse embryos, but the role of this receptor in adult animals is unknown. Here Gpr124 conditional knockout (CKO) in the endothelia of adult mice did not affect homeostatic BBB integrity, but resulted in BBB disruption and microvascular hemorrhage in mouse models of both ischemic stroke and glioblastoma, accompanied by reduced cerebrovascular canonical Wnt-β-catenin signaling. Constitutive activation of Wnt-β-catenin signaling fully corrected the BBB disruption and hemorrhage defects of Gpr124-CKO mice, with rescue of the endothelial gene tight junction, pericyte coverage and extracellular-matrix deficits. We thus identify Gpr124 as an endothelial GPCR specifically required for endothelial Wnt signaling and BBB integrity under pathological conditions in adult mice. This finding implicates Gpr124 as a potential therapeutic target for human CNS disorders characterized by BBB disruption.
Collapse
|
176
|
Alok A, Lei Z, Jagannathan NS, Kaur S, Harmston N, Rozen SG, Tucker-Kellogg L, Virshup DM. Wnt proteins synergize to activate β-catenin signaling. J Cell Sci 2017; 130:1532-1544. [PMID: 28289266 DOI: 10.1242/jcs.198093] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/07/2017] [Indexed: 12/31/2022] Open
Abstract
Wnt ligands are involved in diverse signaling pathways that are active during development, maintenance of tissue homeostasis and in various disease states. While signaling regulated by individual Wnts has been extensively studied, Wnts are rarely expressed alone, and the consequences of Wnt gene co-expression are not well understood. Here, we studied the effect of co-expression of Wnts on the β-catenin signaling pathway. While some Wnts are deemed 'non-canonical' due to their limited ability to activate β-catenin when expressed alone, unexpectedly, we find that multiple Wnt combinations can synergistically activate β-catenin signaling in multiple cell types. WNT1- and WNT7B-mediated synergistic Wnt signaling requires FZD5, FZD8 and LRP6, as well as the WNT7B co-receptors GPR124 (also known as ADGRA2) and RECK. Unexpectedly, this synergistic signaling occurs downstream of β-catenin stabilization, and is correlated with increased lysine acetylation of β-catenin. Wnt synergy provides a general mechanism to confer increased combinatorial control over this important regulatory pathway.
Collapse
Affiliation(s)
- Anshula Alok
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore
| | - Zhengdeng Lei
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore.,Centre for Computational Biology, Duke-NUS Medical School, 169857 Singapore
| | - N Suhas Jagannathan
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore.,Centre for Computational Biology, Duke-NUS Medical School, 169857 Singapore
| | - Simran Kaur
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore
| | - Nathan Harmston
- Centre for Computational Biology, Duke-NUS Medical School, 169857 Singapore
| | - Steven G Rozen
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore.,Centre for Computational Biology, Duke-NUS Medical School, 169857 Singapore
| | - Lisa Tucker-Kellogg
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore.,Centre for Computational Biology, Duke-NUS Medical School, 169857 Singapore
| | - David M Virshup
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, 169857 Singapore .,Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
177
|
|
178
|
Bostaille N, Gauquier A, Twyffels L, Vanhollebeke B. Molecular insights into Adgra2/Gpr124 and Reck intracellular trafficking. Biol Open 2016; 5:1874-1881. [PMID: 27979830 PMCID: PMC5200908 DOI: 10.1242/bio.021287] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adgra2, formerly known as Gpr124, is a key regulator of cerebrovascular development in vertebrates. Together with the GPI-anchored glycoprotein Reck, this adhesion GPCR (aGPCR) stimulates Wnt7-dependent Wnt/β-catenin signaling to promote brain vascular invasion in an endothelial cell-autonomous manner. Adgra2 and Reck have been proposed to assemble a receptor complex at the plasma membrane, but the molecular modalities of their functional synergy remain to be investigated. In particular, as typically found in aGPCRs, the ectodomain of Adgra2 is rich in protein-protein interaction motifs whose contributions to receptor function are unknown. In opposition to the severe ADGRA2 genetic lesions found in previously generated zebrafish and mouse models, the zebrafish ouchless allele encodes an aberrantly-spliced and inactive receptor lacking a single leucine-rich repeat (LRR) unit within its N-terminus. By characterizing this allele we uncover that, in contrast to all other extracellular domains, the precise composition of the LRR domain determines proper receptor trafficking to the plasma membrane. Using CRISPR/Cas9 engineered cells, we further show that Adgra2 trafficking occurs in a Reck-independent manner and that, similarly, Reck reaches the plasma membrane irrespective of Adgra2 expression or localization, suggesting that the partners meet at the plasma membrane after independent intracellular trafficking events. Summary: This work uncovers molecular determinants of Adgra2/Gpr124 and Reck trafficking to the plasma membrane where the partners meet to act as potent Wnt7-specific Wnt/β-catenin signaling co-activators required for brain vascularization.
Collapse
Affiliation(s)
- Naguissa Bostaille
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies B-6041, Belgium
| | - Anne Gauquier
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies B-6041, Belgium
| | - Laure Twyffels
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles (ULB), Gosselies B-6041, Belgium
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies B-6041, Belgium .,Center for Microscopy and Molecular Imaging, Université libre de Bruxelles (ULB), Gosselies B-6041, Belgium
| |
Collapse
|
179
|
Stem Cells as a Promising Tool for the Restoration of Brain Neurovascular Unit and Angiogenic Orientation. Mol Neurobiol 2016; 54:7689-7705. [DOI: 10.1007/s12035-016-0286-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/02/2016] [Indexed: 02/07/2023]
|
180
|
CORRELATION OF INCREASED INTRAVITREOUS WNT3A WITH VASCULAR ENDOTHELIAL GROWTH FACTOR IN PROLIFERATIVE DIABETIC RETINOPATHY. Retina 2016; 36:812-8. [PMID: 26418446 DOI: 10.1097/iae.0000000000000784] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate Wnt3a and vascular endothelial growth factor (VEGF) levels in the vitreous fluid of patients with proliferative diabetic retinopathy (PDR) and to examine their correlation with PDR activity. METHODS Vitreous samples from 45 eyes with PDR and 28 eyes with nondiabetic macular disease were collected. Active PDR was present in 24 patients and inactive PDR in 21 patients, according to retinal neovascularization. The Wnt3a and VEGF level of vitreous fluid samples were measured by enzyme-linked immunosorbent assay. RESULTS Comparison revealed that mean intravitreal levels of Wnt3a increased significantly in PDR eyes compared with control eyes (13.55 ng/mL vs. 1.57 ng/mL, P < 0.001). The mean VEGF concentrations in the vitreous fluid of patients with PDR were also higher than those in nondiabetic controls, with the values being 723.21 pg/mL and 20.81 pg/mL, respectively (P < 0.001). In addition, vitreous concentrations of Wnt3a and VEGF were significantly higher in active PDR than in eyes with inactive PDR (P = 0.016 and P = 0.008, respectively). Furthermore, a significant positive correlation was detected between Wnt3a and VEGF levels in the vitreous. CONCLUSION Intravitreous levels of Wnt3a and VEGF in patients with PDR are increased and correlated mutually. Wnt3a may be an important player in the development of diabetic retinopathy and its activity in vitreous fluid can be biomarker of PDR.
Collapse
|
181
|
Sewduth R, Santoro MM. "Decoding" Angiogenesis: New Facets Controlling Endothelial Cell Behavior. Front Physiol 2016; 7:306. [PMID: 27493632 PMCID: PMC4954849 DOI: 10.3389/fphys.2016.00306] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/06/2016] [Indexed: 01/08/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a unique and crucial biological process occurring during both development and adulthood. A better understanding of the mechanisms that regulates such process is mandatory to intervene in pathophysiological conditions. Here we highlight some recent argument on new players that are critical in endothelial cells, by summarizing novel discoveries that regulate notorious vascular pathways such as Vascular Endothelial Growth Factor (VEGF), Notch and Planar Cell Polarity (PCP), and by discussing more recent findings that put metabolism, redox signaling and hemodynamic forces as novel unforeseen facets in angiogenesis. These new aspects, that critically regulate angiogenesis and vascular homeostasis in health and diseased, represent unforeseen new ground to develop anti-angiogenic therapies.
Collapse
Affiliation(s)
- Raj Sewduth
- Laboratory of Endothelial Molecular Biology, Department of Oncology, Vesalius Research Center, VIB, KU Leuven Leuven, Belgium
| | - Massimo M Santoro
- Laboratory of Endothelial Molecular Biology, Department of Oncology, Vesalius Research Center, VIB, KU LeuvenLeuven, Belgium; Department of Molecular Biotechnology and Health Sciences, University of TurinTorino, Italy
| |
Collapse
|
182
|
Roker LA, Nemri K, Yu J. Wnt7b Signaling from the Ureteric Bud Epithelium Regulates Medullary Capillary Development. J Am Soc Nephrol 2016; 28:250-259. [PMID: 27432740 DOI: 10.1681/asn.2015111205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/26/2016] [Indexed: 01/29/2023] Open
Abstract
The renal vasculature is integral to the physiologic function of the kidneys in regulating hemodynamics of the body and maintaining organ health. The close inter-relationship of capillaries and the renal epithelium is key to renal physiology, but how renal tubules regulate capillary development remains unclear. Our previous work showed that Wnt7b is expressed in the ureteric trunk epithelium and activates canonical Wnt signaling in the surrounding medullary interstitium, where the capillaries reside. In this study, we showed by immunofluorescence that the target interstitial cells of Wnt7b/canonical Wnt signaling are mural cells of periureteric bud capillaries in the nascent renal medulla of embryonic mice. Genetic ablation of Wnt7b enhanced the proliferation of Wnt7b target mural cells, an effect that associated with decreased expression of PDGFRβ and p57kip2, a cyclin-dependent kinase inhibitor, in these cells. Furthermore, Wnt7b regulated lumen formation of the capillary endothelium in the renal medulla. In the absence of Wnt7b signaling, the periureteric bud medullary capillaries displayed narrower lumens lined with less flattened endothelial cells and a significantly increased presence of luminal endothelial cell-cell junctions, a transient configuration in the forming blood vessels in the controls. Moreover, the absence of Wnt7b led to greatly diminished levels of vascular endothelial (VE)-cadherin at the cell surface in these blood vessels. VE-cadherin is essential for blood vessel lumen formation; thus, Wnt7b may regulate lumen formation through modulation of VE-cadherin localization. Overall, these results indicate a novel role of Wnt7b signaling and the ureteric bud epithelium in renal medullary capillary development.
Collapse
Affiliation(s)
| | | | - Jing Yu
- Department of Cell Biology, .,Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia.,Center for Immunity, Inflammation and Regenerative Medicine and
| |
Collapse
|
183
|
Chow BW, Gu C. The molecular constituents of the blood-brain barrier. Trends Neurosci 2016; 38:598-608. [PMID: 26442694 DOI: 10.1016/j.tins.2015.08.003] [Citation(s) in RCA: 277] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) maintains the optimal microenvironment in the central nervous system (CNS) for proper brain function. The BBB comprises specialized CNS endothelial cells with fundamental molecular properties essential for the function and integrity of the BBB. The restrictive nature of the BBB hinders the delivery of therapeutics for many neurological disorders. In addition, recent evidence shows that BBB dysfunction can precede or hasten the progression of several neurological diseases. Despite the physiological significance of the BBB in health and disease, major discoveries of the molecular regulators of BBB formation and function have occurred only recently. This review highlights recent findings describing the molecular determinants and core cellular pathways that confer BBB properties on CNS endothelial cells.
Collapse
Affiliation(s)
- Brian Wai Chow
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MN 02115, USA
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MN 02115, USA.
| |
Collapse
|
184
|
Jiang X, Cong F. Novel Regulation of Wnt Signaling at the Proximal Membrane Level. Trends Biochem Sci 2016; 41:773-783. [PMID: 27377711 DOI: 10.1016/j.tibs.2016.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/03/2016] [Accepted: 06/07/2016] [Indexed: 02/06/2023]
Abstract
Wnt pathways are crucial for embryonic development and adult tissue homeostasis in all multicellular animals. Our understanding of Wnt signaling networks has grown increasingly complex. Recent studies have revealed many regulatory proteins that function at the proximal membrane level to fine-tune signaling output and enhance signaling specificity. These proteins regulate crucial points in Wnt signaling, including post-translational modification of Wnt proteins, regulation of Wnt receptor degradation, internalization of Wnt receptor complex, and specific ligand-receptor complex formation. Such regulators not only provide us with molecular details of Wnt regulation but also serve as potential targets for therapeutic intervention. In this review we highlight new insights into Wnt regulation at the plasma membrane, especially newly identified feedback regulators.
Collapse
Affiliation(s)
- Xiaomo Jiang
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Feng Cong
- Novartis Institutes for Biomedical Research, 181 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
185
|
Scholz B, Korn C, Wojtarowicz J, Mogler C, Augustin I, Boutros M, Niehrs C, Augustin HG. Endothelial RSPO3 Controls Vascular Stability and Pruning through Non-canonical WNT/Ca(2+)/NFAT Signaling. Dev Cell 2016; 36:79-93. [PMID: 26766444 DOI: 10.1016/j.devcel.2015.12.015] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 11/16/2015] [Accepted: 12/11/2015] [Indexed: 12/15/2022]
Abstract
The WNT signaling enhancer R-spondin3 (RSPO3) is prominently expressed in the vasculature. Correspondingly, embryonic lethality of Rspo3-deficient mice is caused by vessel remodeling defects. Yet the mechanisms underlying vascular RSPO3 function remain elusive. Inducible endothelial Rspo3 deletion (Rspo3-iECKO) resulted in perturbed developmental and tumor vascular remodeling. Endothelial cell apoptosis and vascular pruning led to reduced microvessel density in Rspo3-iECKO mice. Rspo3-iECKO mice strikingly phenocopied the non-canonical WNT signaling-induced vascular defects of mice deleted for the WNT secretion factor Evi/Wls. An endothelial screen for RSPO3 and EVI/WLS co-regulated genes identified Rnf213, Usp18, and Trim30α. RNF213 targets filamin A and NFAT1 for proteasomal degradation attenuating non-canonical WNT/Ca(2+) signaling. Likewise, USP18 and TRIM5α inhibited NFAT1 activation. Consequently, NFAT protein levels were decreased in endothelial cells of Rspo3-iECKO mice and pharmacological NFAT inhibition phenocopied Rspo3-iECKO mice. The data identify endothelial RSPO3-driven non-canonical WNT/Ca(2+)/NFAT signaling as a critical maintenance pathway of the remodeling vasculature.
Collapse
Affiliation(s)
- Beate Scholz
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Claudia Korn
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jessica Wojtarowicz
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Carolin Mogler
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Institute of Pathology, Heidelberg University, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Iris Augustin
- Division of Signaling and Functional Genomics, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Cell and Molecular Biology (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Cell and Molecular Biology (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; DNA Demethylation, DNA Repair and Reprogramming, Institute of Molecular Biology, Ackermannweg 4, 55128 Mainz, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; German Cancer Consortium, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| |
Collapse
|
186
|
Jin G, Yang Y, Liu H, Liu K, Zhao J, Chen X, Zhang X, Zhang Y, Lu J, Dong Z. Genome-wide analysis of the effect of esophageal squamous cell carcinoma on human umbilical vein endothelial cells. Oncol Rep 2016; 36:155-64. [PMID: 27222202 DOI: 10.3892/or.2016.4816] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/05/2016] [Indexed: 11/06/2022] Open
Abstract
A large volume of data indicates that controlling tumor-associated angiogenesis is a promising therapy against cancer. However, angiogenesis is a complex process, little is known about the differential gene expression in the process of normal endothelial cell differentiation toward tumor vascular endothelial cells induced by tumor microenvironment. The aim of this study is to investigate the effect of tumor microenvironment simulated by the supernatant of esophageal squamous cancer cells (KYSE70) on normal endothelial cells (HUVECs) at the whole genome level. The gene expression profile was studied through gene ontology and signal pathway analysis. Compared with the normal HUVECs, a total of 3769 differentially expressed genes in induced HUVECs were detected, including 1609 upregulated genes and 2160 downregulated genes. Moreover, the microarray data analysis showed that 11 significant biological processes and 10 significant signaling pathways changed most, which are associated with angiogenesis and cell differentiation. According to the different expression levels in the microarrays and their functions, four differentially expressed genes involved in tumor angiogenesis and cell differentiation (IL6, VEGFA, S1PR1, TYMP) were selected and analyzed by qRT-PCR. The qRT-PCR results were consistent with the microarray data. Furthermore, we simulated the tumor microenvironment by human esophageal carcinoma tissue homogenate to investigate its effect on HUVECs, the qRT-PCR results indicated that the above genes were highly expressed in HUVECs after induction by esophageal carcinoma tissue homogenate. In conclusion, tumor microenvironment impact on normal endothelial cells differentiated toward tumor vascular endothelial cells, and the selected genes, which are associated with tumor angiogenesis, would be anti-angiogenesis targets against esophageal carcinoma.
Collapse
Affiliation(s)
- Guoguo Jin
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yi Yang
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Hangfan Liu
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Kangdong Liu
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jimin Zhao
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xinhuan Chen
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiaoyan Zhang
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yanyan Zhang
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jing Lu
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Ziming Dong
- Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
187
|
Feng J, Xian Q, Guan T, Hu J, Wang M, Huang Y, So KF, Evans SM, Chai G, Goffinet AM, Qu Y, Zhou L. Celsr3 and Fzd3 Organize a Pioneer Neuron Scaffold to Steer Growing Thalamocortical Axons. Cereb Cortex 2016; 26:3323-34. [PMID: 27170656 PMCID: PMC4898681 DOI: 10.1093/cercor/bhw132] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Celsr3 and Fzd3 regulate the development of reciprocal thalamocortical projections independently of their expression in cortical or thalamic neurons. To understand this cell non autonomous mechanism further, we tested whether Celsr3 and Fzd3 could act via Isl1-positive guidepost cells. Isl1-positive cells appear in the forebrain at embryonic day (E) 9.5-E10.5 and, from E12.5, they form 2 contingents in ventral telencephalon and prethalamus. In control mice, corticothalamic axons run in the ventral telencephalic corridor in close contact with Isl1-positive cells. When Celsr3 or Fzd3 is inactivated in Isl1-expressing cells, corticofugal fibers stall and loop in the ventral telencephalic corridor of high Isl1 expression, and thalamic axons fail to cross the diencephalon–telencephalon junction (DTJ). At E12.5, before thalamic and cortical axons emerge, pioneer projections from Isl1-positive cells cross the DTJ from both sides in control but not mutant embryos. These early projections appear to act like a bridge to guide later growing thalamic axons through the DTJ. Our data suggest that Celsr3 and Fzd3 orchestrate the formation of a scaffold of pioneer neurons and their axons. This scaffold extends from prethalamus to ventral telencephalon and subcortex, and steers reciprocal corticothalamic fibers.
Collapse
Affiliation(s)
- Jia Feng
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Quanxiang Xian
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Tingting Guan
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Jing Hu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Meizhi Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Yuhua Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China Department of Anatomy, The University of Hong Kong Pokfulam, Hong Kong SAR, PR China
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Guoliang Chai
- Institute of Neuroscience, Université catholique de Louvain, Brussels B1200, Belgium
| | - Andre M Goffinet
- Institute of Neuroscience, Université catholique de Louvain, Brussels B1200, Belgium
| | - Yibo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China Co-innovation Center of Neuroregeneration, Jiangsu, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Guangdong key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou 510632, PR China Co-innovation Center of Neuroregeneration, Jiangsu, China
| |
Collapse
|
188
|
Noda M, Vallon M, Kuo CJ. The Wnt7's Tale: A story of an orphan who finds her tie to a famous family. Cancer Sci 2016; 107:576-82. [PMID: 26934061 PMCID: PMC4970824 DOI: 10.1111/cas.12924] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/22/2016] [Accepted: 02/29/2016] [Indexed: 12/11/2022] Open
Abstract
The transformation suppressor gene RECK was isolated by cDNA expression cloning (1998), and GPR124/TEM5 was detected as a tumor endothelial marker by differential screening (2000). The importance of Wnt7a/b and Gpr124 in brain angiogenesis was demonstrated by reverse genetics in mice (2008–2010). A series of recent studies using genetically engineered mice and zebrafish as well as luciferase reporter assays in cultured cells led to the discovery of functional interactions among Reck, Gpr124, and Wnt7a/b in triggering canonical Wnt signaling with relevance to embryonic brain angiogenesis and blood–brain barrier formation.
Collapse
Affiliation(s)
- Makoto Noda
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mario Vallon
- Hematology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Calvin J Kuo
- Hematology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
189
|
Zhao Z, Nelson AR, Betsholtz C, Zlokovic BV. Establishment and Dysfunction of the Blood-Brain Barrier. Cell 2016; 163:1064-1078. [PMID: 26590417 DOI: 10.1016/j.cell.2015.10.067] [Citation(s) in RCA: 1178] [Impact Index Per Article: 130.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Indexed: 12/11/2022]
Abstract
Structural and functional brain connectivity, synaptic activity, and information processing require highly coordinated signal transduction between different cell types within the neurovascular unit and intact blood-brain barrier (BBB) functions. Here, we examine the mechanisms regulating the formation and maintenance of the BBB and functions of BBB-associated cell types. Furthermore, we discuss the growing evidence associating BBB breakdown with the pathogenesis of inherited monogenic neurological disorders and complex multifactorial diseases, including Alzheimer's disease.
Collapse
Affiliation(s)
- Zhen Zhao
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Amy R Nelson
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, 75185 Uppsala, Sweden
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
190
|
Lange C, Turrero Garcia M, Decimo I, Bifari F, Eelen G, Quaegebeur A, Boon R, Zhao H, Boeckx B, Chang J, Wu C, Le Noble F, Lambrechts D, Dewerchin M, Kuo CJ, Huttner WB, Carmeliet P. Relief of hypoxia by angiogenesis promotes neural stem cell differentiation by targeting glycolysis. EMBO J 2016; 35:924-41. [PMID: 26856890 DOI: 10.15252/embj.201592372] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/05/2016] [Indexed: 12/23/2022] Open
Abstract
Blood vessels are part of the stem cell niche in the developing cerebral cortex, but their in vivo role in controlling the expansion and differentiation of neural stem cells (NSCs) in development has not been studied. Here, we report that relief of hypoxia in the developing cerebral cortex by ingrowth of blood vessels temporo-spatially coincided with NSC differentiation. Selective perturbation of brain angiogenesis in vessel-specific Gpr124 null embryos, which prevented the relief from hypoxia, increased NSC expansion at the expense of differentiation. Conversely, exposure to increased oxygen levels rescued NSC differentiation in Gpr124 null embryos and increased it further in WT embryos, suggesting that niche blood vessels regulate NSC differentiation at least in part by providing oxygen. Consistent herewith, hypoxia-inducible factor (HIF)-1α levels controlled the switch of NSC expansion to differentiation. Finally, we provide evidence that high glycolytic activity of NSCs is required to prevent their precocious differentiation in vivo Thus, blood vessel function is required for efficient NSC differentiation in the developing cerebral cortex by providing oxygen and possibly regulating NSC metabolism.
Collapse
Affiliation(s)
- Christian Lange
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | | | - Ilaria Decimo
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Francesco Bifari
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Annelies Quaegebeur
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Ruben Boon
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Hui Zhao
- Laboratory of Translational Genetics, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Translational Genetics, Department of Oncology, KU Leuven Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Translational Genetics, Department of Oncology, KU Leuven Leuven, Belgium
| | - Junlei Chang
- Department of Medicine, Hematology Division Stanford University, Stanford, CA, USA
| | - Christine Wu
- Department of Medicine, Hematology Division Stanford University, Stanford, CA, USA
| | - Ferdinand Le Noble
- Angiogenesis and Cardiovascular Pathology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany Department of Cell and Developmental Biology, KIT, Karlsruhe, Germany
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Translational Genetics, Department of Oncology, KU Leuven Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Calvin J Kuo
- Department of Medicine, Hematology Division Stanford University, Stanford, CA, USA
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| |
Collapse
|
191
|
Abstract
Frizzled proteins are the principal receptors for the Wnt family of ligands. They mediate canonical Wnt signaling together with Lrp5 and Lrp6 coreceptors. In conjunction with Celsr, Vangl, and a small number of additional membrane and membrane-associated proteins, they also play a central role in tissue polarity/planar cell polarity (PCP) signaling. Targeted mutations in 9 of the 10 mammalian Frizzled genes have revealed their roles in an extraordinarily diverse set of developmental and homeostatic processes, including morphogenetic movements responsible for palate, ventricular septum, ocular furrow, and neural tube closure; survival of thalamic neurons; bone formation; central nervous system (CNS) angiogenesis and blood-brain barrier formation and maintenance; and a wide variety of processes that orient subcellular, cellular, and multicellular structures relative to the body axes. The last group likely reflects the mammalian equivalent of tissue polarity/PCP signaling, as defined in Drosophila, and it includes CNS axon guidance, hair follicle and tongue papilla orientation, and inner ear sensory hair bundle orientation. Frizzled receptors are ubiquitous among multicellular animals and, with other signaling molecules, they very likely evolved to permit the development of the complex tissue architectures that provide multicellular animals with their enormous selective advantage.
Collapse
Affiliation(s)
- Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hao Chang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
192
|
Bostaille N, Gauquier A, Stainier DYR, Raible DW, Vanhollebeke B. Defective adgra2 (gpr124) splicing and function in zebrafish ouchless mutants. Development 2016; 144:8-11. [DOI: 10.1242/dev.146803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ABSTRACT
A hitherto unidentified N-ethyl-N-nitrosourea (ENU)-induced mutation affects dorsal root ganglia (DRG) formation in ouchless mutant zebrafish larvae. In contrast to previous findings assigning the ouchless phenotypes to downregulated sorbs3 transcript levels, this work re-attributes the phenotypes to an essential splice site mutation affecting adgra2 (gpr124) splicing and function. Accordingly, ouchless mutants fail to complement previously characterized adgra2 mutants and exhibit highly penetrant cerebrovascular defects. The aberrantly spliced adgra2 transcript found in ouchless mutants encodes a receptor lacking a single leucine-rich repeat (LRR) within its N-terminus.
Collapse
Affiliation(s)
- Naguissa Bostaille
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Anne Gauquier
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - David W. Raible
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| |
Collapse
|
193
|
Musa G, Engel FB, Niaudet C. Heart Development, Angiogenesis, and Blood-Brain Barrier Function Is Modulated by Adhesion GPCRs. Handb Exp Pharmacol 2016; 234:351-368. [PMID: 27832496 DOI: 10.1007/978-3-319-41523-9_16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cardiovascular system in adult organisms forms a network of interconnected endothelial cells, supported by mural cells and displaying a high degree of hierarchy: arteries emerging from the heart ramify into arterioles and then capillaries, which return to the venous systems through venules and veins. The cardiovascular system allows blood circulation, which in turn is essential for hemostasis through gas diffusion, nutrient distribution, and cell trafficking. In this chapter, we have summarized the current knowledge on how adhesion GPCRs (aGPCRs) impact heart development, followed by their role in modulating vascular angiogenesis.
Collapse
Affiliation(s)
- Gentian Musa
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 12, Erlangen, 91054, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 12, Erlangen, 91054, Germany.
| | - Colin Niaudet
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, Uppsala, 751 85, Sweden.
| |
Collapse
|
194
|
Wang W, Li M, Wang Y, Li Q, Deng G, Wan J, Yang Q, Chen Q, Wang J. GSK-3β inhibitor TWS119 attenuates rtPA-induced hemorrhagic transformation and activates the Wnt/β-catenin signaling pathway after acute ischemic stroke in rats. Mol Neurobiol 2015; 53:7028-7036. [PMID: 26671619 DOI: 10.1007/s12035-015-9607-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
Abstract
Hemorrhagic transformation (HT) is a devastating complication for patients with acute ischemic stroke who are treated with tissue plasminogen activator (tPA). It is associated with high morbidity and mortality, but no effective treatments are currently available to reduce HT risk. Therefore, methods to prevent HT are urgently needed. In this study, we used TWS119, an inhibitor of glycogen synthase kinase 3β (GSK-3β), to evaluate the role of the Wnt/β-catenin signaling pathway in recombinant tPA (rtPA)-induced HT. Sprague-Dawley rats were subjected to a middle cerebral artery occlusion (MCAO) model of ischemic stroke and then were administered rtPA, rtPA combined with TWS119, or vehicle at 4 h. The animals were sacrificed 24 h after infarct induction. Rats treated with rtPA showed evident HT, had more severe neurologic deficit, brain edema, and blood-brain barrier breakdown, and had larger infarction volume than did the vehicle group. Rats treated with TWS119 had significantly improved outcomes compared with those of rats treated with rtPA alone. In addition, Western blot analysis showed that TWS119 increased the protein expression of β-catenin, claudin-3, and ZO-1 while suppressing the expression of GSK-3β. These results suggest that TWS119 reduces rtPA-induced HT and attenuates blood-brain barrier disruption, possibly through activation of the Wnt/β-catenin signaling pathway. This study provides a potential therapeutic strategy to prevent tPA-induced HT after acute ischemic stroke.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yuefei Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Li
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD, 21205, USA
| | - Gang Deng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jieru Wan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD, 21205, USA
| | - Qingwu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400044, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD, 21205, USA.
| |
Collapse
|
195
|
Ulrich F, Carretero-Ortega J, Menéndez J, Narvaez C, Sun B, Lancaster E, Pershad V, Trzaska S, Véliz E, Kamei M, Prendergast A, Kidd KR, Shaw KM, Castranova DA, Pham VN, Lo BD, Martin BL, Raible DW, Weinstein BM, Torres-Vázquez J. Reck enables cerebrovascular development by promoting canonical Wnt signaling. Development 2015; 143:147-59. [PMID: 26657775 DOI: 10.1242/dev.123059] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 11/25/2015] [Indexed: 01/03/2023]
Abstract
The cerebral vasculature provides the massive blood supply that the brain needs to grow and survive. By acquiring distinctive cellular and molecular characteristics it becomes the blood-brain barrier (BBB), a selectively permeable and protective interface between the brain and the peripheral circulation that maintains the extracellular milieu permissive for neuronal activity. Accordingly, there is great interest in uncovering the mechanisms that modulate the formation and differentiation of the brain vasculature. By performing a forward genetic screen in zebrafish we isolated no food for thought (nft (y72)), a recessive late-lethal mutant that lacks most of the intracerebral central arteries (CtAs), but not other brain blood vessels. We found that the cerebral vascularization deficit of nft (y72) mutants is caused by an inactivating lesion in reversion-inducing cysteine-rich protein with Kazal motifs [reck; also known as suppressor of tumorigenicity 15 protein (ST15)], which encodes a membrane-anchored tumor suppressor glycoprotein. Our findings highlight Reck as a novel and pivotal modulator of the canonical Wnt signaling pathway that acts in endothelial cells to enable intracerebral vascularization and proper expression of molecular markers associated with BBB formation. Additional studies with cultured endothelial cells suggest that, in other contexts, Reck impacts vascular biology via the vascular endothelial growth factor (VEGF) cascade. Together, our findings have broad implications for both vascular and cancer biology.
Collapse
Affiliation(s)
- Florian Ulrich
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Jorge Carretero-Ortega
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Javier Menéndez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Carlos Narvaez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Belinda Sun
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Eva Lancaster
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Valerie Pershad
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Sean Trzaska
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Evelyn Véliz
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Makoto Kamei
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew Prendergast
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Kameha R Kidd
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenna M Shaw
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel A Castranova
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brigid D Lo
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jesús Torres-Vázquez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
196
|
Zhou Y, Williams J, Smallwood PM, Nathans J. Sox7, Sox17, and Sox18 Cooperatively Regulate Vascular Development in the Mouse Retina. PLoS One 2015; 10:e0143650. [PMID: 26630461 PMCID: PMC4667919 DOI: 10.1371/journal.pone.0143650] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 11/06/2015] [Indexed: 02/06/2023] Open
Abstract
Vascular development and maintenance are controlled by a complex transcriptional program, which integrates both extracellular and intracellular signals in endothelial cells. Here we study the roles of three closely related SoxF family transcription factors–Sox7, Sox17, and Sox18 –in the developing and mature mouse vasculature using targeted gene deletion on a mixed C57/129/CD1 genetic background. In the retinal vasculature, each SoxF gene exhibits a distinctive pattern of expression in different classes of blood vessels. On a mixed genetic background, vascular endothelial-specific deletion of individual SoxF genes has little or no effect on vascular architecture or differentiation, a result that can be explained by overlapping function and by reciprocal regulation of gene expression between Sox7 and Sox17. By contrast, combined deletion of Sox7, Sox17, and Sox18 at the onset of retinal angiogenesis leads to a dense capillary plexus with a nearly complete loss of radial arteries and veins, whereas the presence of a single Sox17 allele largely restores arterial identity, as determined by vascular smooth muscle cell coverage. In the developing retina, expression of all three SoxF genes is reduced in the absence of Norrin/Frizzled4-mediated canonical Wnt signaling, but SoxF gene expression is unaffected by reduced VEGF signaling in response to deletion of Neuropilin1 (Npn1). In adulthood, Sox7, Sox17, and Sox18 act in a largely redundant manner to maintain blood vessel function, as adult onset vascular endothelial-specific deletion of all three SoxF genes leads to massive edema despite nearly normal vascular architecture. These data reveal critical and partially redundant roles for Sox7, Sox17 and Sox18 in vascular growth, differentiation, and maintenance.
Collapse
Affiliation(s)
- Yulian Zhou
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - John Williams
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Philip M. Smallwood
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Jeremy Nathans
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- * E-mail:
| |
Collapse
|
197
|
|
198
|
Keaney J, Campbell M. The dynamic blood-brain barrier. FEBS J 2015; 282:4067-79. [DOI: 10.1111/febs.13412] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/20/2015] [Accepted: 08/12/2015] [Indexed: 01/19/2023]
Affiliation(s)
- James Keaney
- Smurfit Institute of Genetics; Trinity College Dublin; Ireland
| | | |
Collapse
|
199
|
Broux B, Gowing E, Prat A. Glial regulation of the blood-brain barrier in health and disease. Semin Immunopathol 2015; 37:577-90. [PMID: 26245144 DOI: 10.1007/s00281-015-0516-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/15/2015] [Indexed: 12/14/2022]
Abstract
The brain is the organ with the highest metabolic demand in the body. Therefore, it needs specialized vasculature to provide it with the necessary oxygen and nutrients, while protecting it against pathogens and toxins. The blood-brain barrier (BBB) is very tightly regulated by specialized endothelial cells, two basement membranes, and astrocytic endfeet. The proximity of astrocytes to the vessel makes them perfect candidates to influence the function of the BBB. Moreover, other glial cells are also known to contribute to either BBB quiescence or breakdown. In this review, we summarize the knowledge on glial regulation of the BBB during development, in homeostatic conditions in the adult, and during neuroinflammatory responses.
Collapse
Affiliation(s)
- Bieke Broux
- Neuroimmunology Unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint Denis Street, Room R9.912, Montréal, Québec, Canada, H2X 0A9
- Department of Neuroscience, Faculté de Médecine, Université de Montréal, Montréal, Canada
- Hasselt University, Biomedical Research Institute and transnationale Universiteit Limburg, School of Life Sciences, Agoralaan, Building C, 3590, Diepenbeek, Belgium
| | - Elizabeth Gowing
- Neuroimmunology Unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint Denis Street, Room R9.912, Montréal, Québec, Canada, H2X 0A9
- Department of Neuroscience, Faculté de Médecine, Université de Montréal, Montréal, Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Rue Saint Denis Street, Room R9.912, Montréal, Québec, Canada, H2X 0A9.
- Department of Neuroscience, Faculté de Médecine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
200
|
Jiang L, Yin M, Wei X, Liu J, Wang X, Niu C, Kang X, Xu J, Zhou Z, Sun S, Wang X, Zheng X, Duan S, Yao K, Qian R, Sun N, Chen A, Wang R, Zhang J, Chen S, Meng D. Bach1 Represses Wnt/β-Catenin Signaling and Angiogenesis. Circ Res 2015; 117:364-375. [PMID: 26123998 PMCID: PMC4676728 DOI: 10.1161/circresaha.115.306829] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/29/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Wnt/β-catenin signaling has an important role in the angiogenic activity of endothelial cells (ECs). Bach1 is a transcription factor and is expressed in ECs, but whether Bach1 regulates angiogenesis is unknown. OBJECTIVE This study evaluated the role of Bach1 in angiogenesis and Wnt/β-catenin signaling. METHODS AND RESULTS Hind-limb ischemia was surgically induced in Bach1(-/-) mice and their wild-type littermates and in C57BL/6J mice treated with adenoviruses coding for Bach1 or GFP. Lack of Bach1 expression was associated with significant increases in perfusion and vascular density and in the expression of proangiogenic cytokines in the ischemic hindlimb of mice, with enhancement of the angiogenic activity of ECs (eg, tube formation, migration, and proliferation). Bach1 overexpression impaired angiogenesis in mice with hind-limb ischemia and inhibited Wnt3a-stimulated angiogenic response and the expression of Wnt/β-catenin target genes, such as interleukin-8 and vascular endothelial growth factor, in human umbilical vein ECs. Interleukin-8 and vascular endothelial growth factor were responsible for the antiangiogenic response of Bach1. Immunoprecipitation and GST pull-down assessments indicated that Bach1 binds directly to TCF4 and reduces the interaction of β-catenin with TCF4. Bach1 overexpression reduces the interaction between p300/CBP and β-catenin, as well as β-catenin acetylation, and chromatin immunoprecipitation experiments confirmed that Bach1 occupies the TCF4-binding site of the interleukin-8 promoter and recruits histone deacetylase 1 to the interleukin-8 promoter in human umbilical vein ECs. CONCLUSIONS Bach1 suppresses angiogenesis after ischemic injury and impairs Wnt/β-catenin signaling by disrupting the interaction between β-catenin and TCF4 and by recruiting histone deacetylase 1 to the promoter of TCF4-targeted genes.
Collapse
Affiliation(s)
- Li Jiang
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Meng Yin
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Xiangxiang Wei
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Junxu Liu
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Xinhong Wang
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Cong Niu
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Xueling Kang
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Jie Xu
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Zhongwei Zhou
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Shaoyang Sun
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Xu Wang
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Xiaojun Zheng
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Shengzhong Duan
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Kang Yao
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Ning Sun
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Alex Chen
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Rui Wang
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Jianyi Zhang
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Sifeng Chen
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| | - Dan Meng
- Department of Physiology and Pathophysiology (L.J., Xiangxiang Wei, J.L., Xinhong Wang, C.N., X.K., J.X., Z.Z., R.Q., N.S., A.C., R.W., S.C., D.M.) and Department of Biochemistry and Molecular Biology (S.S., Xu Wang), School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China (M.Y.); Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China (X.Z., S.D.); Department of Cardiology, Shanghai Institute of Cardiovascular Disease, ZhongShan Hospital, Fudan University, Shanghai, China (K.Y.); Center for Vascular Disease and Translational Medicine, Xiangya Third Hospital, Central South University, Changsha, China (A.C.); Department of Biology, Laurentian University, Sudbury, Ontario, Canada (R.W.); and Division of Cardiology, Department of Medicine, Stem Cell Institute, University of Minnesota Medical School, Minneapolis (J.Z.)
| |
Collapse
|