1
|
Wang Y, Yang M, Wang X, Zou H, Chen X, Yuan R. Role of Gpr124 in the Migration and Proliferation of Retinal Microvascular Endothelial Cells and Microangiopathies in Diabetic Retinopathy. Mol Biotechnol 2025; 67:2467-2480. [PMID: 38862861 DOI: 10.1007/s12033-024-01210-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/27/2024] [Indexed: 06/13/2024]
Abstract
Retinal microangiopathies, such as neovascularization and preretinal and vitreous hemorrhages, are the primary pathological features of diabetic retinopathy (DR). These conditions can worsen visual impairment and may result in blindness. Furthermore, multiple metabolic pathways are associated with microangiopathy in DR. However, the specific underlying pathological mechanisms remain unclear. Several studies have demonstrated the important role of G protein-coupled receptor 124 (Gpr124) in cerebral vascular endothelial cells, but its effect on the retinal endothelium has not been elucidated. In this study, we found that Gpr124 is expressed in both pathological retinal fibrous vascular membranes of DR patients and retinal blood vessels of mice, with elevated protein expression specifically observed in the retinas of DR model mice. Furthermore, Gpr124 expression was elevated after high-glucose treatment of human retinal microvascular endothelial cells (HRMECs). Inhibition of Gpr124 expression affected the high glucose-induced proliferation, migration, and tube-forming ability of HRMECs. We concluded that Gpr124 expression was upregulated in DR and promoted HRMECs angiogenesis in a high-glucose environment. This finding may help to elucidate the pathogenesis of DR and provide a critical research basis for identifying effective treatments.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Xinqiao Road, Shapingba District, Chongqing, 400037, China
| | - Mei Yang
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Xinqiao Road, Shapingba District, Chongqing, 400037, China
| | - Xuan Wang
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huan Zou
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Xinqiao Road, Shapingba District, Chongqing, 400037, China.
| | - Xiaofan Chen
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Xinqiao Road, Shapingba District, Chongqing, 400037, China
| | - Rongdi Yuan
- Department of Ophthalmology, Xinqiao Hospital, Army Medical University, Xinqiao Road, Shapingba District, Chongqing, 400037, China.
| |
Collapse
|
2
|
He T, Zhang M, Qin J, Wang Y, Li S, Du C, Jiao J, Ji F. Endothelial PD-1 Regulates Vascular Homeostasis and Oligodendrogenesis during Brain Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417410. [PMID: 40013943 PMCID: PMC12021089 DOI: 10.1002/advs.202417410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/14/2025] [Indexed: 02/28/2025]
Abstract
Appropriate vascular and neural development is essential for central nervous system (CNS). Although programmed cell death receptor 1 (PD-1) mediates neurogenesis, its role in cerebrovascular development remains poorly understood. Here, a correlation between cerebral vessels and oligodendrocyte precursor cells (OPCs) is revealed during brain development. The ablation of endothelial PD-1 triggers cortical hypervascularization through excessive angiogenic sprouting, concomitantly driving OPC differentiation. These alterations disrupt blood brain barrier (BBB) maturation, induce dysmyelination, and ultimately result in abnormal behavior in mice. Mechanistically, the loss of endothelial PD-1 suppresses the activity of the Wnt/β-catenin signaling pathway, thereby disrupting normal angiogenesis. Concurrently, it activates the MEK1/2-ERK1/2-GLI1 pathway, leading to increased GREMLIN1 (GREM1) expression. Elevated GREM1 secretion inhibits the BMP/SMAD1/5/SMAD4 signaling cascade in OPCs, which inhibits oligodendrogenesis and myelination. These findings indicate the importance of endothelial cell-intrinsic PD-1 in regulating the oligovascular niche, and suggest potential therapeutic implications for neurological disorders associated with disrupted vascular development.
Collapse
Affiliation(s)
- Tingting He
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing100190China
| | - Mengtian Zhang
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jie Qin
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yanyan Wang
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Sihan Li
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Chaoyi Du
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jianwei Jiao
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| | - Fen Ji
- State Key Laboratory of Organ Regeneration and Reconstruction,Institute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| |
Collapse
|
3
|
Li Y, Duan Y, Chu Q, Lv H, Li J, Guo X, Gao Y, Liu M, Tang W, Hu H, Liu H, Sun J, Wang X, Yi F. G-protein coupled receptor GPR124 protects against podocyte senescence and injury in diabetic kidney disease. Kidney Int 2025; 107:652-665. [PMID: 39828038 DOI: 10.1016/j.kint.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
Although emerging studies highlight the pivotal role of podocyte senescence in the pathogenesis of diabetic kidney disease (DKD) and aging-related kidney diseases, therapeutic strategies for preventing podocyte senescence are still lacking. Here, we identified a previously unrecognized role of GPR124, a novel adhesion G protein-coupled receptor, in maintaining podocyte structure and function by regulation of cellular senescence in DKD. Podocyte GPR124 was significantly reduced in db/db diabetic (a type 2 diabetic mouse model) and streptozocin-induced diabetic mice (a type 1 diabetic model), which was further confirmed in kidney biopsies from patients with DKD. The level of GPR124 in glomeruli was positively correlated with the estimated glomerular filtration rate and negatively correlated with serum creatinine levels. Podocyte-specific deficiency of GPR124 significantly aggravated podocyte injury and proteinuria in the two models of diabetic mice. Moreover, GPR124 regulated podocyte senescence in both diabetic and aged mice. Mechanistically, GPR124 directly bound with vinculin and negatively regulated focal adhesion kinase (FAK) signaling, thereby mediating podocyte senescence and function. Importantly, overexpression of GPR124 or pharmacological inhibition of FAK protected against podocyte senescence and injury under diabetic conditions. Our studies suggest that targeting GPR124 may be an innovative therapeutic strategy for patients with DKD and aging-related kidney diseases.
Collapse
MESH Headings
- Podocytes/pathology
- Podocytes/metabolism
- Animals
- Cellular Senescence
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/genetics
- Diabetic Nephropathies/etiology
- Diabetic Nephropathies/prevention & control
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/deficiency
- Humans
- Male
- Mice, Inbred C57BL
- Signal Transduction
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/metabolism
- Mice
- Mice, Knockout
- Focal Adhesion Kinase 1/metabolism
- Focal Adhesion Kinase 1/antagonists & inhibitors
- Proteinuria/pathology
- Proteinuria/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/metabolism
- Glomerular Filtration Rate
- Cells, Cultured
Collapse
Affiliation(s)
- Yujia Li
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, China
| | - Yiqi Duan
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qingqing Chu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hang Lv
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jing Li
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiangyun Guo
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yanjiao Gao
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Min Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Huili Hu
- Department of Systems Biomedicine and Research Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Jinpeng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Xiaojie Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, China.
| |
Collapse
|
4
|
Olaniru OE, Toczyska K, Guccio N, Giera S, Piao X, King AJF, Jones PM, Persaud SJ. Spatiotemporal profiling of adhesion G protein-coupled receptors in developing mouse and human pancreas reveals a role for GPR56 in islet development. Cell Mol Life Sci 2025; 82:129. [PMID: 40137991 PMCID: PMC11947406 DOI: 10.1007/s00018-025-05659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
INTRODUCTION G protein-coupled receptors (GPCRs) are cell-surface proteins that are targeted therapeutically for a range of disorders, including diabetes. Adhesion GPCRs (aGPCRs) are the second largest class of the GPCR superfamily and some members of this family have been implicated in appropriate organ development. However, the role of aGPCRs in endocrine pancreas specification is not yet known. METHODS Here, we systematically characterised expression of mRNAs encoding aGPCRs and their ligands in developing mouse and human pancreas using our own and publicly available single-cell RNA sequencing and spatial transcriptomics data, and we conducted qPCR analysis of aGPCR expression in human pancreas at different gestational stages. We then investigated the role of GPR56 (ADGRG1), the most abundant aGPCR in pancreatic endocrine progenitors, in islet development using Gpr56 null mice and their wildtype littermates. RESULTS We demonstrated that aGPCRs are dynamically expressed during mouse and human pancreas development, with specific aGPCR mRNAs expressed in distinct endocrine, endothelial, mesenchymal, acinar, ductal, and immune cell clusters. aGPCR ligand mRNAs were mostly expressed by non-endocrine cells, and the most highly expressed receptor-ligand interacting mRNA pairs were those encoding GPR56 and COL3A1. Deletion of Gpr56 in neonatal mice was associated with an altered α-/β-/δ-cell ratio and reduced β-cell proliferation. CONCLUSION Our data show that aGPCRs are expressed at key stages of human and mouse pancreas endocrine lineage decisions, and analysis of pancreases from Gpr56 knockout mice implicate this aGPCR in the development of a full complement of β-cells.
Collapse
Affiliation(s)
- Oladapo E Olaniru
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Klaudia Toczyska
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Nunzio Guccio
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Stefanie Giera
- Department of Medicine, Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xianhua Piao
- Department of Medicine, Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pediatrics, University of California at San Francisco, San Francisco, CA, USA
| | - Aileen J F King
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Peter M Jones
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Shanta J Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
5
|
Sebo DJ, Ali I, Fetsko AR, Trimbach AA, Taylor MR. Activation of Wnt/β-catenin in neural progenitor cells regulates blood-brain barrier development and promotes neuroinflammation. Sci Rep 2025; 15:3496. [PMID: 39875426 PMCID: PMC11775206 DOI: 10.1038/s41598-025-85784-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
The central nervous system (CNS) requires specialized blood vessels to support neural function within specific microenvironments. During neurovascular development, endothelial Wnt/β-catenin signaling is required for BBB development within the brain parenchyma, whereas fenestrated blood vessels that lack BBB properties do not require Wnt/β-catenin signaling. Here, we used zebrafish to further characterize this phenotypic heterogeneity of the CNS vasculature. Using transgenic reporters of Wnt/β-catenin transcriptional activity, we found an inverse correlation between activated Wnt/β-catenin signaling in endothelial cells (ECs) versus non-ECs within these distinct microenvironments. Our results indicated that the level of Wnt/β-catenin signaling in non-ECs may regulate Wnt/β-catenin activity in adjacent ECs. To further test this concept, we generated a transgenic Tet-On inducible system to drive constitutively active β-catenin expression in neural progenitor cells (NPCs). We found that dose-dependent activation of Wnt/β-catenin in NPCs caused severe deficiency in CNS angiogenesis and BBB development. Additionally, we discovered a significant increase in the proliferation of microglia and infiltration of peripheral neutrophils indicative of a stereotypical neuroinflammatory response. In conclusion, our results demonstrate the importance of proper Wnt/β-catenin signaling within specific CNS microenvironments and highlights the potentially deleterious consequences of aberrant Wnt activation.
Collapse
Affiliation(s)
- Dylan J Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Irshad Ali
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Audrey R Fetsko
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Aubrey A Trimbach
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael R Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
6
|
Lee NJ, Parab S, Lam AE, Leong JX, Matsuoka RL. Angiogenic mechanisms governing the segregation of blood-brain barrier and fenestrated capillaries derived from a multipotent cerebrovascular niche. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.10.627641. [PMID: 39868183 PMCID: PMC11760744 DOI: 10.1101/2024.12.10.627641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Cerebrovascular endothelial cell (EC) subtypes characterized by blood-brain barrier (BBB) properties or fenestrated pores are essential components of brain-blood interfaces, supporting brain function and homeostasis. To date, the origins and developmental mechanisms underlying this heterogeneous EC network remain largely unclear. Using single-cell-resolution lineage tracing in zebrafish, we discover a multipotent vascular niche at embryonic capillary borders that generates ECs with BBB or fenestrated molecular identity. RNAscope analysis demonstrates restricted expression of flt4 in sprouting ECs contributing to fenestrated choroid plexus (CP) vasculature, identifying an early molecular distinction from adjacent BBB vessels. Mechanistically, flt4 null and cytoplasmic-domain-deletion mutants exhibit CP vascularization defects when combined with vegfr2 zebrafish paralog deletion. Pharmacological results support this co-requirement of Flt4 and Vegfr2 signaling and suggest the PI3K and ERK pathways as downstream effectors. These findings reveal a specialized developmental origin for BBB and fenestrated EC subtypes, and establish Flt4 as a crucial guidance receptor mediating their angiogenic segregation.
Collapse
|
7
|
Lin HH. An Alternative Mode of GPCR Transactivation: Activation of GPCRs by Adhesion GPCRs. Int J Mol Sci 2025; 26:552. [PMID: 39859266 PMCID: PMC11765499 DOI: 10.3390/ijms26020552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
G protein-coupled receptors (GPCRs), critical for cellular communication and signaling, represent the largest cell surface protein family and play important roles in numerous pathophysiological processes. Consequently, GPCRs have become a primary focus in drug discovery efforts. Beyond their traditional G protein-dependent signaling pathways, GPCRs are also capable of activating alternative signaling mechanisms, including G protein-independent signaling, biased signaling, and signaling crosstalk. A particularly novel signaling mode employed by these receptors is GPCR transactivation, which enables cross-communication between GPCRs and other receptor types. Intriguingly, GPCR transactivation by distinct GPCRs has also been identified. In this review, I provide an overview of the known GPCR transactivation mechanisms and explore recently uncovered GPCR transactivation mediated by adhesion-class GPCRs (aGPCRs). These aGPCR-GPCR transactivation processes regulate unique cell type-specific functions, offering an exciting opportunity to develop therapies that precisely modulate specific GPCR-mediated biological effects.
Collapse
Affiliation(s)
- Hsi-Hsien Lin
- Department of Microbiology and Immunology, Graduate School of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; ; Tel.: +886-03-2118800-3321
- Center for Molecular and Clinical Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung 20401, Taiwan
| |
Collapse
|
8
|
Lin WY, Dong YL, Lin Y, Sunchuri D, Guo ZL. Potential role of G protein‑coupled receptor 124 in cardiovascular and cerebrovascular disease (Review). Exp Ther Med 2025; 29:2. [PMID: 39534284 PMCID: PMC11552082 DOI: 10.3892/etm.2024.12752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
G protein-coupled receptor 124 (GPR124) has a key role in regulating the proliferation and differentiation of endothelial cells, activating inflammatory bodies and promoting angiogenesis and other processes, thus affecting various pathological and physiological processes in the body. GPR124 is vital for promoting the development of the nervous system and maintaining the stability of the blood-brain barrier, and is also associated with cardiovascular and cerebrovascular diseases and cancer. This article will elaborate on the biological information regarding GPR124 published in recent years and its possible related signaling pathways in the field of diseases and provide a reference for further revealing the role of GPR124 in the occurrence and development of diseases.
Collapse
Affiliation(s)
- Wan-Yun Lin
- Health Management Center, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
- School of Dentistry, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Yu-Lei Dong
- Health Management Center, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
- School of Dentistry, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Yang Lin
- School of Dentistry, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Diwas Sunchuri
- School of International Education, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Zhu-Ling Guo
- Health Management Center, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
- School of Dentistry, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| |
Collapse
|
9
|
Nogueira Pinto H, Zarekiani P, de Vries HE. Neuroglia and the blood-brain barrier. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:127-141. [PMID: 40122621 DOI: 10.1016/b978-0-443-19104-6.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The blood-brain barrier (BBB) is a highly dynamic and complex structure, present throughout the brain vasculature, that safeguards the brain against blood-borne insults. Neuroglial cells play a major role in its development, function, and homeostasis of the BBB by establishing intricate interactions via direct cell-cell contacts and paracrine signaling. Astrocytes, pericytes, oligodendrocytes, and microglia, alongside specialized brain endothelial cells, orchestrate key events in the brain in health and disease, which can be partially recapitulated by in vitro and in vivo models for biomedical research. This chapter presents a detailed description of the main cellular and molecular mechanisms that govern the neuroglia-BBB crosstalk and the available models for its investigation, emphasizing the importance of each cell population and the synergistic roles they play in the brain.
Collapse
Affiliation(s)
- Henrique Nogueira Pinto
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands
| | - Parand Zarekiani
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Helga E de Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; MS Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Chen J, Choi JJY, Lin PY, Huang EJ. Pathogenesis of Germinal Matrix Hemorrhage: Insights from Single-Cell Transcriptomics. ANNUAL REVIEW OF PATHOLOGY 2025; 20:221-243. [PMID: 39401848 PMCID: PMC11759652 DOI: 10.1146/annurev-pathmechdis-111523-023446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
The germinal matrix harbors neurogenic niches in the subpallium of the prenatal human brain that produce abundant GABAergic neurons. In preterm infants, the germinal matrix is particularly vulnerable to developing hemorrhage, which disrupts neurogenesis and causes severe neurodevelopmental sequelae. However, the disease mechanisms that promote germinal matrix hemorrhage remain unclear. Here, we review recent advances using single-cell transcriptomics to uncover novel mechanisms that govern neurogenesis and angiogenesis in the germinal matrix of the prenatal human brain. These approaches also reveal the critical role of immune-vascular interaction that promotes vascular morphogenesis in the germinal matrix and how proinflammatory factors from activated neutrophils and monocytes can disrupt this process, leading to hemorrhage. Collectively, these results reveal fundamental disease mechanisms and therapeutic interventions for germinal matrix hemorrhage.
Collapse
Affiliation(s)
- Jiapei Chen
- Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Department of Pathology, University of California, San Francisco, California, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, California, USA;
| | - Jennifer Ja-Yoon Choi
- Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Department of Pathology, University of California, San Francisco, California, USA
| | - Pin-Yeh Lin
- Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Department of Pathology, University of California, San Francisco, California, USA
| | - Eric J Huang
- Pathology Service, Veterans Administration Health Care System, San Francisco, California, USA
- Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Department of Pathology, University of California, San Francisco, California, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, California, USA;
| |
Collapse
|
11
|
Sadanandan J, Thomas S, Mathew IE, Huang Z, Blackburn SL, Tandon N, Lokhande H, McCrea PD, Bresnick EH, Dash PK, McBride DW, Harmanci A, Ahirwar LK, Jose D, Dienel AC, Zeineddine HA, Hong S, Kumar T P. Key epigenetic and signaling factors in the formation and maintenance of the blood-brain barrier. eLife 2024; 12:RP86978. [PMID: 39670988 PMCID: PMC11643625 DOI: 10.7554/elife.86978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024] Open
Abstract
The blood-brain barrier (BBB) controls the movement of molecules into and out of the central nervous system (CNS). Since a functional BBB forms by mouse embryonic day E15.5, we reasoned that gene cohorts expressed in CNS endothelial cells (EC) at E13.5 contribute to BBB formation. In contrast, adult gene signatures reflect BBB maintenance mechanisms. Supporting this hypothesis, transcriptomic analysis revealed distinct cohorts of EC genes involved in BBB formation and maintenance. Here, we demonstrate that epigenetic regulator's histone deacetylase 2 (HDAC2) and polycomb repressive complex 2 (PRC2) control EC gene expression for BBB development and prevent Wnt/β-catenin (Wnt) target genes from being expressed in adult CNS ECs. Low Wnt activity during development modifies BBB genes epigenetically for the formation of functional BBB. As a Class-I HDAC inhibitor induces adult CNS ECs to regain Wnt activity and BBB genetic signatures that support BBB formation, our results inform strategies to promote BBB repair.
Collapse
Affiliation(s)
- Jayanarayanan Sadanandan
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Sithara Thomas
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Iny Elizabeth Mathew
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Zhen Huang
- Departments of Neurology & Neuroscience, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
| | - Spiros L Blackburn
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Nitin Tandon
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | | | - Pierre D McCrea
- Department of Genetics, TheUniversity of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Emery H Bresnick
- Wisconsin Blood Cancer Research Institute, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
| | - Pramod K Dash
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Devin W McBride
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Arif Harmanci
- UTHealth School of Biomedical InformaticsHoustonUnited States
| | - Lalit K Ahirwar
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Dania Jose
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Ari C Dienel
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Hussein A Zeineddine
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Sungha Hong
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Peeyush Kumar T
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| |
Collapse
|
12
|
Kordon SP, Cechova K, Bandekar SJ, Leon K, Dutka P, Siffer G, Kossiakoff AA, Vafabakhsh R, Araç D. Conformational coupling between extracellular and transmembrane domains modulates holo-adhesion GPCR function. Nat Commun 2024; 15:10545. [PMID: 39627215 PMCID: PMC11615224 DOI: 10.1038/s41467-024-54836-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 11/20/2024] [Indexed: 12/06/2024] Open
Abstract
Adhesion G Protein-Coupled Receptors (aGPCRs) are key cell-adhesion molecules involved in numerous physiological functions. aGPCRs have large multi-domain extracellular regions (ECRs) containing a conserved GAIN domain that precedes their seven-pass transmembrane domain (7TM). Ligand binding and mechanical force applied on the ECR regulate receptor function. However, how the ECR communicates with the 7TM remains elusive, because the relative orientation and dynamics of the ECR and 7TM within a holoreceptor is unclear. Here, we describe the cryo-EM reconstruction of an aGPCR, Latrophilin3/ADGRL3, and reveal that the GAIN domain adopts a parallel orientation to the transmembrane region and has constrained movement. Single-molecule FRET experiments unveil three slow-exchanging FRET states of the ECR relative to the transmembrane region within the holoreceptor. GAIN-targeted antibodies, and cancer-associated mutations at the GAIN-7TM interface, alter FRET states, cryo-EM conformations, and receptor signaling. Altogether, this data demonstrates conformational and functional coupling between the ECR and 7TM, suggesting an ECR-mediated mechanism for aGPCR activation.
Collapse
Affiliation(s)
- Szymon P Kordon
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Center for Mechanical Excitability, University of Chicago, Chicago, IL, USA
| | - Kristina Cechova
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Sumit J Bandekar
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Center for Mechanical Excitability, University of Chicago, Chicago, IL, USA
| | - Katherine Leon
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
| | - Przemysław Dutka
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Department of Structural Biology, Genentech, South San Francisco, CA, USA
| | - Gracie Siffer
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Reza Vafabakhsh
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA.
| | - Demet Araç
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA.
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA.
- Center for Mechanical Excitability, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Huang X, Wei P, Fang C, Yu M, Yang S, Qiu L, Wang Y, Xu A, Hoo RLC, Chang J. Compromised endothelial Wnt/β-catenin signaling mediates the blood-brain barrier disruption and leads to neuroinflammation in endotoxemia. J Neuroinflammation 2024; 21:265. [PMID: 39427196 PMCID: PMC11491032 DOI: 10.1186/s12974-024-03261-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
The blood-brain barrier (BBB) is a critical interface that maintains the central nervous system homeostasis by controlling the exchange of substances between the blood and the brain. Disruption of the BBB plays a vital role in the development of neuroinflammation and neurological dysfunction in sepsis, but the mechanisms by which the BBB becomes disrupted during sepsis are not well understood. Here, we induced endotoxemia, a major type of sepsis, in mice by intraperitoneal injection of lipopolysaccharide (LPS). LPS acutely increased BBB permeability, activated microglia, and heightened inflammatory responses in brain endothelium and parenchyma. Concurrently, LPS or proinflammatory cytokines activated the NF-κB pathway, inhibiting Wnt/β-catenin signaling in brain endothelial cells in vitro and in vivo. Cell culture study revealed that NF-κB p65 directly interacted with β-catenin to suppress Wnt/β-catenin signaling. Pharmacological NF-κB pathway inhibition restored brain endothelial Wnt/β-catenin signaling activity and mitigated BBB disruption and neuroinflammation in septic mice. Furthermore, genetic or pharmacological activation of brain endothelial Wnt/β-catenin signaling substantially alleviated LPS-induced BBB leakage and neuroinflammation, while endothelial conditional ablation of the Wnt7a/7b co-receptor Gpr124 exacerbated the BBB leakage caused by LPS. Mechanistically, Wnt/β-catenin signaling activation rectified the reduced expression levels of tight junction protein ZO-1 and transcytosis suppressor Mfsd2a in brain endothelial cells of mice with endotoxemia, inhibiting both paracellular and transcellular permeability of the BBB. Our findings demonstrate that endotoxemia-associated systemic inflammation decreases endothelial Wnt/β-catenin signaling through activating NF-κB pathway, resulting in acute BBB disruption and neuroinflammation. Targeting the endothelial Wnt/β-catenin signaling may offer a promising therapeutic strategy for preserving BBB integrity and treating neurological dysfunction in sepsis.
Collapse
Affiliation(s)
- Xiaowen Huang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Avenue 1068, Nanshan, Shenzhen, Guangdong, 518055, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- State Key Laboratory of Pharmacological Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Pengju Wei
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Avenue 1068, Nanshan, Shenzhen, Guangdong, 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Cheng Fang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Avenue 1068, Nanshan, Shenzhen, Guangdong, 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Min Yu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Avenue 1068, Nanshan, Shenzhen, Guangdong, 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Shilun Yang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Avenue 1068, Nanshan, Shenzhen, Guangdong, 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Linhui Qiu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Avenue 1068, Nanshan, Shenzhen, Guangdong, 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Yu Wang
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- State Key Laboratory of Pharmacological Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Aimin Xu
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- State Key Laboratory of Pharmacological Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ruby Lai Chong Hoo
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- State Key Laboratory of Pharmacological Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Junlei Chang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Xueyuan Avenue 1068, Nanshan, Shenzhen, Guangdong, 518055, China.
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
14
|
Zhang Y, Shen X, Deng S, Chen Q, Xu B. Neural Regulation of Vascular Development: Molecular Mechanisms and Interactions. Biomolecules 2024; 14:966. [PMID: 39199354 PMCID: PMC11353022 DOI: 10.3390/biom14080966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
As a critical part of the circulatory system, blood vessels transport oxygen and nutrients to every corner of the body, nourishing each cell, and also remove waste and toxins. Defects in vascular development and function are closely associated with many diseases, such as heart disease, stroke, and atherosclerosis. In the nervous system, the nervous and vascular systems are intricately connected in both development and function. First, peripheral blood vessels and nerves exhibit parallel distribution patterns. In the central nervous system (CNS), nerves and blood vessels form a complex interface known as the neurovascular unit. Second, the vascular system employs similar cellular and molecular mechanisms as the nervous system for its development. Third, the development and function of CNS vasculature are tightly regulated by CNS-specific signaling pathways and neural activity. Additionally, vascular endothelial cells within the CNS are tightly connected and interact with pericytes, astrocytes, neurons, and microglia to form the blood-brain barrier (BBB). The BBB strictly controls material exchanges between the blood and brain, maintaining the brain's microenvironmental homeostasis, which is crucial for the normal development and function of the CNS. Here, we comprehensively summarize research on neural regulation of vascular and BBB development and propose directions for future research.
Collapse
Affiliation(s)
- Yu Zhang
- School of Life Sciences, Nantong University, Nantong 226019, China
| | - Xinyu Shen
- School of Life Sciences, Nantong University, Nantong 226019, China
| | - Shunze Deng
- School of Life Sciences, Nantong University, Nantong 226019, China
| | - Qiurong Chen
- School of Life Sciences, Nantong University, Nantong 226019, China
| | - Bing Xu
- School of Life Sciences, Nantong University, Nantong 226019, China
| |
Collapse
|
15
|
Wälchli T, Ghobrial M, Schwab M, Takada S, Zhong H, Suntharalingham S, Vetiska S, Gonzalez DR, Wu R, Rehrauer H, Dinesh A, Yu K, Chen ELY, Bisschop J, Farnhammer F, Mansur A, Kalucka J, Tirosh I, Regli L, Schaller K, Frei K, Ketela T, Bernstein M, Kongkham P, Carmeliet P, Valiante T, Dirks PB, Suva ML, Zadeh G, Tabar V, Schlapbach R, Jackson HW, De Bock K, Fish JE, Monnier PP, Bader GD, Radovanovic I. Single-cell atlas of the human brain vasculature across development, adulthood and disease. Nature 2024; 632:603-613. [PMID: 38987604 PMCID: PMC11324530 DOI: 10.1038/s41586-024-07493-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 04/30/2024] [Indexed: 07/12/2024]
Abstract
A broad range of brain pathologies critically relies on the vasculature, and cerebrovascular disease is a leading cause of death worldwide. However, the cellular and molecular architecture of the human brain vasculature remains incompletely understood1. Here we performed single-cell RNA sequencing analysis of 606,380 freshly isolated endothelial cells, perivascular cells and other tissue-derived cells from 117 samples, from 68 human fetuses and adult patients to construct a molecular atlas of the developing fetal, adult control and diseased human brain vasculature. We identify extensive molecular heterogeneity of the vasculature of healthy fetal and adult human brains and across five vascular-dependent central nervous system (CNS) pathologies, including brain tumours and brain vascular malformations. We identify alteration of arteriovenous differentiation and reactivated fetal as well as conserved dysregulated genes and pathways in the diseased vasculature. Pathological endothelial cells display a loss of CNS-specific properties and reveal an upregulation of MHC class II molecules, indicating atypical features of CNS endothelial cells. Cell-cell interaction analyses predict substantial endothelial-to-perivascular cell ligand-receptor cross-talk, including immune-related and angiogenic pathways, thereby revealing a central role for the endothelium within brain neurovascular unit signalling networks. Our single-cell brain atlas provides insights into the molecular architecture and heterogeneity of the developing, adult/control and diseased human brain vasculature and serves as a powerful reference for future studies.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada.
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich and University Hospital Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
| | - Moheb Ghobrial
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Laboratory of Exercise and Health, Institute of Exercise and Health, Department of Health Sciences and Technology; Swiss Federal Institute of Technology (ETH Zurich), Zurich, Switzerland
| | - Marc Schwab
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Shigeki Takada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hang Zhong
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Laboratory of Exercise and Health, Institute of Exercise and Health, Department of Health Sciences and Technology; Swiss Federal Institute of Technology (ETH Zurich), Zurich, Switzerland
| | - Samuel Suntharalingham
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Vetiska
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | | - Ruilin Wu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Anuroopa Dinesh
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Ontario, Canada
| | - Kai Yu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Edward L Y Chen
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jeroen Bisschop
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Fiona Farnhammer
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ann Mansur
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Luca Regli
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Karl Schaller
- Department of Neurosurgery, University of Geneva Medical Center & Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Karl Frei
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, University of Zurich and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Troy Ketela
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Mark Bernstein
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Sprott Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Paul Kongkham
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Sprott Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters-Hamilton Centre for Neuro-Oncology Research, University Health Network, Toronto, Ontario, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB & Department of Oncology, KU Leuven, Leuven, Belgium
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, P. R. China
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Taufik Valiante
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Sprott Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Krembil Brain Institute, Division of Clinical and Computational Neuroscience, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomaterials and Biomedical Engineering and Electrical and Computer Engineering, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter B Dirks
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Arthur and Sonia Labatt Brain Tumor Research Center, Departments of Surgery and Molecular Genetics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mario L Suva
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Sprott Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Viviane Tabar
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ralph Schlapbach
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Hartland W Jackson
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Ontario Institute of Cancer Research, Toronto, Ontario, Canada
| | - Katrien De Bock
- Laboratory of Exercise and Health, Institute of Exercise and Health, Department of Health Sciences and Technology; Swiss Federal Institute of Technology (ETH Zurich), Zurich, Switzerland
| | - Jason E Fish
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Research Institute, Vision Division, Krembil Discovery Tower, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gary D Bader
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Ivan Radovanovic
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Sprott Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Post Y, Lu C, Fletcher RB, Yeh WC, Nguyen H, Lee SJ, Li Y. Design principles and therapeutic applications of novel synthetic WNT signaling agonists. iScience 2024; 27:109938. [PMID: 38832011 PMCID: PMC11145361 DOI: 10.1016/j.isci.2024.109938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Wingless-related integration site or Wingless and Int-1 or Wingless-Int (WNT) signaling is crucial for embryonic development, and adult tissue homeostasis and regeneration, through its essential roles in cell fate, patterning, and stem cell regulation. The biophysical characteristics of WNT ligands have hindered efforts to interrogate ligand activity in vivo and prevented their development as therapeutics. Recent breakthroughs have enabled the generation of synthetic WNT signaling molecules that possess characteristics of natural ligands and potently activate the pathway, while also providing distinct advantages for therapeutic development and manufacturing. This review provides a detailed discussion of the protein engineering of these molecular platforms for WNT signaling agonism. We discuss the importance of WNT signaling in several organs and share insights from the initial application of these new classes of molecules in vitro and in vivo. These molecules offer a unique opportunity to enhance our understanding of how WNT signaling agonism promotes tissue repair, enabling targeted development of tailored therapeutics.
Collapse
Affiliation(s)
- Yorick Post
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Chenggang Lu
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Russell B. Fletcher
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Wen-Chen Yeh
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Huy Nguyen
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Sung-Jin Lee
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Yang Li
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| |
Collapse
|
17
|
Dao L, You Z, Lu L, Xu T, Sarkar AK, Zhu H, Liu M, Calandrelli R, Yoshida G, Lin P, Miao Y, Mierke S, Kalva S, Zhu H, Gu M, Vadivelu S, Zhong S, Huang LF, Guo Z. Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids. Cell Stem Cell 2024; 31:818-833.e11. [PMID: 38754427 PMCID: PMC11162335 DOI: 10.1016/j.stem.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 02/24/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
The human blood-brain barrier (hBBB) is a highly specialized structure that regulates passage across blood and central nervous system (CNS) compartments. Despite its critical physiological role, there are no reliable in vitro models that can mimic hBBB development and function. Here, we constructed hBBB assembloids from brain and blood vessel organoids derived from human pluripotent stem cells. We validated the acquisition of blood-brain barrier (BBB)-specific molecular, cellular, transcriptomic, and functional characteristics and uncovered an extensive neuro-vascular crosstalk with a spatial pattern within hBBB assembloids. When we used patient-derived hBBB assembloids to model cerebral cavernous malformations (CCMs), we found that these assembloids recapitulated the cavernoma anatomy and BBB breakdown observed in patients. Upon comparison of phenotypes and transcriptome between patient-derived hBBB assembloids and primary human cavernoma tissues, we uncovered CCM-related molecular and cellular alterations. Taken together, we report hBBB assembloids that mimic the core properties of the hBBB and identify a potentially underlying cause of CCMs.
Collapse
Affiliation(s)
- Lan Dao
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zhen You
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lu Lu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tianyang Xu
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Avijite Kumer Sarkar
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hui Zhu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Miao Liu
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Riccardo Calandrelli
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Yoshida
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pei Lin
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yifei Miao
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sarah Mierke
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Srijan Kalva
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Haining Zhu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sudhakar Vadivelu
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Sheng Zhong
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | - L Frank Huang
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
18
|
Yuki K, Vallon M, Ding J, Rada CC, Tang AT, Vilches-Moure JG, McCormick AK, Henao Echeverri MF, Alwahabi S, Braunger BM, Ergün S, Kahn ML, Kuo CJ. GPR124 regulates murine brain embryonic angiogenesis and BBB formation by an intracellular domain-independent mechanism. Development 2024; 151:dev202794. [PMID: 38682276 PMCID: PMC11213517 DOI: 10.1242/dev.202794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
The GPR124/RECK/WNT7 pathway is an essential regulator of CNS angiogenesis and blood-brain barrier (BBB) function. GPR124, a brain endothelial adhesion seven-pass transmembrane protein, associates with RECK, which binds and stabilizes newly synthesized WNT7 that is transferred to frizzled (FZD) to initiate canonical β-catenin signaling. GPR124 remains enigmatic: although its extracellular domain (ECD) is essential, the poorly conserved intracellular domain (ICD) appears to be variably required in mammals versus zebrafish, potentially via adaptor protein bridging of GPR124 and FZD ICDs. GPR124 ICD deletion impairs zebrafish angiogenesis, but paradoxically retains WNT7 signaling upon mammalian transfection. We thus investigated GPR124 ICD function using the mouse deletion mutant Gpr124ΔC. Despite inefficiently expressed GPR124ΔC protein, Gpr124ΔC/ΔC mice could be born with normal cerebral cortex angiogenesis, in comparison with Gpr124-/- embryonic lethality, forebrain avascularity and hemorrhage. Gpr124ΔC/ΔC vascular phenotypes were restricted to sporadic ganglionic eminence angiogenic defects, attributable to impaired GPR124ΔC protein expression. Furthermore, Gpr124ΔC and the recombinant GPR124 ECD rescued WNT7 signaling in culture upon brain endothelial Gpr124 knockdown. Thus, in mice, GPR124-regulated CNS forebrain angiogenesis and BBB function are exerted by ICD-independent functionality, extending the signaling mechanisms used by adhesion seven-pass transmembrane receptors.
Collapse
Affiliation(s)
- Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mario Vallon
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Wuerzburg, 97070 Wuerzburg, Germany
| | - Jie Ding
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cara C. Rada
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan T. Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - José G. Vilches-Moure
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aaron K. McCormick
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maria F. Henao Echeverri
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Samira Alwahabi
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Barbara M. Braunger
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Wuerzburg, 97070 Wuerzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Wuerzburg, 97070 Wuerzburg, Germany
| | - Mark L. Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Calvin J. Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
19
|
Fetsko AR, Sebo DJ, Budzynski LB, Scharbarth A, Taylor MR. IL-1β disrupts the initiation of blood-brain barrier development by inhibiting endothelial Wnt/β-catenin signaling. iScience 2024; 27:109651. [PMID: 38638574 PMCID: PMC11025013 DOI: 10.1016/j.isci.2024.109651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/06/2024] [Accepted: 03/29/2024] [Indexed: 04/20/2024] Open
Abstract
During neuroinflammation, the proinflammatory cytokine interleukin-1β (IL-1β) impacts blood-brain barrier (BBB) function by disrupting brain endothelial tight junctions, promoting vascular permeability, and increasing transmigration of immune cells. Here, we examined the effects of Il-1β on the in vivo initiation of BBB development. We generated doxycycline-inducible transgenic zebrafish to secrete Il-1β in the CNS. To validate the utility of our model, we showed Il-1β dose-dependent mortality, recruitment of neutrophils, and expansion of microglia. Using live imaging, we discovered that Il-1β causes a significant reduction in CNS angiogenesis and barriergenesis. To demonstrate specificity, we rescued the Il-1β induced phenotypes by targeting the zebrafish il1r1 gene using CRISPR-Cas9. Mechanistically, we determined that Il-1β disrupts the initiation of BBB development by decreasing Wnt/β-catenin transcriptional activation in brain endothelial cells. Given that several neurodevelopmental disorders are associated with inflammation, our findings support further investigation into the connections between proinflammatory cytokines, neuroinflammation, and neurovascular development.
Collapse
Affiliation(s)
- Audrey R. Fetsko
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dylan J. Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Lilyana B. Budzynski
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Alli Scharbarth
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michael R. Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
20
|
Schevenels G, Cabochette P, America M, Vandenborne A, De Grande L, Guenther S, He L, Dieu M, Christou B, Vermeersch M, Germano RFV, Perez-Morga D, Renard P, Martin M, Vanlandewijck M, Betsholtz C, Vanhollebeke B. A brain-specific angiogenic mechanism enabled by tip cell specialization. Nature 2024; 628:863-871. [PMID: 38570687 PMCID: PMC11041701 DOI: 10.1038/s41586-024-07283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Vertebrate organs require locally adapted blood vessels1,2. The gain of such organotypic vessel specializations is often deemed to be molecularly unrelated to the process of organ vascularization. Here, opposing this model, we reveal a molecular mechanism for brain-specific angiogenesis that operates under the control of Wnt7a/b ligands-well-known blood-brain barrier maturation signals3-5. The control mechanism relies on Wnt7a/b-dependent expression of Mmp25, which we find is enriched in brain endothelial cells. CRISPR-Cas9 mutagenesis in zebrafish reveals that this poorly characterized glycosylphosphatidylinositol-anchored matrix metalloproteinase is selectively required in endothelial tip cells to enable their initial migration across the pial basement membrane lining the brain surface. Mechanistically, Mmp25 confers brain invasive competence by cleaving meningeal fibroblast-derived collagen IV α5/6 chains within a short non-collagenous region of the central helical part of the heterotrimer. After genetic interference with the pial basement membrane composition, the Wnt-β-catenin-dependent organotypic control of brain angiogenesis is lost, resulting in properly patterned, yet blood-brain-barrier-defective cerebrovasculatures. We reveal an organ-specific angiogenesis mechanism, shed light on tip cell mechanistic angiodiversity and thereby illustrate how organs, by imposing local constraints on angiogenic tip cells, can select vessels matching their distinctive physiological requirements.
Collapse
Affiliation(s)
- Giel Schevenels
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Pauline Cabochette
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Michelle America
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Arnaud Vandenborne
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Line De Grande
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Stefan Guenther
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, Germany
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Marc Dieu
- Mass Spectrometry Facility (MaSUN), University of Namur, Namur, Belgium
| | - Basile Christou
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Raoul F V Germano
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - David Perez-Morga
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Patricia Renard
- Mass Spectrometry Facility (MaSUN), University of Namur, Namur, Belgium
| | - Maud Martin
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium.
| |
Collapse
|
21
|
Kordon SP, Cechova K, Bandekar SJ, Leon K, Dutka P, Siffer G, Kossiakoff AA, Vafabakhsh R, Araç D. Structural analysis and conformational dynamics of a holo-adhesion GPCR reveal interplay between extracellular and transmembrane domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.25.581807. [PMID: 38464178 PMCID: PMC10925191 DOI: 10.1101/2024.02.25.581807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Adhesion G Protein-Coupled Receptors (aGPCRs) are key cell-adhesion molecules involved in numerous physiological functions. aGPCRs have large multi-domain extracellular regions (ECR) containing a conserved GAIN domain that precedes their seven-pass transmembrane domain (7TM). Ligand binding and mechanical force applied on the ECR regulate receptor function. However, how the ECR communicates with the 7TM remains elusive, because the relative orientation and dynamics of the ECR and 7TM within a holoreceptor is unclear. Here, we describe the cryo-EM reconstruction of an aGPCR, Latrophilin3/ADGRL3, and reveal that the GAIN domain adopts a parallel orientation to the membrane and has constrained movement. Single-molecule FRET experiments unveil three slow-exchanging FRET states of the ECR relative to the 7TM within the holoreceptor. GAIN-targeted antibodies, and cancer-associated mutations at the GAIN-7TM interface, alter FRET states, cryo-EM conformations, and receptor signaling. Altogether, this data demonstrates conformational and functional coupling between the ECR and 7TM, suggesting an ECR-mediated mechanism of aGPCR activation.
Collapse
|
22
|
Xu Y, Fang X, Zhao Z, Wu H, Fan H, Zhang Y, Meng Q, Rong Q, Fukunaga K, Guo Q, Liu Q. GPR124 induces NLRP3 inflammasome-mediated pyroptosis in endothelial cells during ischemic injury. Eur J Pharmacol 2024; 962:176228. [PMID: 38042462 DOI: 10.1016/j.ejphar.2023.176228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
OBJECTIVE G protein-coupled receptor 124 (GPR124) regulates central nervous system angiogenesis and blood-brain barrier (BBB) integrity, and its deficiency aggravates BBB breakdown and hemorrhagic transformation in ischemic mice. However, excessive GPR124 expression promotes inflammation in atherosclerotic mice. In this study, we aimed to elucidate the role of GPR124 in hypoxia/ischemia-induced cerebrovascular endothelial cell injury. METHODS bEnd.3 cells were exposed to oxygen-glucose deprivation (OGD), and time-dependent changes in GPR124 mRNA and protein expression were evaluated using reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting. The effects of GPR124 overexpression or knockdown on the expression of pyroptosis-related genes were assessed at the mRNA and protein levels. Tadehaginoside (TA) was screened as a potential small molecule targeting GPR124, and its effects on pyroptosis-related signaling pathways were investigated. Finally, the therapeutic efficacy of TA was evaluated using a rat model of transient middle cerebral artery occlusion/reperfusion (tMCAO/R). RESULTS During OGD, the expression of GPR124 initially increased and then decreased over time, with the highest levels observed 1 h after OGD. The overexpression of GPR124 enhanced the OGD-induced expression of NLRP3, Caspase-1, and Gasdermin D (GSDMD) in bEnd.3 cells, whereas GPR124 knockdown reduced pyroptosis. Additionally, TA exhibited a high targeting ability to GPR124, significantly inhibiting its function and expression and suppressing the expression of pyroptosis-related proteins during OGD. Furthermore, TA treatment significantly reduced the cerebral infarct volume and pyroptotic signaling in tMCAO/R rats. CONCLUSIONS Our findings suggest that GPR124 mediates pyroptotic signaling in endothelial cells during the early stages of hypoxia/ischemia, thereby exacerbating ischemic injury.
Collapse
Affiliation(s)
- Yiqian Xu
- Department of Pharmacy & Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Xingyue Fang
- Department of Pharmacy & Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Zhenqiang Zhao
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Haolin Wu
- Department of Pharmacy & Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Haofei Fan
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Ya Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou 571199, China
| | - Qingwen Meng
- Department of Pharmacology, School of Basic and Life Science, Hainan Medical University, Haikou 571199, China
| | - Qiongwen Rong
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Qingyun Guo
- Department of Pharmacology, School of Basic and Life Science, Hainan Medical University, Haikou 571199, China; Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, China.
| | - Qibing Liu
- Department of Pharmacy & Engineering Research Center of Tropical Medicine Innovation and Transformation, Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China; Department of Pharmacology, School of Basic and Life Science, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
23
|
Yu M, Nie Y, Yang J, Yang S, Li R, Rao V, Hu X, Fang C, Li S, Song D, Guo F, Snyder MP, Chang HY, Kuo CJ, Xu J, Chang J. Integrative multi-omic profiling of adult mouse brain endothelial cells and potential implications in Alzheimer's disease. Cell Rep 2023; 42:113392. [PMID: 37925638 PMCID: PMC10843806 DOI: 10.1016/j.celrep.2023.113392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 09/11/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023] Open
Abstract
The blood-brain barrier (BBB) is primarily manifested by a variety of physiological properties of brain endothelial cells (ECs), but the molecular foundation for these properties remains incompletely clear. Here, we generate a comprehensive molecular atlas of adult brain ECs using acutely purified mouse ECs and integrated multi-omics. Using RNA sequencing (RNA-seq) and proteomics, we identify the transcripts and proteins selectively enriched in brain ECs and demonstrate that they are partially correlated. Using single-cell RNA-seq, we dissect the molecular basis of functional heterogeneity of brain ECs. Using integrative epigenomics and transcriptomics, we determine that TCF/LEF, SOX, and ETS families are top-ranked transcription factors regulating the BBB. We then validate the identified brain-EC-enriched proteins and transcription factors in normal mouse and human brain tissue and assess their expression changes in mice with Alzheimer's disease. Overall, we present a valuable resource with broad implications for regulation of the BBB and treatment of neurological disorders.
Collapse
Affiliation(s)
- Min Yu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yage Nie
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jiawen Yang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Shilun Yang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Varsha Rao
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoyan Hu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cheng Fang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Simeng Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dengpan Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Fuyou Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Jin Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, China.
| | - Junlei Chang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
24
|
Salahudeen AA, Seoane JA, Yuki K, Mah AT, Smith AR, Kolahi K, De la O SM, Hart DJ, Ding J, Ma Z, Barkal SA, Shukla ND, Zhang CH, Cantrell MA, Batish A, Usui T, Root DE, Hahn WC, Curtis C, Kuo CJ. Functional screening of amplification outlier oncogenes in organoid models of early tumorigenesis. Cell Rep 2023; 42:113355. [PMID: 37922313 PMCID: PMC10841581 DOI: 10.1016/j.celrep.2023.113355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/30/2023] [Accepted: 10/12/2023] [Indexed: 11/05/2023] Open
Abstract
Somatic copy number gains are pervasive across cancer types, yet their roles in oncogenesis are insufficiently evaluated. This inadequacy is partly due to copy gains spanning large chromosomal regions, obscuring causal loci. Here, we employed organoid modeling to evaluate candidate oncogenic loci identified via integrative computational analysis of extreme copy gains overlapping with extreme expression dysregulation in The Cancer Genome Atlas. Subsets of "outlier" candidates were contextually screened as tissue-specific cDNA lentiviral libraries within cognate esophagus, oral cavity, colon, stomach, pancreas, and lung organoids bearing initial oncogenic mutations. Iterative analysis nominated the kinase DYRK2 at 12q15 as an amplified head and neck squamous carcinoma oncogene in p53-/- oral mucosal organoids. Similarly, FGF3, amplified at 11q13 in 41% of esophageal squamous carcinomas, promoted p53-/- esophageal organoid growth reversible by small molecule and soluble receptor antagonism of FGFRs. Our studies establish organoid-based contextual screening of candidate genomic drivers, enabling functional evaluation during early tumorigenesis.
Collapse
Affiliation(s)
- Ameen A Salahudeen
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA; University of Illinois at Chicago College of Medicine, Department of Medicine, Division of Hematology and Oncology, Chicago, IL 60612, USA; Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA; University of Illinois Cancer Center, Chicago, IL 60612, USA.
| | - Jose A Seoane
- Stanford University School of Medicine, Department of Medicine, Divisions of Oncology, Stanford, CA 94305, USA; Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain.
| | - Kanako Yuki
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Amanda T Mah
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Amber R Smith
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Kevin Kolahi
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Sean M De la O
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Daniel J Hart
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Jie Ding
- Stanford University School of Medicine, Department of Medicine, Divisions of Oncology, Stanford, CA 94305, USA
| | - Zhicheng Ma
- Stanford University School of Medicine, Department of Medicine, Divisions of Oncology, Stanford, CA 94305, USA
| | - Sammy A Barkal
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Navika D Shukla
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Chuck H Zhang
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Michael A Cantrell
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Arpit Batish
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - Tatsuya Usui
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA
| | - David E Root
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - William C Hahn
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA; Dana-Farber Cancer Institute, Department of Medical Oncology, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Christina Curtis
- Stanford University School of Medicine, Department of Medicine, Divisions of Oncology, Stanford, CA 94305, USA; Stanford University School of Medicine, Department of Medicine, Divisions of Genetics, Stanford, CA 94305, USA
| | - Calvin J Kuo
- Stanford University School of Medicine, Department of Medicine, Divisions of Hematology, Stanford, CA 94305, USA.
| |
Collapse
|
25
|
Lin HH. Functional partnerships between GPI-anchored proteins and adhesion GPCRs. Bioessays 2023; 45:e2300115. [PMID: 37526334 DOI: 10.1002/bies.202300115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023]
Abstract
Specific extracellular interaction between glycophosphatidylinositol (GPI)-anchored proteins and adhesion G protein-coupled receptors (aGPCRs) plays an important role in unique biological functions. GPI-anchored proteins are derived from a novel post-translational modification of single-span membrane molecules, while aGPCRs are bona fide seven-span transmembrane proteins with a long extracellular domain. Although various members of the two structurally-distinct protein families are known to be involved in a wide range of biological processes, many remain as orphans. Interestingly, accumulating evidence has pointed to a complex interaction and functional synergy between these two protein families. I discuss herein current understanding of specific functional partnerships between GPI-anchored proteins and aGPCRs.
Collapse
Affiliation(s)
- Hsi-Hsien Lin
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate School of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
- Division of Rheumatology, Allergy, and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung, Taiwan
| |
Collapse
|
26
|
Rao S, Liu M, Iosef C, Knutsen C, Alvira CM. Endothelial-specific loss of IKKβ disrupts pulmonary endothelial angiogenesis and impairs postnatal lung growth. Am J Physiol Lung Cell Mol Physiol 2023; 325:L299-L313. [PMID: 37310763 PMCID: PMC10625829 DOI: 10.1152/ajplung.00034.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023] Open
Abstract
Pulmonary angiogenesis drives alveolarization, but the transcriptional regulators directing pulmonary angiogenesis remain poorly defined. Global, pharmacological inhibition of nuclear factor-kappa B (NF-κB) impairs pulmonary angiogenesis and alveolarization. However, establishing a definitive role for NF-κB in pulmonary vascular development has been hindered by embryonic lethality induced by constitutive deletion of NF-κB family members. We created a mouse model allowing inducible deletion of the NF-κB activator, IKKβ, in endothelial cells (ECs) and assessed the effect on lung structure, endothelial angiogenic function, and the lung transcriptome. Embryonic deletion of IKKβ permitted lung vascular development but resulted in a disorganized vascular plexus, while postnatal deletion significantly decreased radial alveolar counts, vascular density, and proliferation of both endothelial and nonendothelial lung cells. Loss of IKKβ impaired survival, proliferation, migration, and angiogenesis in primary lung ECs in vitro, in association with decreased expression of VEGFR2 and activation of downstream effectors. Loss of endothelial IKKβ in vivo induced broad changes in the lung transcriptome with downregulation of genes related to mitotic cell cycle, extracellular matrix (ECM)-receptor interaction, and vascular development, and the upregulation of genes related to inflammation. Computational deconvolution suggested that loss of endothelial IKKβ decreased general capillary, aerocyte capillary, and alveolar type I cell abundance. Taken together, these data definitively establish an essential role for endogenous endothelial IKKβ signaling during alveolarization. A deeper understanding of the mechanisms directing this developmental, physiological activation of IKKβ in the lung vasculature may provide novel targets for the development of strategies to enhance beneficial proangiogenic signaling in lung development and disease.NEW & NOTEWORTHY This study highlights the cell-specific complexity of nuclear factor kappa B signaling in the developing lung by demonstrating that inducible loss of IKKβ in endothelial cells impairs alveolarization, disrupts EC angiogenic function, and broadly represses genes important for vascular development.
Collapse
Affiliation(s)
- Shailaja Rao
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Min Liu
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristiana Iosef
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Carsten Knutsen
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| |
Collapse
|
27
|
Shu J, Wang C, Tao Y, Wang S, Cheng F, Zhang Y, Shi K, Xia K, Wang R, Wang J, Yu C, Chen J, Huang X, Xu H, Zhou X, Wu H, Liang C, Chen Q, Yan S, Li F. Thermosensitive hydrogel-based GPR124 delivery strategy for rebuilding blood-spinal cord barrier. Bioeng Transl Med 2023; 8:e10561. [PMID: 37693060 PMCID: PMC10486335 DOI: 10.1002/btm2.10561] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/20/2023] [Accepted: 05/25/2023] [Indexed: 09/12/2023] Open
Abstract
Spinal cord injury (SCI) causes blood-spinal cord barrier (BSCB) disruption, leading to secondary damage, such as hemorrhagic infiltration, inflammatory response, and neuronal cell death. It is of great significance to rebuild the BSCB at the early stage of SCI to alleviate the secondary injury for better prognosis. Yet, current research involved in the reconstruction of BSCB is insufficient. Accordingly, we provide a thermosensitive hydrogel-based G protein-coupled receptor 124 (GPR124) delivery strategy for rebuilding BSCB. Herein, we firstly found that the expression of GPR124 decreased post-SCI and demonstrated that treatment with recombinant GPR124 could partially alleviate the disruption of BSCB post-SCI by restoring tight junctions (TJs) and promoting migration and tube formation of endothelial cells. Interestingly, GPR124 could also boost the energy metabolism of endothelial cells. However, the absence of physicochemical stability restricted the wide usage of GPR124. Hence, we fabricated a thermosensitive heparin-poloxamer (HP) hydrogel that demonstrated sustained GPR124 production and maintained the bioactivity of GPR124 (HP@124) for rebuilding the BSCB and eventually enhancing functional motor recovery post-SCI. HP@124 hydrogel can encapsulate GPR124 at the lesion site by injection, providing prolonged release, preserving wounded tissues, and filling injured tissue cavities. Consequently, it induces synergistically efficient integrated regulation by blocking BSCB rupture, decreasing fibrotic scar formation, minimizing inflammatory response, boosting remyelination, and regenerating axons. Mechanistically, giving GPR124 activates energy metabolism via elevating the expression of phosphoenolpyruvate carboxykinase 2 (PCK2), and eventually restores the poor state of endothelial cells. This research demonstrated that early intervention by combining GPR124 with bioactive multifunctional hydrogel may have tremendous promise for restoring locomotor recovery in patients with central nervous system disorders, in addition to a translational approach for the medical therapy of SCI.
Collapse
Affiliation(s)
- Jiawei Shu
- International Institutes of MedicineThe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwuZhejiangPeople's Republic of China
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Chenggui Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangPeople's Republic of China
| | - Yiqing Tao
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Shaoke Wang
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Feng Cheng
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yuang Zhang
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Kesi Shi
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Kaishun Xia
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Ronghao Wang
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Jingkai Wang
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Chao Yu
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Jiangjie Chen
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Xianpeng Huang
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Haibin Xu
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Xiaopeng Zhou
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Haobo Wu
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Chengzhen Liang
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Qixin Chen
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Shigui Yan
- International Institutes of MedicineThe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwuZhejiangPeople's Republic of China
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Fangcai Li
- Department of Orthopedics SurgeryThe Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Orthopedics Research Institute of Zhejiang University, Zhejiang UniversityHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| |
Collapse
|
28
|
Xie Y, He L, Zhang Y, Huang H, Yang F, Chao M, Cao H, Wang J, Li Y, Zhang L, Xin L, Xiao B, Shi X, Zhang X, Tang J, Uhrbom L, Dimberg A, Wang L, Zhang L. Wnt signaling regulates MFSD2A-dependent drug delivery through endothelial transcytosis in glioma. Neuro Oncol 2023; 25:1073-1084. [PMID: 36591963 PMCID: PMC10237416 DOI: 10.1093/neuonc/noac288] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Systemic delivery of anti-tumor therapeutic agents to brain tumors is thwarted by the blood-brain barrier (BBB), an organotypic specialization of brain endothelial cells (ECs). A failure of pharmacological compounds to cross BBB is one culprit for the dismal prognosis of glioblastoma (GBM) patients. Identification of novel vascular targets to overcome the challenges posed by the BBB in tumors for GBM treatment is urgently needed. METHODS Temozolomide (TMZ) delivery was investigated in CT2A and PDGFB-driven RCAS/tv-a orthotopic glioma models. Transcriptome analysis was performed on ECs from murine gliomas. Mfsd2a deficient, Cav1 deficient, and Mfsd2a EC-specific inducible mice were developed to study the underlying molecular mechanisms. RESULTS We demonstrated that inhibiting Wnt signaling by LGK974 could increase TMZ delivery and sensitize glioma to chemotherapy in both murine glioma models. Transcriptome analysis of ECs from murine gliomas revealed that Wnt signaling inhibition enhanced vascular transcytosis as indicated by the upregulation of PLVAP and downregulation of MFSD2A. Mfsd2a deficiency in mice enhances TMZ delivery in tumors, whereas constitutive expression of Mfsd2a in ECs suppresses the enhanced TMZ delivery induced by Wnt pathway inhibition in murine glioma. In addition, Wnt signaling inhibition enhanced caveolin-1 (Cav1)-positive caveolae-mediated transcytosis in tumor ECs. Moreover, Wnt signaling inhibitor or Mfsd2a deficiency fails to enhance TMZ penetration in tumors from Cav1-deficient mice. CONCLUSIONS These results demonstrated that Wnt signaling regulates MFSD2A-dependent TMZ delivery through a caveolae-mediated EC transcytosis pathway. Our findings identify Wnt signaling as a promising therapeutic target to improve drug delivery for GBM treatment.
Collapse
Affiliation(s)
- Yuan Xie
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, 710119, China
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, 710032, China
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Fan Yang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuro-injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Min Chao
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi’an, 710038, China
| | - Haiyan Cao
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi’an, 710038, China
| | - Jianhao Wang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuro-injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Yaling Li
- Department of Obstetrics and Gynaecology, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, 710005, China
| | - Lingxue Zhang
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, 710119, China
| | - Lele Xin
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, 710119, China
| | - Bing Xiao
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, 710119, China
| | - Xinxin Shi
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, 710119, China
| | - Xue Zhang
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, 710119, China
| | - Jiefu Tang
- Trauma Center, First Affiliated Hospital of Hunan University of Medicine, Huaihua, 418000, China
| | - Lene Uhrbom
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, 75185, Uppsala, Sweden
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, 569 Xinsi Road, Xi’an, 710038, China
| | - Lei Zhang
- China-Sweden International Joint Research Center for Brain Diseases, Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, 710119, China
| |
Collapse
|
29
|
Parab S, Card OA, Chen Q, America M, Buck LD, Quick RE, Horrigan WF, Levkowitz G, Vanhollebeke B, Matsuoka RL. Local angiogenic interplay of Vegfc/d and Vegfa controls brain region-specific emergence of fenestrated capillaries. eLife 2023; 12:e86066. [PMID: 37191285 PMCID: PMC10229134 DOI: 10.7554/elife.86066] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/15/2023] [Indexed: 05/17/2023] Open
Abstract
Fenestrated and blood-brain barrier (BBB)-forming endothelial cells constitute major brain capillaries, and this vascular heterogeneity is crucial for region-specific neural function and brain homeostasis. How these capillary types emerge in a brain region-specific manner and subsequently establish intra-brain vascular heterogeneity remains unclear. Here, we performed a comparative analysis of vascularization across the zebrafish choroid plexuses (CPs), circumventricular organs (CVOs), and retinal choroid, and show common angiogenic mechanisms critical for fenestrated brain capillary formation. We found that zebrafish deficient for Gpr124, Reck, or Wnt7aa exhibit severely impaired BBB angiogenesis without any apparent defect in fenestrated capillary formation in the CPs, CVOs, and retinal choroid. Conversely, genetic loss of various Vegf combinations caused significant disruptions in Wnt7/Gpr124/Reck signaling-independent vascularization of these organs. The phenotypic variation and specificity revealed heterogeneous endothelial requirements for Vegfs-dependent angiogenesis during CP and CVO vascularization, identifying unexpected interplay of Vegfc/d and Vegfa in this process. Mechanistically, expression analysis and paracrine activity-deficient vegfc mutant characterization suggest that endothelial cells and non-neuronal specialized cell types present in the CPs and CVOs are major sources of Vegfs responsible for regionally restricted angiogenic interplay. Thus, brain region-specific presentations and interplay of Vegfc/d and Vegfa control emergence of fenestrated capillaries, providing insight into the mechanisms driving intra-brain vascular heterogeneity and fenestrated vessel formation in other organs.
Collapse
Affiliation(s)
- Sweta Parab
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Olivia A Card
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Qiyu Chen
- Departments of Molecular Cell Biology and Molecular Neuroscience, The Weizmann Institute of ScienceRehovotIsrael
| | - Michelle America
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de BruxellesGosseliesBelgium
| | - Luke D Buck
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Rachael E Quick
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - William F Horrigan
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Gil Levkowitz
- Departments of Molecular Cell Biology and Molecular Neuroscience, The Weizmann Institute of ScienceRehovotIsrael
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de BruxellesGosseliesBelgium
| | - Ryota L Matsuoka
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve UniversityClevelandUnited States
| |
Collapse
|
30
|
Rada CC, Yuki K, Ding J, Kuo CJ. Regulation of the Blood-Brain Barrier in Health and Disease. Cold Spring Harb Perspect Med 2023; 13:a041191. [PMID: 36987582 PMCID: PMC10691497 DOI: 10.1101/cshperspect.a041191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The neurovascular unit is a dynamic microenvironment with tightly controlled signaling and transport coordinated by the blood-brain barrier (BBB). A properly functioning BBB allows sufficient movement of ions and macromolecules to meet the high metabolic demand of the central nervous system (CNS), while protecting the brain from pathogenic and noxious insults. This review describes the main cell types comprising the BBB and unique molecular signatures of these cells. Additionally, major signaling pathways for BBB development and maintenance are highlighted. Finally, we describe the pathophysiology of BBB diseases, their relationship to barrier dysfunction, and identify avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Cara C Rada
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Jie Ding
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
31
|
Wälchli T, Bisschop J, Carmeliet P, Zadeh G, Monnier PP, De Bock K, Radovanovic I. Shaping the brain vasculature in development and disease in the single-cell era. Nat Rev Neurosci 2023; 24:271-298. [PMID: 36941369 PMCID: PMC10026800 DOI: 10.1038/s41583-023-00684-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/23/2023]
Abstract
The CNS critically relies on the formation and proper function of its vasculature during development, adult homeostasis and disease. Angiogenesis - the formation of new blood vessels - is highly active during brain development, enters almost complete quiescence in the healthy adult brain and is reactivated in vascular-dependent brain pathologies such as brain vascular malformations and brain tumours. Despite major advances in the understanding of the cellular and molecular mechanisms driving angiogenesis in peripheral tissues, developmental signalling pathways orchestrating angiogenic processes in the healthy and the diseased CNS remain incompletely understood. Molecular signalling pathways of the 'neurovascular link' defining common mechanisms of nerve and vessel wiring have emerged as crucial regulators of peripheral vascular growth, but their relevance for angiogenesis in brain development and disease remains largely unexplored. Here we review the current knowledge of general and CNS-specific mechanisms of angiogenesis during brain development and in brain vascular malformations and brain tumours, including how key molecular signalling pathways are reactivated in vascular-dependent diseases. We also discuss how these topics can be studied in the single-cell multi-omics era.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada.
| | - Jeroen Bisschop
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB & Department of Oncology, KU Leuven, Leuven, Belgium
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Donald K. Johnson Research Institute, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Science and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ivan Radovanovic
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
32
|
Wang F, Wang Y, Qiu W, Zhang Q, Yang H, Song G. Crystal Structure of the Extracellular Domains of GPR110. J Mol Biol 2023; 435:167979. [PMID: 36716818 DOI: 10.1016/j.jmb.2023.167979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
Adhesion G protein-coupled receptors (aGPCRs) play a pivotal role in human immune responses, cellular communication, organ development, and other processes. GPR110 belongs to the aGPCR subfamily VI and was initially identified as an oncogene involved in lung and prostate cancers. GPR110 contains tandem adhesion domains at the extracellular region that mediate inter-cellular signaling. However, the structural organization and signaling mechanism for these tandem domains remain unclear. Here, we report the crystal structure of a GPR110 fragment composing the SEA, HormR, and GAIN domains at 2.9 Å resolution. The structure together with MD simulations reveal rigid connections between these domains that are stabilized by complementary interfaces. Strikingly, we found N-linked carbohydrates attached to N389 of the GAIN domain form extensive contacts with the preceding HormR domain. These interactions appear to be critical for folding, as removal of the glycosylation site greatly decreases expression of the GPR110 extracellular fragment. We further demonstrate that the ligand synaptamide fits well within the hydrophobic pocket occupied by the Stachel peptide in the rest state. This suggests that the agonist may function by removing the Stachel peptide which in turn redocks to the orthosteric pocket for receptor activation. Taken together, our structural findings and analyses provide novel insights into the activation mechanism for aGPCRs.
Collapse
Affiliation(s)
- Fangfang Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yang Wang
- Center of Biomedical Physics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
| | - Weicheng Qiu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiansen Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Huaiyu Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Gaojie Song
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
33
|
Yao D, Zhang R, Xie M, Ding F, Wang M, Wang W. Updated Understanding of the Glial-Vascular Unit in Central Nervous System Disorders. Neurosci Bull 2023; 39:503-518. [PMID: 36374471 PMCID: PMC10043098 DOI: 10.1007/s12264-022-00977-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022] Open
Abstract
The concept of the glial-vascular unit (GVU) was raised recently to emphasize the close associations between brain cells and cerebral vessels, and their coordinated reactions to diverse neurological insults from a "glio-centric" view. GVU is a multicellular structure composed of glial cells, perivascular cells, and perivascular space. Each component is closely linked, collectively forming the GVU. The central roles of glial and perivascular cells and their multi-level interconnections in the GVU under normal conditions and in central nervous system (CNS) disorders have not been elucidated in detail. Here, we comprehensively review the intensive interactions between glial cells and perivascular cells in the niche of perivascular space, which take part in the modulation of cerebral blood flow and angiogenesis, formation of the blood-brain barrier, and clearance of neurotoxic wastes. Next, we discuss dysfunctions of the GVU in various neurological diseases, including ischemic stroke, spinal cord injury, Alzheimer's disease, and major depression disorder. In addition, we highlight the possible therapies targeting the GVU, which may have potential clinical applications.
Collapse
Affiliation(s)
- Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ruoying Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
34
|
Isoform- and ligand-specific modulation of the adhesion GPCR ADGRL3/Latrophilin3 by a synthetic binder. Nat Commun 2023; 14:635. [PMID: 36746957 PMCID: PMC9902482 DOI: 10.1038/s41467-023-36312-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 01/24/2023] [Indexed: 02/08/2023] Open
Abstract
Adhesion G protein-coupled receptors (aGPCRs) are cell-surface proteins with large extracellular regions that bind to multiple ligands to regulate key biological functions including neurodevelopment and organogenesis. Modulating a single function of a specific aGPCR isoform while affecting no other function and no other receptor is not trivial. Here, we engineered an antibody, termed LK30, that binds to the extracellular region of the aGPCR ADGRL3, and specifically acts as an agonist for ADGRL3 but not for its isoform, ADGRL1. The LK30/ADGRL3 complex structure revealed that the LK30 binding site on ADGRL3 overlaps with the binding site for an ADGRL3 ligand - teneurin. In cellular-adhesion assays, LK30 specifically broke the trans-cellular interaction of ADGRL3 with teneurin, but not with another ADGRL3 ligand - FLRT3. Our work provides proof of concept for the modulation of isoform- and ligand-specific aGPCR functions using unique tools, and thus establishes a foundation for the development of fine-tuned aGPCR-targeted therapeutics.
Collapse
|
35
|
Fetsko AR, Sebo DJ, Taylor MR. Brain endothelial cells acquire blood-brain barrier properties in the absence of Vegf-dependent CNS angiogenesis. Dev Biol 2023; 494:46-59. [PMID: 36502932 PMCID: PMC9870987 DOI: 10.1016/j.ydbio.2022.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/08/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
During neurovascular development, brain endothelial cells (BECs) respond to secreted signals from the neuroectoderm that regulate CNS angiogenesis, the formation of new blood vessels in the brain, and barriergenesis, the acquisition of blood-brain barrier (BBB) properties. Wnt/β-catenin signaling and Vegf signaling are both required for CNS angiogenesis; however, the relationship between these pathways is not understood. Furthermore, while Wnt/β-catenin signaling is essential for barriergenesis, the role of Vegf signaling in this vital process remains unknown. Here, we provide the first direct evidence, to our knowledge, that Vegf signaling is not required for barriergenesis and that activation of Wnt/β-catenin in BECs is independent of Vegf signaling during neurovascular development. Using double transgenic glut1b:mCherry and plvap:EGFP zebrafish (Danio rerio) to visualize the developing brain vasculature, we performed a forward genetic screen and identified a new mutant allele of kdrl, an ortholog of mammalian Vegfr2. The kdrl mutant lacks CNS angiogenesis but, unlike the Wnt/β-catenin pathway mutant gpr124, acquires BBB properties in BECs. To examine Wnt/β-catenin pathway activation in BECs, we chemically inhibited Vegf signaling and found robust expression of the Wnt/β-catenin transcriptional reporter line 7xtcf-Xla.Siam:EGFP. Taken together, our results establish that Vegf signaling is essential for CNS angiogenesis but is not required for Wnt/β-catenin-dependent barriergenesis. Given the clinical significance of either inhibiting pathological angiogenesis or stimulating neovascularization, our study provides valuable new insights that are critical for the development of effective therapies that target the vasculature in neurological disorders.
Collapse
Affiliation(s)
- Audrey R Fetsko
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Dylan J Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael R Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
36
|
Adhesion G protein-coupled receptors-Structure and functions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 195:1-25. [PMID: 36707149 DOI: 10.1016/bs.pmbts.2022.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Adhesion G protein-coupled receptors (aGPCRs) are an ancient class of receptors that represent some of the largest transmembrane-integrated proteins in humans. First recognized as surface markers on immune cells, it took more than a decade to appreciate their 7-transmembrane structure, which is reminiscent of GPCRs. Roughly 30 years went by before the first functional proof of an interaction with a G protein was published. Besides classic features of GPCRs (extracellular N terminus, 7-transmembrane region, intracellular C terminus), aGPCRs display a distinct N-terminal structure, which harbors the highly conserved GPCR autoproteolysis-inducing (GAIN) domain with the GPCR proteolysis site (GPS) in addition to several functional domains. Several human diseases have been associated with variants of aGPCRs and subsequent animal models have been established to investigate these phenotypes. Much progress has been made in recent years to decipher the structure and functions of these receptors. This chapter gives an overview of our current understanding with respect to the molecular structural patterns governing aGPCR activation and the contribution of these giant molecules to the development of pathologies.
Collapse
|
37
|
Vieira JR, Shah B, Dupraz S, Paredes I, Himmels P, Schermann G, Adler H, Motta A, Gärtner L, Navarro-Aragall A, Ioannou E, Dyukova E, Bonnavion R, Fischer A, Bonanomi D, Bradke F, Ruhrberg C, Ruiz de Almodóvar C. Endothelial PlexinD1 signaling instructs spinal cord vascularization and motor neuron development. Neuron 2022; 110:4074-4089.e6. [PMID: 36549270 PMCID: PMC9796814 DOI: 10.1016/j.neuron.2022.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/04/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
How the vascular and neural compartment cooperate to achieve such a complex and highly specialized structure as the central nervous system is still unclear. Here, we reveal a crosstalk between motor neurons (MNs) and endothelial cells (ECs), necessary for the coordinated development of MNs. By analyzing cell-to-cell interaction profiles of the mouse developing spinal cord, we uncovered semaphorin 3C (Sema3C) and PlexinD1 as a communication axis between MNs and ECs. Using cell-specific knockout mice and in vitro assays, we demonstrate that removal of Sema3C in MNs, or its receptor PlexinD1 in ECs, results in premature and aberrant vascularization of MN columns. Those vascular defects impair MN axon exit from the spinal cord. Impaired PlexinD1 signaling in ECs also causes MN maturation defects at later stages. This study highlights the importance of a timely and spatially controlled communication between MNs and ECs for proper spinal cord development.
Collapse
Affiliation(s)
- José Ricardo Vieira
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Im Neuenheimer 234, 69120 Heidelberg, Germany
| | - Bhavin Shah
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Sebastian Dupraz
- Institute for Neurovascular Cell Biology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Isidora Paredes
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Faculty of Biosciences, Heidelberg University, Im Neuenheimer 234, 69120 Heidelberg, Germany
| | - Patricia Himmels
- Faculty of Biosciences, Heidelberg University, Im Neuenheimer 234, 69120 Heidelberg, Germany
| | - Géza Schermann
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Heike Adler
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Alessia Motta
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 60, 20132 Milan, Italy
| | - Lea Gärtner
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Ariadna Navarro-Aragall
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, EC1V 9EL London, UK
| | - Elena Ioannou
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, EC1V 9EL London, UK
| | - Elena Dyukova
- Max-Planck-Institute for Heart and Lung Research, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Remy Bonnavion
- Max-Planck-Institute for Heart and Lung Research, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Andreas Fischer
- Department of Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany; Division Vascular Signaling and Cancer, German Cancer Research Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Dario Bonanomi
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 60, 20132 Milan, Italy
| | - Frank Bradke
- Laboratory of Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, EC1V 9EL London, UK
| | - Carmen Ruiz de Almodóvar
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany; Institute for Neurovascular Cell Biology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany; Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
38
|
Petridi S, Dubal D, Rikhy R, van den Ameele J. Mitochondrial respiration and dynamics of in vivo neural stem cells. Development 2022; 149:285126. [PMID: 36445292 PMCID: PMC10112913 DOI: 10.1242/dev.200870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Neural stem cells (NSCs) in the developing and adult brain undergo many different transitions, tightly regulated by extrinsic and intrinsic factors. While the role of signalling pathways and transcription factors is well established, recent evidence has also highlighted mitochondria as central players in NSC behaviour and fate decisions. Many aspects of cellular metabolism and mitochondrial biology change during NSC transitions, interact with signalling pathways and affect the activity of chromatin-modifying enzymes. In this Spotlight, we explore recent in vivo findings, primarily from Drosophila and mammalian model systems, about the role that mitochondrial respiration and morphology play in NSC development and function.
Collapse
Affiliation(s)
- Stavroula Petridi
- Department of Clinical Neurosciences and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Dnyanesh Dubal
- Department of Clinical Neurosciences and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK.,Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| | - Richa Rikhy
- Biology, Indian Institute of Science Education and Research, Homi Bhabha Road, Pashan, Pune 411008, India
| | - Jelle van den Ameele
- Department of Clinical Neurosciences and MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| |
Collapse
|
39
|
Liebscher I, Cevheroğlu O, Hsiao CC, Maia AF, Schihada H, Scholz N, Soave M, Spiess K, Trajković K, Kosloff M, Prömel S. A guide to adhesion GPCR research. FEBS J 2022; 289:7610-7630. [PMID: 34729908 DOI: 10.1111/febs.16258] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/20/2021] [Accepted: 11/01/2021] [Indexed: 01/14/2023]
Abstract
Adhesion G protein-coupled receptors (aGPCRs) are a class of structurally and functionally highly intriguing cell surface receptors with essential functions in health and disease. Thus, they display a vastly unexploited pharmacological potential. Our current understanding of the physiological functions and signaling mechanisms of aGPCRs form the basis for elucidating further molecular aspects. Combining these with novel tools and methodologies from different fields tailored for studying these unusual receptors yields a powerful potential for pushing aGPCR research from singular approaches toward building up an in-depth knowledge that will facilitate its translation to applied science. In this review, we summarize the state-of-the-art knowledge on aGPCRs in respect to structure-function relations, physiology, and clinical aspects, as well as the latest advances in the field. We highlight the upcoming most pressing topics in aGPCR research and identify strategies to tackle them. Furthermore, we discuss approaches how to promote, stimulate, and translate research on aGPCRs 'from bench to bedside' in the future.
Collapse
Affiliation(s)
- Ines Liebscher
- Division of Molecular Biochemistry, Medical Faculty, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Germany
| | | | - Cheng-Chih Hsiao
- Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - André F Maia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,IBMC - Instituto Biologia Molecular e Celular, Universidade do Porto, Portugal
| | - Hannes Schihada
- C3 Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Nicole Scholz
- Division of General Biochemistry, Medical Faculty, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Germany
| | - Mark Soave
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, UK
| | - Katja Spiess
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Katarina Trajković
- Biology of Robustness Group, Mediterranean Institute for Life Sciences, Split, Croatia
| | - Mickey Kosloff
- Department of Human Biology, Faculty of Natural Sciences, The University of Haifa, Israel
| | - Simone Prömel
- Institute of Cell Biology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
40
|
Goncalves A, Antonetti DA. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS. Fluids Barriers CNS 2022; 19:86. [PMID: 36320068 PMCID: PMC9628113 DOI: 10.1186/s12987-022-00386-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
The unique environment of the brain and retina is tightly regulated by blood-brain barrier and the blood-retinal barrier, respectively, to ensure proper neuronal function. Endothelial cells within these tissues possess distinct properties that allow for controlled passage of solutes and fluids. Pericytes, glia cells and neurons signal to endothelial cells (ECs) to form and maintain the barriers and control blood flow, helping to create the neurovascular unit. This barrier is lost in a wide range of diseases affecting the central nervous system (CNS) and retina such as brain tumors, stroke, dementia, and in the eye, diabetic retinopathy, retinal vein occlusions and age-related macular degeneration to name prominent examples. Recent studies directly link barrier changes to promotion of disease pathology and degradation of neuronal function. Understanding how these barriers form and how to restore these barriers in disease provides an important point for therapeutic intervention. This review aims to describe the fundamentals of the blood-tissue barriers of the CNS and how the use of transgenic animal models led to our current understanding of the molecular framework of these barriers. The review also highlights examples of targeting barrier properties to protect neuronal function in disease states.
Collapse
Affiliation(s)
- Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA.
| |
Collapse
|
41
|
Lala T, Hall RA. Adhesion G protein-coupled receptors: structure, signaling, physiology, and pathophysiology. Physiol Rev 2022; 102:1587-1624. [PMID: 35468004 PMCID: PMC9255715 DOI: 10.1152/physrev.00027.2021] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/11/2022] [Accepted: 04/16/2022] [Indexed: 01/17/2023] Open
Abstract
Adhesion G protein-coupled receptors (AGPCRs) are a family of 33 receptors in humans exhibiting a conserved general structure but diverse expression patterns and physiological functions. The large NH2 termini characteristic of AGPCRs confer unique properties to each receptor and possess a variety of distinct domains that can bind to a diverse array of extracellular proteins and components of the extracellular matrix. The traditional view of AGPCRs, as implied by their name, is that their core function is the mediation of adhesion. In recent years, though, many surprising advances have been made regarding AGPCR signaling mechanisms, activation by mechanosensory forces, and stimulation by small-molecule ligands such as steroid hormones and bioactive lipids. Thus, a new view of AGPCRs has begun to emerge in which these receptors are seen as massive signaling platforms that are crucial for the integration of adhesive, mechanosensory, and chemical stimuli. This review article describes the recent advances that have led to this new understanding of AGPCR function and also discusses new insights into the physiological actions of these receptors as well as their roles in human disease.
Collapse
Affiliation(s)
- Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
42
|
Evaluation of the Synergistic Potential of Simultaneous Pan- or Isoform-Specific BET and SYK Inhibition in B-Cell Lymphoma: An In Vitro Approach. Cancers (Basel) 2022; 14:cancers14194691. [PMID: 36230614 PMCID: PMC9564024 DOI: 10.3390/cancers14194691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary B-cell lymphomas represent the majority of non-Hodgkin lymphomas and are the most common lymphoid malignancies in the Western world. Genetic alterations or epigenetic modulations can lead to tumor initiation and tumor progression. Aside from standard care, targeted, individualized therapies can be highly effective. Here, we evaluated the impact of simultaneous specific inhibition of two key regulators involved in B lymphoid tumor progression. Spleen tyrosine kinase (SYK) is a B-cell receptor-associated kinase acting as a proto-oncogene in B-cell malignancies, while bromodomain and extra-terminal domain (BET) proteins are epigenetic reader proteins involved in histone recognition and transcription regulation. The simultaneous inhibition of SYK and BET showed enhanced anti-proliferative effects, as well as inducing a distinct combination-specific gene expression profile, suggesting SYK and BET inhibition as a promising combination in the treatment of B-cell lymphoma. Abstract Background: Both bromodomain and extra-terminal domain (BET) proteins and spleen tyrosine kinase (SYK) represent promising targets in diffuse large B-cell (DLBCL) and Burkitt’s lymphoma (BL). We evaluated the anti-lymphoma activity of the isoform-specific bivalent BET inhibitor AZD5153 (AZD) and the pan-BET inhibitor I-BET151 (I-BET) as single agents and in combination with SYK inhibitor Entospletinib (Ento) in vitro. Methods: The effect of the single agents on cell proliferation and metabolic activity was evaluated in two DLBCL and two BL cell lines. Proliferation, metabolic activity, apoptosis, cell cycle and morphology were further investigated after a combined treatment of AZD or I-BET and Ento. RNAseq profiling of combined AZD+Ento treatment was performed in SU-DHL-4 cells. Results: Both BET inhibitors reduced cell proliferation and metabolic activity in a dose- and time-dependent manner. Combined BET and SYK inhibition enhanced the anti-proliferative effect and induced a G0/G1 cell cycle arrest. SU-DHL-4 demonstrated a pronounced modulation of gene expression by AZD, which was markedly increased by additional SYK inhibition. Functional enrichment analyses identified combination-specific GO terms related to DNA replication and cell division. Genes such as ADGRA2, MYB, TNFRSF11A, S100A10, PLEKHH3, DHRS2 and FOXP1-AS1 were identified as possible key regulators. Conclusion: Simultaneous inhibition of BET and SYK enhanced the anti-proliferative effects, and induced a combination-specific gene expression signature.
Collapse
|
43
|
Yin A, Guan X, Zhang JV, Niu J. Focusing on the role of secretin/adhesion (Class B) G protein-coupled receptors in placental development and preeclampsia. Front Cell Dev Biol 2022; 10:959239. [PMID: 36187484 PMCID: PMC9515905 DOI: 10.3389/fcell.2022.959239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Preeclampsia, a clinical syndrome mainly characterized by hypertension and proteinuria, with a worldwide incidence of 3–8% and high maternal mortality, is a risk factor highly associated with maternal and offspring cardiovascular disease. However, the etiology and pathogenesis of preeclampsia are complicated and have not been fully elucidated. Obesity, immunological diseases and endocrine metabolic diseases are high-risk factors for the development of preeclampsia. Effective methods to treat preeclampsia are lacking, and termination of pregnancy remains the only curative treatment for preeclampsia. The pathogenesis of preeclampsia include poor placentation, uteroplacental malperfusion, oxidative stress, endoplasmic reticulum stress, dysregulated immune tolerance, vascular inflammation and endothelial cell dysfunction. The notion that placenta is the core factor in the pathogenesis of preeclampsia is still prevailing. G protein-coupled receptors, the largest family of membrane proteins in eukaryotes and the largest drug target family to date, exhibit diversity in structure and function. Among them, the secretin/adhesion (Class B) G protein-coupled receptors are essential drug targets for human diseases, such as endocrine diseases and cardiometabolic diseases. Given the great value of the secretin/adhesion (Class B) G protein-coupled receptors in the regulation of cardiovascular system function and the drug target exploration, we summarize the role of these receptors in placental development and preeclampsia, and outlined the relevant pathological mechanisms, thereby providing potential drug targets for preeclampsia treatment.
Collapse
Affiliation(s)
- Aiqi Yin
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Xiaonian Guan
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Jian V. Zhang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory of Metabolic Health, Shenzhen, China
- *Correspondence: Jian V. Zhang, ; Jianmin Niu,
| | - Jianmin Niu
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
- *Correspondence: Jian V. Zhang, ; Jianmin Niu,
| |
Collapse
|
44
|
Sato Y, Falcone-Juengert J, Tominaga T, Su H, Liu J. Remodeling of the Neurovascular Unit Following Cerebral Ischemia and Hemorrhage. Cells 2022; 11:2823. [PMID: 36139398 PMCID: PMC9496956 DOI: 10.3390/cells11182823] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Formulated as a group effort of the stroke community, the transforming concept of the neurovascular unit (NVU) depicts the structural and functional relationship between brain cells and the vascular structure. Composed of both neural and vascular elements, the NVU forms the blood-brain barrier that regulates cerebral blood flow to meet the oxygen demand of the brain in normal physiology and maintain brain homeostasis. Conversely, the dysregulation and dysfunction of the NVU is an essential pathological feature that underlies neurological disorders spanning from chronic neurodegeneration to acute cerebrovascular events such as ischemic stroke and cerebral hemorrhage, which were the focus of this review. We also discussed how common vascular risk factors of stroke predispose the NVU to pathological changes. We synthesized existing literature and first provided an overview of the basic structure and function of NVU, followed by knowledge of how these components remodel in response to ischemic stroke and brain hemorrhage. A greater understanding of the NVU dysfunction and remodeling will enable the design of targeted therapies and provide a valuable foundation for relevant research in this area.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Jaime Falcone-Juengert
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| | - Teiji Tominaga
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hua Su
- Department of Anesthesia, UCSF, San Francisco, CA 94143, USA
- Center for Cerebrovascular Research, UCSF, San Francisco, CA 94143, USA
| | - Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| |
Collapse
|
45
|
Abstract
The central nervous system (CNS) has been viewed as an immunologically privileged site, but emerging works are uncovering a large array of neuroimmune interactions primarily occurring at its borders. CNS barriers sites host diverse population of both innate and adaptive immune cells capable of, directly and indirectly, influence the function of the residing cells of the brain parenchyma. These structures are only starting to reveal their role in controlling brain function under normal and pathological conditions and represent an underexplored therapeutic target for the treatment of brain disorders. This review will highlight the development of the CNS barriers to host neuro-immune interactions and emphasize their newly described roles in neurodevelopmental, neurological, and neurodegenerative disorders, particularly for the meninges.
Collapse
Affiliation(s)
- Natalie M Frederick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gabriel A Tavares
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Antoine Louveau
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Cleveland Clinic College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Kent University, Neurosciences, School of Biomedical Sciences, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Sreepada A, Tiwari M, Pal K. Adhesion G protein-coupled receptor gluing action guides tissue development and disease. J Mol Med (Berl) 2022; 100:1355-1372. [PMID: 35969283 DOI: 10.1007/s00109-022-02240-0] [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: 01/25/2022] [Revised: 06/23/2022] [Accepted: 07/21/2022] [Indexed: 10/15/2022]
Abstract
Phylogenetic analysis of human G protein-coupled receptors (GPCRs) divides these transmembrane signaling proteins into five groups: glutamate, rhodopsin, adhesion, frizzled, and secretin families, commonly abbreviated as the GRAFS classification system. The adhesion GPCR (aGPCR) sub-family comprises 33 different receptors in humans. Majority of the aGPCRs are orphan receptors with unknown ligands, structures, and tissue expression profiles. They have a long N-terminal extracellular domain (ECD) with several adhesion sites similar to integrin receptors. Many aGPCRs undergo autoproteolysis at the GPCR proteolysis site (GPS), enclosed within the larger GPCR autoproteolysis inducing (GAIN) domain. Recent breakthroughs in aGPCR research have created new paradigms for understanding their roles in organogenesis. They play crucial roles in multiple aspects of organ development through cell signaling, intercellular adhesion, and cell-matrix associations. They are involved in essential physiological processes like regulation of cell polarity, mitotic spindle orientation, cell adhesion, and migration. Multiple aGPCRs have been associated with the development of the brain, musculoskeletal system, kidneys, cardiovascular system, hormone secretion, and regulation of immune functions. Since aGPCRs have crucial roles in tissue patterning and organogenesis, mutations in these receptors are often associated with diseases with loss of tissue integrity. Thus, aGPCRs include a group of enigmatic receptors with untapped potential for elucidating novel signaling pathways leading to drug discovery. We summarized the current knowledge on how aGPCRs play critical roles in organ development and discussed how aGPCR mutations/genetic variants cause diseases.
Collapse
Affiliation(s)
- Abhijit Sreepada
- Department of Biology, Ashoka University, Rajiv Gandhi Education City, Sonipat, Haryana, 131029, India
| | - Mansi Tiwari
- Department of Biology, Ashoka University, Rajiv Gandhi Education City, Sonipat, Haryana, 131029, India
| | - Kasturi Pal
- Department of Biology, Ashoka University, Rajiv Gandhi Education City, Sonipat, Haryana, 131029, India.
| |
Collapse
|
47
|
ADGRL1 haploinsufficiency causes a variable spectrum of neurodevelopmental disorders in humans and alters synaptic activity and behavior in a mouse model. Am J Hum Genet 2022; 109:1436-1457. [PMID: 35907405 PMCID: PMC9388395 DOI: 10.1016/j.ajhg.2022.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/22/2022] [Indexed: 02/06/2023] Open
Abstract
ADGRL1 (latrophilin 1), a well-characterized adhesion G protein-coupled receptor, has been implicated in synaptic development, maturation, and activity. However, the role of ADGRL1 in human disease has been elusive. Here, we describe ten individuals with variable neurodevelopmental features including developmental delay, intellectual disability, attention deficit hyperactivity and autism spectrum disorders, and epilepsy, all heterozygous for variants in ADGRL1. In vitro, human ADGRL1 variants expressed in neuroblastoma cells showed faulty ligand-induced regulation of intracellular Ca2+ influx, consistent with haploinsufficiency. In vivo, Adgrl1 was knocked out in mice and studied on two genetic backgrounds. On a non-permissive background, mice carrying a heterozygous Adgrl1 null allele exhibited neurological and developmental abnormalities, while homozygous mice were non-viable. On a permissive background, knockout animals were also born at sub-Mendelian ratios, but many Adgrl1 null mice survived gestation and reached adulthood. Adgrl1-/- mice demonstrated stereotypic behaviors, sexual dysfunction, bimodal extremes of locomotion, augmented startle reflex, and attenuated pre-pulse inhibition, which responded to risperidone. Ex vivo synaptic preparations displayed increased spontaneous exocytosis of dopamine, acetylcholine, and glutamate, but Adgrl1-/- neurons formed synapses in vitro poorly. Overall, our findings demonstrate that ADGRL1 haploinsufficiency leads to consistent developmental, neurological, and behavioral abnormalities in mice and humans.
Collapse
|
48
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15. [DOI: https:/doi.org/10.3389/fnmol.2022.910543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
|
49
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15:910543. [PMID: 35935338 PMCID: PMC9354928 DOI: 10.3389/fnmol.2022.910543] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
Affiliation(s)
- Hao Wu
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Min Wei
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Yuping Li
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Qiang Ma
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Hengzhu Zhang
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| |
Collapse
|
50
|
Rattner A, Wang Y, Nathans J. Signaling Pathways in Neurovascular Development. Annu Rev Neurosci 2022; 45:87-108. [PMID: 35803586 DOI: 10.1146/annurev-neuro-111020-102127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During development, the central nervous system (CNS) vasculature grows to precisely meet the metabolic demands of neurons and glia. In addition, the vast majority of the CNS vasculature acquires a unique set of molecular and cellular properties-collectively referred to as the blood-brain barrier-that minimize passive diffusion of molecules between the blood and the CNS parenchyma. Both of these processes are controlled by signals emanating from neurons and glia. In this review, we describe the nature and mechanisms-of-action of these signals, with an emphasis on vascular endothelial growth factor (VEGF) and beta-catenin (canonical Wnt) signaling, the two best-understood systems that regulate CNS vascular development. We highlight foundational discoveries, interactions between different signaling systems, the integration of genetic and cell biological studies, advances that are of clinical relevance, and questions for future research.
Collapse
Affiliation(s)
- Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States;
| | - Yanshu Wang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States; .,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States; .,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Departments of Neuroscience and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|