1
|
Zhang CL, Ma JJ, Li X, Yan HQ, Gui YK, Yan ZX, You MF, Zhang P. The role of transcytosis in the blood-retina barrier: from pathophysiological functions to drug delivery. Front Pharmacol 2025; 16:1565382. [PMID: 40308764 PMCID: PMC12040858 DOI: 10.3389/fphar.2025.1565382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
The blood-retina barrier (BRB) serves as a critical interface that separates the retina from the circulatory system, playing an essential role in preserving the homeostasis of the microenvironment within the retina. Specialized tight junctions and limited vesicle trafficking restrict paracellular and transcellular transport, respectively, thereby maintaining BRB barrier properties. Additionally, transcytosis of macromolecules through retinal vascular endothelial cells constitutes a primary mechanism for transporting substances from the vascular compartment into the surrounding tissue. This review summarizes the fundamental aspects of transcytosis including its function in the healthy retina, the biochemical properties of transcytosis, and the methodologies used to study this process. Furthermore, we discuss the current understanding of transcytosis in the context of pathological BRB breakdown and present recent findings that highlight significant advances in drug delivery to the retina based on transcytosis.
Collapse
Affiliation(s)
- Chun-Lin Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jing-Jie Ma
- Department of Audit, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiang Li
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Hai-Qing Yan
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yong-Kun Gui
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhi-Xin Yan
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ming-Feng You
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ping Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 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
|
Dobariya KH, Goyal D, Kumar H. Molecular signature-based labeling techniques for vascular endothelial cells. Acta Histochem 2025; 127:152222. [PMID: 39644518 DOI: 10.1016/j.acthis.2024.152222] [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: 07/13/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Vascular endothelial cells (VECs) play a crucial role in the development and maintenance of vascular biology specific to the tissue types. Molecular signature-based labeling and imaging of VECs help researchers understand potential mechanisms linking VECs to disease pathology, serving as valuable biomarkers in clinical settings and trials. Labeling techniques involve selectively tagging or marking VECs for visualization. Immunolabeled employs antibodies that specifically bind to VECs markers, while fluorescent tracers or dyes can directly label VECs for imaging. Some techniques use specific carbohydrate residues on cell surface, while others employ endothelial-specific promoters to express fluorescent proteins. Additionally, VEC can be labeled with contrast agents, radiolabeled tracers, and nanoparticles. The choice of labeling technique depends on study context, including whether it involves animal models, in vitro cell cultures, or clinical applications. Herein, we discussed the various labeling methods utilized to label VECs and the techniques to visualize them.
Collapse
Affiliation(s)
- Krutika H Dobariya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Divya Goyal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
4
|
Estrada CC, Wilson C, Gujarati N, Ahmed S, Bronstein R, Daehn IS, Revelo MP, Guo Y, Mallipattu SK. Induction of plasmalemmal vesicle-associated protein exacerbates glomerular endothelial injury in thrombotic microangiopathy. Am J Physiol Renal Physiol 2025; 328:F190-F203. [PMID: 39706169 DOI: 10.1152/ajprenal.00283.2024] [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: 10/03/2024] [Revised: 11/19/2024] [Accepted: 12/07/2024] [Indexed: 12/23/2024] Open
Abstract
Glomerular endothelial cell (GEnC) injury is a common feature across the wide spectrum of glomerular diseases. We recently reported that the endothelial-specific knockout of Krüppel-like factor 4 (Klf4) increases the susceptibility to GEnC injury and subsequent development of subacute thrombotic microangiopathy (TMA). However, the mechanism(s) mediating GEnCs response to injury in TMA are poorly understood. Single-nucleus RNA-sequencing demonstrated enrichment in pathways involved in angiogenesis, permeability, focal adhesion, dedifferentiation, and cytoskeletal organization in the endothelial cluster in mice with TMA. Plasmalemmal vesicle-associated protein (Plvap), a structural component of fenestral diaphragms, was highly enriched specifically in injured GEnCs. Induction of Plvap in cultured GEnCs increased proliferation, migration, and cell permeability with an accompanying loss of mature GEnC markers. Immunostaining for PLVAP in human kidney biopsies confirmed the increase in glomerular PLVAP in TMA, which correlated with a higher grade of glomerular injury. To date, this is the first study to show that the induction of Plvap in GEnCs shifts the cells to an immature state, which might exacerbate glomerular injury in TMA.NEW & NOTEWORTHY This study investigated the mechanism(s) underlying glomerular endothelial cell (GEnC) injury in thrombotic microangiopathy (TMA). We identified plasmalemmal vesicle-associated protein (PLVAP) as specifically upregulated in injured GEnCs in TMA, which was accompanied by pathways involved in angiogenesis and loss of differentiation. Induction of Plvap increased proliferation and migration of GEnCs. Human kidney biopsies with TMA demonstrated an increase in glomerular PLVAP, which correlated with histological markers of GEnC injury, confirming its pathologic role in TMA.
Collapse
Affiliation(s)
- Chelsea C Estrada
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Division of Nephrology, Department of Medicine, Northport Veterans Affairs Medical Center, Northport, New York, United States
| | - Craig Wilson
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Division of Nephrology, Department of Medicine, Northport Veterans Affairs Medical Center, Northport, New York, United States
| | - Nehaben Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Sumaya Ahmed
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Robert Bronstein
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Ilse S Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Monica P Revelo
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States
| | - Yiqing Guo
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Division of Nephrology, Department of Medicine, Northport Veterans Affairs Medical Center, Northport, New York, United States
| |
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 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
|
8
|
Gajbhiye DS, Fernandes GL, Oz I, Nahmias Y, Golan M. A transient neurohormonal circuit controls hatching in fish. Science 2024; 386:1173-1178. [PMID: 39636978 DOI: 10.1126/science.ado8929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024]
Abstract
Hatching is a critical event in the life history of oviparous species. The decision to hatch is often carefully timed to coincide with favorable conditions that will improve survival through early life stages. However, how the relevant cues are relayed to trigger hatching remains unknown. In this work, we show that thyrotropin-releasing hormone (Trh) is the neuroendocrine activator of hatching in zebrafish. To elicit hatching, Trh neurons form a transient circuit that deposits the peptide into the embryo's circulation. Trh also activates hatching in a distantly related fish species that separated more than 200 million years ago. Our results reveal an evolutionarily conserved neuroendocrine circuit that controls a major life event in oviparous fish species.
Collapse
Affiliation(s)
- Deodatta S Gajbhiye
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Genevieve L Fernandes
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Itay Oz
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuni Nahmias
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Matan Golan
- Department of Aquaculture and Poultry, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, Rishon Letziyon, Israel
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
9
|
Bassi I, Grunspan M, Hen G, Ravichandran KA, Moshe N, Gutierrez-Miranda L, Safriel SR, Kostina D, Shen A, Ruiz de Almodovar C, Yaniv K. Endolysosomal dysfunction in radial glia progenitor cells leads to defective cerebral angiogenesis and compromised blood-brain barrier integrity. Nat Commun 2024; 15:8158. [PMID: 39289367 PMCID: PMC11408700 DOI: 10.1038/s41467-024-52365-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
The neurovascular unit (NVU) is a complex multicellular structure that helps maintain cerebral homeostasis and blood-brain barrier (BBB) integrity. While extensive evidence links NVU alterations to cerebrovascular diseases and neurodegeneration, the underlying molecular mechanisms remain unclear. Here, we use zebrafish embryos carrying a mutation in Scavenger Receptor B2, a highly conserved endolysosomal protein expressed predominantly in Radial Glia Cells (RGCs), to investigate the interplay among different NVU components. Through live imaging and genetic manipulations, we demonstrate that compromised acidification of the endolysosomal compartment in mutant RGCs leads to impaired Notch3 signaling, thereby inducing excessive neurogenesis and reduced glial differentiation. We further demonstrate that alterations to the neuron/glia balance result in impaired VEGF and Wnt signaling, leading to severe vascular defects, hemorrhages, and a leaky BBB. Altogether, our findings provide insights into NVU formation and function and offer avenues for investigating diseases involving white matter defects and vascular abnormalities.
Collapse
Affiliation(s)
- Ivan Bassi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Grunspan
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gideon Hen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kishore A Ravichandran
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Noga Moshe
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Laura Gutierrez-Miranda
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Stav R Safriel
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Kostina
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Amitay Shen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Carmen Ruiz de Almodovar
- Institute for Neurovascular Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
- Schlegel Chair for Neurovascular Cell Biology, University of Bonn, Bonn, Germany
| | - Karina Yaniv
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
10
|
Kiper K, Mild B, Chen J, Yuan C, Wells EM, Zheng W, Freeman JL. Cerebral Vascular Toxicity after Developmental Exposure to Arsenic (As) and Lead (Pb) Mixtures. TOXICS 2024; 12:624. [PMID: 39330552 PMCID: PMC11435665 DOI: 10.3390/toxics12090624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/28/2024]
Abstract
Arsenic (As) and lead (Pb) are environmental pollutants found in common sites linked to similar adverse health effects. This study determined driving factors of neurotoxicity on the developing cerebral vasculature with As and Pb mixture exposures. Cerebral vascular toxicity was evaluated at mixture concentrations of As and Pb representing human exposures levels (10 or 100 parts per billion; ppb; µg/L) in developing zebrafish by assessing behavior, morphology, and gene expression. In the visual motor response assay, hyperactivity was observed in all three outcomes in dark phases in larvae with exposure (1-120 h post fertilization, hpf) to 10 ppb As, 10 ppb Pb, or 10 ppb mix treatment. Time spent moving exhibited hyperactivity in dark phases for 100 ppb As and 100 ppb mix treatment groups only. A decreased brain length and ratio of brain length to total length in the 10 ppb mix group was measured with no alterations in other treatment groups or other endpoints (i.e., total larval length, head length, or head width). Alternatively, measurements of cerebral vasculature in the midbrain and cerebellum uncovered decreased total vascularization at 72 hpf in all treatment groups in the mesencephalon and in all treatment groups, except the 100 ppb Pb and 10 ppb As groups, in the cerebellum. In addition, decreased sprouting and branching occurred in the mesencephalon, while only decreased branching was measured in the cerebellum. The 10 ppb Pb group showed several cerebral vasculature modifications that were aligned with a specific gene expression alteration pattern different from other treatment groups. Additionally, the 100 ppb As group drove gene alterations, along with several other endpoints, for changes observed in the 100 ppb mix treatment group. Perturbations assessed in this study displayed non-linear concentration-responses, which are important to consider in environmental health outcomes for As and Pb neurotoxicity.
Collapse
Affiliation(s)
- Keturah Kiper
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Breeann Mild
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jenny Chen
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Ellen M. Wells
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
11
|
Kakogiannos N, Scalise AA, Martini E, Maderna C, Benvenuto AF, D’Antonio M, Carmignani L, Magni S, Gullotta GS, Lampugnani MG, Iannelli F, Beznoussenko GV, Mironov AA, Cerutti C, Bentley K, Philippides A, Zanardi F, Bacigaluppi M, Sigismund S, Bassani C, Farina C, Martino G, De Giovanni M, Dejana E, Iannacone M, Inverso D, Giannotta M. GPR126 is a specifier of blood-brain barrier formation in the mouse central nervous system. J Clin Invest 2024; 134:e165368. [PMID: 39087467 PMCID: PMC11290973 DOI: 10.1172/jci165368] [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: 09/13/2022] [Accepted: 06/04/2024] [Indexed: 08/02/2024] Open
Abstract
The blood-brain barrier (BBB) acquires unique properties to regulate neuronal function during development. The formation of the BBB, which occurs in tandem with angiogenesis, is directed by the Wnt/β-catenin signaling pathway. Yet the exact molecular interplay remains elusive. Our study reveals the G protein-coupled receptor GPR126 as a critical target of canonical Wnt signaling, essential for the development of the BBB's distinctive vascular characteristics and its functional integrity. Endothelial cell-specific deletion of the Gpr126 gene in mice induced aberrant vascular morphogenesis, resulting in disrupted BBB organization. Simultaneously, heightened transcytosis in vitro compromised barrier integrity, resulting in enhanced vascular permeability. Mechanistically, GPR126 enhanced endothelial cell migration, pivotal for angiogenesis, acting through an interaction between LRP1 and β1 integrin, thereby balancing the levels of β1 integrin activation and recycling. Overall, we identified GPR126 as a specifier of an organotypic vascular structure, which sustained angiogenesis and guaranteed the acquisition of the BBB properties during development.
Collapse
Affiliation(s)
| | | | - Emanuele Martini
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Claudio Maderna
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Michele D’Antonio
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Carmignani
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Serena Magni
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giorgia Serena Gullotta
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS, San Raffaele Hospital, Milan, Italy
| | | | - Fabio Iannelli
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | | | | | - Camilla Cerutti
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Katie Bentley
- The Francis Crick Institute, London, United Kingdom
- Department of Informatics, King’s College London, London, United Kingdom
| | - Andrew Philippides
- Department of Informatics, University of Sussex, Brighton, United Kingdom
| | - Federica Zanardi
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Bacigaluppi
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS, San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Sara Sigismund
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Claudia Bassani
- Immunobiology of Neurological Disorders Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinthia Farina
- Immunobiology of Neurological Disorders Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS, San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marco De Giovanni
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Monica Giannotta
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
12
|
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
|
13
|
Thomas JR, Frye WJE, Robey RW, Warner AC, Butcher D, Matta JL, Morgan TC, Edmondson EF, Salazar PB, Ambudkar SV, Gottesman MM. Abcg2a is the functional homolog of human ABCG2 expressed at the zebrafish blood-brain barrier. Fluids Barriers CNS 2024; 21:27. [PMID: 38491505 PMCID: PMC10941402 DOI: 10.1186/s12987-024-00529-5] [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: 12/21/2023] [Accepted: 03/04/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND A principal protective component of the mammalian blood-brain barrier (BBB) is the high expression of the multidrug efflux transporters P-glycoprotein (P-gp, encoded by ABCB1) and ABCG2 (encoded by ABCG2) on the lumenal surface of endothelial cells. The zebrafish P-gp homolog Abcb4 is expressed at the BBB and phenocopies human P-gp. Comparatively little is known about the four zebrafish homologs of the human ABCG2 gene: abcg2a, abcg2b, abcg2c, and abcg2d. Here we report the functional characterization and brain tissue distribution of zebrafish ABCG2 homologs. METHODS To determine substrates of the transporters, we stably expressed each in HEK-293 cells and performed cytotoxicity and fluorescent efflux assays with known ABCG2 substrates. To assess the expression of transporter homologs, we used a combination of RNAscope in situ hybridization probes and immunohistochemistry to stain paraffin-embedded sections of adult and larval zebrafish. RESULTS We found Abcg2a had the greatest substrate overlap with ABCG2, and Abcg2d appeared to be the least functionally similar. We identified abcg2a as the only homolog expressed at the adult and larval zebrafish BBB, based on its localization to claudin-5 positive brain vasculature. CONCLUSIONS These results demonstrate the conserved function of zebrafish Abcg2a and suggest that zebrafish may be an appropriate model organism for studying the role of ABCG2 at the BBB.
Collapse
Affiliation(s)
- Joanna R Thomas
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 2108, Bethesda, MD, 20892, USA
| | - William J E Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 2108, Bethesda, MD, 20892, USA
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 2108, Bethesda, MD, 20892, USA
| | - Andrew C Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jennifer L Matta
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tamara C Morgan
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Elijah F Edmondson
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Paula B Salazar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 2108, Bethesda, MD, 20892, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 2108, Bethesda, MD, 20892, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 2108, Bethesda, MD, 20892, USA.
| |
Collapse
|
14
|
Fetsko AR, Sebo DJ, Budzynski LB, Scharbarth A, Taylor MR. IL-1β disrupts blood-brain barrier development by inhibiting endothelial Wnt/β-catenin signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.04.569943. [PMID: 38106202 PMCID: PMC10723338 DOI: 10.1101/2023.12.04.569943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
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 development of the BBB. We generated a doxycycline-inducible transgenic zebrafish model that drives secretion of 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 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, USA
| | - Dylan J. Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lilyana B. Budzynski
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Alli Scharbarth
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael R. Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
15
|
Thomas JR, Frye WJE, Robey RW, Warner AC, Butcher D, Matta JL, Morgan TC, Edmondson EF, Salazar PB, Ambudkar SV, Gottesman MM. Abcg2a is the functional homolog of human ABCG2 expressed at the zebrafish blood-brain barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.18.539313. [PMID: 37425689 PMCID: PMC10327217 DOI: 10.1101/2023.05.18.539313] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background A principal protective component of the mammalian blood-brain barrier (BBB) is the high expression of the multidrug efflux transporters P-glycoprotein (P-gp, encoded by ABCB1) and ABCG2 (encoded by ABCG2) on the lumenal surface of endothelial cells. The zebrafish P-gp homolog Abcb4 is expressed at the BBB and phenocopies human P-gp. Comparatively little is known about the four zebrafish homologs of the human ABCG2 gene: abcg2a, abcg2b, abcg2c, and abcg2d. Here we report the functional characterization and brain tissue distribution of zebrafish ABCG2 homologs. Methods To determine substrates of the transporters, we stably expressed each in HEK-293 cells and performed cytotoxicity and fluorescent efflux assays with known ABCG2 substrates. To assess the expression of transporter homologs, we used a combination of RNAscope in situ hybridization probes and immunohistochemistry to stain paraffin-embedded sections of adult and larval zebrafish. Results We found Abcg2a had the greatest substrate overlap with ABCG2, and Abcg2d appeared to be the least functionally similar. We identified abcg2a as the only homolog expressed at the adult and larval zebrafish BBB, based on its localization to claudin-5 positive brain vasculature. Conclusions These results demonstrate the conserved function of zebrafish Abcg2a and suggest that zebrafish may be an appropriate model organism for the studying the role of ABCG2 at the BBB.
Collapse
Affiliation(s)
- Joanna R. Thomas
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William J. E. Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Robert W. Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrew C. Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jennifer L. Matta
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tamara C. Morgan
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Elijah F. Edmondson
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Paula B. Salazar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Suresh V. Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael M. Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Badaut J, Ghersi-Egea JF, Thorne RG, Konsman JP. Blood-brain borders: a proposal to address limitations of historical blood-brain barrier terminology. Fluids Barriers CNS 2024; 21:3. [PMID: 38183042 PMCID: PMC10770911 DOI: 10.1186/s12987-023-00478-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/11/2023] [Indexed: 01/07/2024] Open
Abstract
Many neuroscientists use the term Blood-Brain Barrier (BBB) to emphasize restrictiveness, often equating or reducing the notion of BBB properties to tight junction molecules physically sealing cerebral endothelial cells, rather than pointing out the complexity of this biological interface with respect to its selectivity and variety of exchange between the general blood circulation and the central nervous tissue. Several authors in the field find it unfortunate that the exquisitely dynamic interfaces between blood and brain continue to be viewed primarily as obstructive barriers to transport. Although the term blood-brain interface is an excellent descriptor that does not convey the idea of a barrier, it is important and preferable for the spreading of an idea beyond specialist communities to try to appeal to well-chosen metaphors. Recent evidence reviewed here indicates that blood-brain interfaces are more than selective semi-permeable membranes in that they display many dynamic processes and complex mechanisms for communication. They are thus more like 'geopolitical borders'. Furthermore, some authors working on blood-brain interface-relevant issues have started to use the word border, for example in border-associated macrophages. Therefore, we suggest adopting the term Blood-Brain Border to better communicate the flexibility of and movement across blood-brain interfaces.
Collapse
Affiliation(s)
- Jerome Badaut
- Brain Molecular Imaging Lab, UMR 5536, CNRS, RMSB, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
- Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Jean-François Ghersi-Egea
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR 5292, Lyon-1 University, Bron, France.
| | - Robert G Thorne
- Denali Therapeutics, Inc, 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
- Department of Pharmaceutics, University of Minnesota, 9-177 Weaver-Densford Hall, 308 Harvard St. SE, Minneapolis, MN, 55455, USA.
| | - Jan Pieter Konsman
- UMR 5164, CNRS, ImmunoConcEpT, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
| |
Collapse
|
17
|
O'Brown NM, Patel NB, Hartmann U, Klein AM, Gu C, Megason SG. The secreted neuronal signal Spock1 promotes blood-brain barrier development. Dev Cell 2023; 58:1534-1547.e6. [PMID: 37437574 PMCID: PMC10525910 DOI: 10.1016/j.devcel.2023.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/07/2023] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
The blood-brain barrier (BBB) is a unique set of properties of the brain vasculature which severely restrict its permeability to proteins and small molecules. Classic chick-quail chimera studies have shown that these properties are not intrinsic to the brain vasculature but rather are induced by surrounding neural tissue. Here, we identify Spock1 as a candidate neuronal signal for regulating BBB permeability in zebrafish and mice. Mosaic genetic analysis shows that neuronally expressed Spock1 is cell non-autonomously required for a functional BBB. Leakage in spock1 mutants is associated with altered extracellular matrix (ECM), increased endothelial transcytosis, and altered pericyte-endothelial interactions. Furthermore, a single dose of recombinant SPOCK1 partially restores BBB function in spock1 mutants by quenching gelatinase activity and restoring vascular expression of BBB genes including mcamb. These analyses support a model in which neuronally secreted Spock1 initiates BBB properties by altering the ECM, thereby regulating pericyte-endothelial interactions and downstream vascular gene expression.
Collapse
Affiliation(s)
- Natasha M O'Brown
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA.
| | - Nikit B Patel
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 52, 50931 Cologne, Germany
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | - Chenghua Gu
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Zabrodskaya Y, Paramonova N, Litovchenko A, Bazhanova E, Gerasimov A, Sitovskaya D, Nezdorovina V, Kravtsova S, Malyshev S, Skiteva E, Samochernykh K. Neuroinflammatory Dysfunction of the Blood-Brain Barrier and Basement Membrane Dysplasia Play a Role in the Development of Drug-Resistant Epilepsy. Int J Mol Sci 2023; 24:12689. [PMID: 37628870 PMCID: PMC10454729 DOI: 10.3390/ijms241612689] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Drug-resistance epilepsy (DRE) is a key problem in neurology. It is possible that damage to the blood-brain barrier (BBB) may affect resistance in DRE. The aim of this work was to assess the damage and dysfunction in the BBB in the area of epileptic foci in patients with DRE under conditions of neuroinflammation. The changes to the BBB in temporal lobe epilepsy (by immunohistochemistry and transmission electron microscopy), levels of neuroinflammatory proteins, and cytokine levels in the blood (by multiplex analysis) were studied. Increased levels of vascular endothelial growth factor (VEGF) and growth-regulated protein (GRO), and decreased levels of epidermal growth factor (EGF) in plasma, combined with overexpression of the VEGF-A receptor by endotheliocytes were detected. Malformation-like growths of the basement membrane of the capillaries of the brain complicate the delivery of antiepileptic drugs (AEDs). Dysplasia of the basement membrane is the result of inadequate reparative processes in chronic inflammation. In conclusion, it should be noted that damage to the microcirculatory network of the brain should be considered one of the leading factors contributing to DRE.
Collapse
Affiliation(s)
- Yulia Zabrodskaya
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
| | - Natalia Paramonova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia; (N.P.); (A.L.); (E.B.)
- State Research Testing Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - Anastasia Litovchenko
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia; (N.P.); (A.L.); (E.B.)
| | - Elena Bazhanova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia; (N.P.); (A.L.); (E.B.)
- Golikov Research Center of Toxicology, 192019 St. Petersburg, Russia
| | - Aleksandr Gerasimov
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
| | - Darya Sitovskaya
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
| | - Victoria Nezdorovina
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
| | - Svetlana Kravtsova
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
| | - Stanislav Malyshev
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
| | - Ekaterina Skiteva
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
- State Scientific Center of the Russian Federation, Institute of Biomedical Problems of the Russian Academy of Sciences, 123007 Moscow, Russia
| | - Konstantin Samochernykh
- Polenov Neurosurgical Institute—Branch of the Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.G.); (D.S.); (V.N.); (S.K.); (S.M.); (E.S.); (K.S.)
| |
Collapse
|
19
|
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
|
20
|
Denzer L, Muranyi W, Schroten H, Schwerk C. The role of PLVAP in endothelial cells. Cell Tissue Res 2023; 392:393-412. [PMID: 36781482 PMCID: PMC10172233 DOI: 10.1007/s00441-023-03741-1] [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: 08/16/2022] [Accepted: 01/18/2023] [Indexed: 02/15/2023]
Abstract
Endothelial cells play a major part in the regulation of vascular permeability and angiogenesis. According to their duty to fit the needs of the underlying tissue, endothelial cells developed different subtypes with specific endothelial microdomains as caveolae, fenestrae and transendothelial channels which regulate nutrient exchange, leukocyte migration, and permeability. These microdomains can exhibit diaphragms that are formed by the endothelial cell-specific protein plasmalemma vesicle-associated protein (PLVAP), the only known protein component of these diaphragms. Several studies displayed an involvement of PLVAP in diseases as cancer, traumatic spinal cord injury, acute ischemic brain disease, transplant glomerulopathy, Norrie disease and diabetic retinopathy. Besides an upregulation of PLVAP expression within these diseases, pro-angiogenic or pro-inflammatory responses were observed. On the other hand, loss of PLVAP in knockout mice leads to premature mortality due to disrupted homeostasis. Generally, PLVAP is considered as a major factor influencing the permeability of endothelial cells and, finally, to be involved in the regulation of vascular permeability. Following these observations, PLVAP is debated as a novel therapeutic target with respect to the different vascular beds and tissues. In this review, we highlight the structure and functions of PLVAP in different endothelial types in health and disease.
Collapse
Affiliation(s)
- Lea Denzer
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Walter Muranyi
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Horst Schroten
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Christian Schwerk
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
21
|
Arias‐Alpizar G, Papadopoulou P, Rios X, Pulagam KR, Moradi M, Pattipeiluhu R, Bussmann J, Sommerdijk N, Llop J, Kros A, Campbell F. Phase-Separated Liposomes Hijack Endogenous Lipoprotein Transport and Metabolism Pathways to Target Subsets of Endothelial Cells In Vivo. Adv Healthc Mater 2023; 12:e2202709. [PMID: 36565694 PMCID: PMC11469146 DOI: 10.1002/adhm.202202709] [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: 11/08/2022] [Revised: 12/14/2022] [Indexed: 12/25/2022]
Abstract
Plasma lipid transport and metabolism are essential to ensure correct cellular function throughout the body. Dynamically regulated in time and space, the well-characterized mechanisms underpinning plasma lipid transport and metabolism offers an enticing, but as yet underexplored, rationale to design synthetic lipid nanoparticles with inherent cell/tissue selectivity. Herein, a systemically administered liposome formulation, composed of just two lipids, that is capable of hijacking a triglyceride lipase-mediated lipid transport pathway resulting in liposome recognition and uptake within specific endothelial cell subsets is described. In the absence of targeting ligands, liposome-lipase interactions are mediated by a unique, phase-separated ("parachute") liposome morphology. Within the embryonic zebrafish, selective liposome accumulation is observed at the developing blood-brain barrier. In mice, extensive liposome accumulation within the liver and spleen - which is reduced, but not eliminated, following small molecule lipase inhibition - supports a role for endothelial lipase but highlights these liposomes are also subject to significant "off-target" by reticuloendothelial system organs. Overall, these compositionally simplistic liposomes offer new insights into the discovery and design of lipid-based nanoparticles that can exploit endogenous lipid transport and metabolism pathways to achieve cell selective targeting in vivo.
Collapse
Affiliation(s)
- Gabriela Arias‐Alpizar
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Division of BioTherapeuticsLeiden Academic Centre for Drug ResearchLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Panagiota Papadopoulou
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Xabier Rios
- CIC biomaGUNEBasque Research and Technology Alliance (BRTA)San Sebastián20014Spain
| | | | - Mohammad‐Amin Moradi
- Materials and Interface ChemistryDepartment of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600The Netherlands
| | - Roy Pattipeiluhu
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Jeroen Bussmann
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Division of BioTherapeuticsLeiden Academic Centre for Drug ResearchLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Nico Sommerdijk
- Department of BiochemistryRadboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegen6525The Netherlands
- Electron Microscopy CentreRadboudumc Technology Center MicroscopyRadboud University Medical CenterGeert Grooteplein Zuid 28Nijmegen6525The Netherlands
| | - Jordi Llop
- Materials and Interface ChemistryDepartment of Chemical Engineering and ChemistryEindhoven University of TechnologyP.O. Box 513Eindhoven5600The Netherlands
| | - Alexander Kros
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
| | - Frederick Campbell
- Supramolecular and Biomaterials ChemistryLeiden Institute of ChemistryLeiden UniversityP.O. Box 9502Leiden2300The Netherlands
- Present address:
NanoVation Therapeutics2405 Wesbrook Mall 4th floorVancouverBCV6T 1Z3Canada
| |
Collapse
|
22
|
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: 11] [Impact Index Per Article: 5.5] [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
|
23
|
Analysis of Vascular Morphogenesis in Zebrafish. Methods Mol Biol 2023; 2608:425-450. [PMID: 36653721 DOI: 10.1007/978-1-0716-2887-4_24] [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: 01/19/2023]
Abstract
Analysis of cardiovascular development in zebrafish embryos has become a major driver of vascular research in recent years. Imaging-based analyses have allowed the discovery or verification of morphologically distinct processes and mechanisms of, e.g., endothelial cell migration, angiogenic sprouting, tip or stalk cell behavior, and vessel anastomosis. In this chapter, we describe the techniques and tools used for confocal imaging of zebrafish endothelial development in combination with general experimental approaches for molecular dissection of involved signaling pathways.
Collapse
|
24
|
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
|
25
|
The presence of BBB hastens neuronal differentiation of cerebral organoids - The potential role of endothelial derived BDNF. Biochem Biophys Res Commun 2022; 626:30-37. [PMID: 35970042 DOI: 10.1016/j.bbrc.2022.07.112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 07/30/2022] [Indexed: 11/23/2022]
Abstract
Despite remaining the best in vitro model to resemble the human brain, a weakness of human cerebral organoids is the lack of the endothelial component that in vivo organizes in the blood brain barrier (BBB). Since the BBB is crucial to control the microenvironment of the nervous system, this study proposes a co-culture of BBB and cerebral organoids. We utilized a BBB model consisting of primary human brain microvascular endothelial cells and astrocytes in a transwell system. Starting from induced Pluripotent Stem Cells (iPSCs) we generated human cerebral organoids which were then cultured in the absence or presence of an in vitro model of BBB to evaluate potential effects on the maturation of cerebral organoids. By morphological analysis, it emerges that in the presence of the BBB the cerebral organoids are better organized than controls in the absence of the BBB. This effect might be due to Brain Derived Neurotrophic Factor (BDNF), a neurotrophic factor released by the endothelial component of the BBB, which is involved in neurodevelopment, neuroplasticity and neurosurvival.
Collapse
|
26
|
Tran KA, Baldwin-Leclair A, DeOre BJ, Antisell M, Galie PA. Oxygen gradients dictate angiogenesis but not barriergenesis in a 3D brain microvascular model. J Cell Physiol 2022; 237:3872-3882. [PMID: 35901247 DOI: 10.1002/jcp.30840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/01/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022]
Abstract
A variety of biophysical properties are known to regulate angiogenic sprouting, and in vitro systems can parse the individual effects of these factors in a controlled setting. Here, a three-dimensional brain microvascular model interrogates how variables including extracellular matrix composition, fluid shear stress, and radius of curvature affect angiogenic sprouting of cerebral endothelial cells. Tracking endothelial migration over several days reveals that application of fluid shear stress and enlarged vessel radius of curvature both attenuate sprouting. Computational modeling informed by oxygen consumption assays suggests that sprouting correlates to reduced oxygen concentration: both fluid shear stress and vessel geometry alter the local oxygen levels dictated by both ambient conditions and cellular respiration. Moreover, increasing cell density and consequently lowering the local oxygen levels yields significantly more sprouting. Further analysis reveals that the magnitude of oxygen concentration is not as important as its spatial concentration gradient: decreasing ambient oxygen concentration causes significantly less sprouting than applying an external oxygen gradient to the vessels. In contrast, barriergenesis is dictated by shear stress independent of local oxygen concentrations, suggesting that different mechanisms mediate angiogenesis and barrier formation and that angiogenic sprouting can occur without compromising the barrier. Overall, these results improve our understanding of how specific biophysical variables regulate the function and activation of cerebral vasculature, and identify spatial oxygen gradients as the driving factor of angiogenesis in the brain.
Collapse
Affiliation(s)
- Kiet A Tran
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
| | | | - Brandon J DeOre
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
| | - Morgan Antisell
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
| | - Peter A Galie
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
| |
Collapse
|
27
|
Ramos-Zaldívar HM, Polakovicova I, Salas-Huenuleo E, Corvalán AH, Kogan MJ, Yefi CP, Andia ME. Extracellular vesicles through the blood-brain barrier: a review. Fluids Barriers CNS 2022; 19:60. [PMID: 35879759 PMCID: PMC9310691 DOI: 10.1186/s12987-022-00359-3] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/15/2022] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are particles naturally released from cells that are delimited by a lipid bilayer and are unable to replicate. How the EVs cross the Blood–Brain barrier (BBB) in a bidirectional manner between the bloodstream and brain parenchyma remains poorly understood. Most in vitro models that have evaluated this event have relied on monolayer transwell or microfluidic organ-on-a-chip techniques that do not account for the combined effect of all cellular layers that constitute the BBB at different sites of the Central Nervous System. There has not been direct transcytosis visualization through the BBB in mammals in vivo, and evidence comes from in vivo experiments in zebrafish. Literature is scarce on this topic, and techniques describing the mechanisms of EVs motion through the BBB are inconsistent. This review will focus on in vitro and in vivo methodologies used to evaluate EVs transcytosis, how EVs overcome this fundamental structure, and discuss potential methodological approaches for future analyses to clarify these issues. Understanding how EVs cross the BBB will be essential for their future use as vehicles in pharmacology and therapeutics.
Collapse
Affiliation(s)
- Héctor M Ramos-Zaldívar
- Doctoral Program in Medical Sciences, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago de Chile, Chile.
| | - Iva Polakovicova
- Advanced Center for Chronic Diseases, Santiago, Chile.,Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Alejandro H Corvalán
- Advanced Center for Chronic Diseases, Santiago, Chile.,Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo J Kogan
- Advanced Center for Chronic Diseases, Santiago, Chile.,Departamento de Química Farmacológica Y Toxicológica, Facultad de Ciencias Químicas Y Farmacéuticas, Laboratorio de Nanobiotecnología, Universidad de Chile, Carlos Lorca 964, Independencia, Chile
| | - Claudia P Yefi
- Escuela de Medicina Veterinaria, Facultad de Agronomía E Ingeniería Forestal, Facultad de Ciencias Biológicas Y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo E Andia
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute for Intelligent Healthcare Engineering, Santiago, Chile
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Matsuoka RL, Buck LD, Vajrala KP, Quick RE, Card OA. Historical and current perspectives on blood endothelial cell heterogeneity in the brain. Cell Mol Life Sci 2022; 79:372. [PMID: 35726097 PMCID: PMC9209386 DOI: 10.1007/s00018-022-04403-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Dynamic brain activity requires timely communications between the brain parenchyma and circulating blood. Brain-blood communication is facilitated by intricate networks of brain vasculature, which display striking heterogeneity in structure and function. This vascular cell heterogeneity in the brain is fundamental to mediating diverse brain functions and has long been recognized. However, the molecular basis of this biological phenomenon has only recently begun to be elucidated. Over the past century, various animal species and in vitro systems have contributed to the accumulation of our fundamental and phylogenetic knowledge about brain vasculature, collectively advancing this research field. Historically, dye tracer and microscopic observations have provided valuable insights into the anatomical and functional properties of vasculature across the brain, and these techniques remain an important approach. Additionally, recent advances in molecular genetics and omics technologies have revealed significant molecular heterogeneity within brain endothelial and perivascular cell types. The combination of these conventional and modern approaches has enabled us to identify phenotypic differences between healthy and abnormal conditions at the single-cell level. Accordingly, our understanding of brain vascular cell states during physiological, pathological, and aging processes has rapidly expanded. In this review, we summarize major historical advances and current knowledge on blood endothelial cell heterogeneity in the brain, and discuss important unsolved questions in the field.
Collapse
Affiliation(s)
- Ryota L Matsuoka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.
| | - Luke D Buck
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Keerti P Vajrala
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA.,Kansas City University College of Osteopathic Medicine, Kansas City, MO 64106, USA
| | - Rachael E Quick
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Olivia A Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
| |
Collapse
|
31
|
Li Y, Wang C, Zhang L, Chen B, Mo Y, Zhang J. Claudin-5a is essential for the functional formation of both zebrafish blood-brain barrier and blood-cerebrospinal fluid barrier. Fluids Barriers CNS 2022; 19:40. [PMID: 35658877 PMCID: PMC9164509 DOI: 10.1186/s12987-022-00337-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/04/2022] [Indexed: 11/25/2022] Open
Abstract
Background Mammalian Claudin-5 is the main endothelial tight junction component maintaining blood-brain barrier (BBB) permeability, while Claudin-1 and -3 seal the paracellular space of choroid plexus (CP) epithelial cells contributing to the blood-cerebrospinal fluid barrier (BCSFB). In zebrafish, two paralogs of claudin-5a and -5b are expressed while their roles in the formation of BBB and BCSFB are unclear. Methods The expression patterns of Claudin-5a and -5b in zebrafish brains were systematically analyzed by immunofluorescence (IF) assay. The developmental functions of Claudin-5a and -5b were characterized by generating of claudin-5a and -5b mutants respectively. Meanwhile, the cerebral inflammation and cell apoptosis in claudin-5a-/- were assessed by live imaging of transgenic zebrafish, RT-qPCR, IF, and TUNEL assay. The integrity of BBB and BCSFB was evaluated by in vivo angiographic and dye permeation assay. Finally, RT-qPCR, whole-mount RNA in situ hybridization (WISH), and transmission electron microscopy (TEM) analyses were performed to investigate the development of cerebral vessels and choroid plexus. Results We showed that Claudin-5a and -5b are both expressed in zebrafish cerebrovascular endothelial cells (ECs). In addition, Claudin-5a was strongly expressed in CP epithelial cells. Loss of Claudin-5b showed no effect on zebrafish vasculogenesis or BBB function. In contrast, the knockout of claudin-5a caused a lethal phenotype of severe whole-brain oedema, ventricular dilatation, and cerebral hernia in zebrafish larvae, although the cerebral vasculogenesis and the development of CP were not altered. In claudin-5a-/- , although ultrastructural analysis of CP and cerebral capillary showed intact integrity of epithelial and endothelial tight junctions, permeability assay indicated a disruption of both BBB and BCSFB functions. On the molecular level, it was found that ZO-1 was upregulated in the CP epithelium of claudin-5a-/-, while the notch and shh pathway responsible for CP development was not affected due to loss of Claudin-5a. Conclusions Our findings verified a non-functional role of zebrafish Claudin-5b in the BBB and identified Claudin-5a as the ortholog of mammalian Claudin-5, contributing to the development and the functional maintenance of both BBB and BCSFB. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00337-9.
Collapse
Affiliation(s)
- Yanyu Li
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chunchun Wang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China
| | - Liang Zhang
- College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Bing Chen
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yuqian Mo
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China.,School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang, 524001, China. .,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, China.
| |
Collapse
|
32
|
Abstract
The brain is arguably the most fascinating and complex organ in the human body. Recreating the brain in vitro is an ambition restricted by our limited understanding of its structure and interacting elements. One of these interacting parts, the brain microvasculature, is distinguished by a highly selective barrier known as the blood-brain barrier (BBB), limiting the transport of substances between the blood and the nervous system. Numerous in vitro models have been used to mimic the BBB and constructed by implementing a variety of microfabrication and microfluidic techniques. However, currently available models still cannot accurately imitate the in vivo characteristics of BBB. In this article, we review recent BBB models by analyzing each parameter affecting the accuracy of these models. Furthermore, we propose an investigation of the synergy between BBB models and neuronal tissue biofabrication, which results in more advanced models, including neurovascular unit microfluidic models and vascularized brain organoid-based models.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China.,School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Hongzhao Zhou
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China.,School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Weikang Lv
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China.,School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaobin Xu
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China.,School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310058, China.,School of Mechanical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
33
|
VPS28 regulates brain vasculature by controlling neuronal VEGF trafficking through extracellular vesicle secretion. iScience 2022; 25:104042. [PMID: 35330682 PMCID: PMC8938284 DOI: 10.1016/j.isci.2022.104042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/27/2022] [Accepted: 03/04/2022] [Indexed: 11/21/2022] Open
Abstract
Extracellular vesicles (EVs) participate in intercellular communication and contribute to the angiogenesis. However, the understanding of the mechanisms underlying EVs secretion by neurons and their action on the vascular system of the central nervous system (CNS) remain rudimentary. Here, we show that vacuolar protein sorting 28 (Vps28) is essential for the sprouting of brain central arteries (CtAs) and for the integrity of blood-brain barrier (BBB) in zebrafish. Disruption of neuron-enriched Vps28 significantly decreased EVs secretion by regulating the formation of intracellular multivesicular bodies (MVBs). EVs derived from zebrafish embryos or mouse cortical neurons partially rescued the brain vasculature defect and brain leakage. Further investigations revealed that neuronal EVs containing vascular endothelial growth factor A (VEGF-A) are key regulators in neurovascular communication. Our results indicate that Vps28 acts as an intercellular endosomal regulator mediating the secretion of neuronal EVs, which in turn communicate with endothelial cells to mediate angiogenesis through VEGF-A trafficking. Vps28 is highly expressed in neurons and involved in the secretion of neuronal EVs Vps28, as a subunit of ESCRT-1 complexes, participates in the formation of MVB Vps28 plays an important role in VEGFA transport and promotes neurovascular communication
Collapse
|
34
|
Bowley G, Kugler E, Wilkinson R, Lawrie A, van Eeden F, Chico TJA, Evans PC, Noël ES, Serbanovic-Canic J. Zebrafish as a tractable model of human cardiovascular disease. Br J Pharmacol 2022; 179:900-917. [PMID: 33788282 DOI: 10.1111/bph.15473] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Mammalian models including non-human primates, pigs and rodents have been used extensively to study the mechanisms of cardiovascular disease. However, there is an increasing desire for alternative model systems that provide excellent scientific value while replacing or reducing the use of mammals. Here, we review the use of zebrafish, Danio rerio, to study cardiovascular development and disease. The anatomy and physiology of zebrafish and mammalian cardiovascular systems are compared, and we describe the use of zebrafish models in studying the mechanisms of cardiac (e.g. congenital heart defects, cardiomyopathy, conduction disorders and regeneration) and vascular (endothelial dysfunction and atherosclerosis, lipid metabolism, vascular ageing, neurovascular physiology and stroke) pathologies. We also review the use of zebrafish for studying pharmacological responses to cardiovascular drugs and describe several features of zebrafish that make them a compelling model for in vivo screening of compounds for the treatment cardiovascular disease. LINKED ARTICLES: This article is part of a themed issue on Preclinical Models for Cardiovascular disease research (BJP 75th Anniversary). To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.5/issuetoc.
Collapse
Affiliation(s)
- George Bowley
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Elizabeth Kugler
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
- Institute of Ophthalmology, Faculty of Brain Sciences, University College London, London, UK
| | - Rob Wilkinson
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Freek van Eeden
- Bateson Centre, University of Sheffield, Sheffield, UK
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Tim J A Chico
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Emily S Noël
- Bateson Centre, University of Sheffield, Sheffield, UK
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
35
|
Lanham KA, Nedden ML, Wise VE, Taylor MR. Genetically inducible and reversible zebrafish model of systemic inflammation. Biol Open 2022; 11:274172. [PMID: 35099005 PMCID: PMC8918989 DOI: 10.1242/bio.058559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/20/2022] [Indexed: 11/20/2022] Open
Abstract
The inflammatory response is a vital defense mechanism against trauma and pathogen induced damage, but equally important is its appropriate resolution. In some instances of severe trauma or sustained infection, inappropriate and persistent activation of the immune response can occur resulting in a dangerous systemic inflammatory response. Untreated, this systemic inflammatory response can lead to tissue damage, organ shutdown, and death. Replicating this condition in tractable model organisms can provide insight into the mechanisms involved in the induction, maintenance, and resolution of inflammation. To that end, we developed a non-invasive, inducible, and reversible model of systemic inflammation in zebrafish. Using the Gal4-EcR/UAS system activated by the ecdysone analog tebufenozide, we generated transgenic zebrafish that allow for chemically-induced, ubiquitous secretion of the mature form of zebrafish interleukin-1β (Il-1βmat) in both larval and adult developmental stages. To ensure a robust immune response, we attached a strong signal peptide from the Gaussia princeps luciferase enzyme to promote active secretion of the cytokine. We observe a dose-dependent inflammatory response involving neutrophil expansion accompanied by tissue damage and reduced survival. Washout of tebufenozide permits inflammation resolution. We also establish the utility of this model for the identification of small molecule anti-inflammatory compounds by treatment with the immunosuppressant rapamycin. Taken together, these features make this model a valuable new tool that can aid in identifying potential new therapies while broadening our understanding of systemic inflammation, its impact on the immune system and its resolution.
Collapse
Affiliation(s)
- Kevin A Lanham
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Megan L Nedden
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Virginia E Wise
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Michael R Taylor
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
36
|
Song X, Cui Y, Wang Y, Zhang Y, He Q, Yu Z, Xu C, Ning H, Han Y, Cai Y, Cheng X, Wang J, Teng Y, Yang X, Wang J. Genome Editing with AAV-BR1-CRISPR in Postnatal Mouse Brain Endothelial Cells. Int J Biol Sci 2022; 18:652-660. [PMID: 35002515 PMCID: PMC8741854 DOI: 10.7150/ijbs.64188] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/27/2021] [Indexed: 12/03/2022] Open
Abstract
Brain endothelial cells (ECs) are an important component of the blood-brain barrier (BBB) and play key roles in restricting entrance of possible toxic components and pathogens into the brain. However, identifying endothelial genes that regulate BBB homeostasis remains a time-consuming process. Although somatic genome editing has emerged as a powerful tool for discovery of essential genes regulating tissue homeostasis, its application in brain ECs is yet to be demonstrated in vivo. Here, we used an adeno-associated virus targeting brain endothelium (AAV-BR1) combined with the CRISPR/Cas9 system (AAV-BR1-CRISPR) to specifically knock out genes of interest in brain ECs of adult mice. We first generated a mouse model expressing Cas9 in ECs (Tie2 Cas9). We selected endothelial β-catenin (Ctnnb1) gene, which is essential for maintaining adult BBB integrity, as the target gene. After intravenous injection of AAV-BR1-sgCtnnb1-tdTomato in 4-week-old Tie2 Cas9 transgenic mice resulted in mutation of 36.1% of the Ctnnb1 alleles, thereby leading to a dramatic decrease in the level of CTNNB1 in brain ECs. Consequently, Ctnnb1 gene editing in brain ECs resulted in BBB breakdown. Taken together, these results demonstrate that the AAV-BR1-CRISPR system is a useful tool for rapid identification of endothelial genes that regulate BBB integrity in vivo.
Collapse
Affiliation(s)
- Xiaopeng Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yaxiong Cui
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yanxiao Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yizhe Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Qi He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Zhenyang Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chengfang Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Huimin Ning
- Department of Immunology, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, China
| | - Yuying Han
- Department of Immunology, College of Basic Medicine, Qingdao University, Qingdao, Shandong 266071, China
| | - Yunting Cai
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xuan Cheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yan Teng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jun Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
37
|
Lee S, Chung M, Jeon NL. BBB-on-a-Chip: Modeling Functional Human Blood-Brain Barrier by Mimicking 3D Brain Angiogenesis Using Microfluidic Chip. Methods Mol Biol 2022; 2492:251-263. [PMID: 35733049 DOI: 10.1007/978-1-0716-2289-6_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Organ-on-a-chip enables human cell-based 3D tissue culture, which recapitulates the physiological structure and function of the tissue. In terms of the blood-brain barrier (BBB) modeling, the 3D structure of the vessel is essential for studying the cellular interactions among BBB composing cells and investigating the barrier function. Here, we describe a BBB-on-a-chip model with 3D perfusable human vasculature tri-cultured with pericytes and astrocytes. The culture method is based on mimicking angiogenic sprouting since the barrier formation is parallel with angiogenesis during the developmental process. This microfluidic-based 3D tri-culture system enables the comparative study on how surrounding BBB-related cells affect brain angiogenic sprouting. Moreover, the engineered perfusable vasculature is eligible for quantitative analysis on barrier function such as efflux transport system. We expect the BBB-on-a-chip could be used to enhance understanding BBB-related pathologies as well as the drug modulating barrier function of BBB.
Collapse
Affiliation(s)
- Somin Lee
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Minhwan Chung
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, Republic of Korea.
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Gastfriend BD, Nishihara H, Canfield SG, Foreman KL, Engelhardt B, Palecek SP, Shusta EV. Wnt signaling mediates acquisition of blood-brain barrier properties in naïve endothelium derived from human pluripotent stem cells. eLife 2021; 10:70992. [PMID: 34755601 PMCID: PMC8664294 DOI: 10.7554/elife.70992] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
Endothelial cells (ECs) in the central nervous system (CNS) acquire their specialized blood-brain barrier (BBB) properties in response to extrinsic signals, with Wnt/β-catenin signaling coordinating multiple aspects of this process. Our knowledge of CNS EC development has been advanced largely by animal models, and human pluripotent stem cells (hPSCs) offer the opportunity to examine BBB development in an in vitro human system. Here we show that activation of Wnt signaling in hPSC-derived naïve endothelial progenitors, but not in matured ECs, leads to robust acquisition of canonical BBB phenotypes including expression of GLUT-1, increased claudin-5, decreased PLVAP and decreased permeability. RNA-seq revealed a transcriptome profile resembling ECs with CNS-like characteristics, including Wnt-upregulated expression of LEF1, APCDD1, and ZIC3. Together, our work defines effects of Wnt activation in naïve ECs and establishes an improved hPSC-based model for interrogation of CNS barriergenesis.
Collapse
Affiliation(s)
- Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | | | - Scott G Canfield
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | - Koji L Foreman
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | | | - Sean P Palecek
- Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | - Eric V Shusta
- Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| |
Collapse
|
39
|
Ando K, Ishii T, Fukuhara S. Zebrafish Vascular Mural Cell Biology: Recent Advances, Development, and Functions. Life (Basel) 2021; 11:1041. [PMID: 34685412 PMCID: PMC8537713 DOI: 10.3390/life11101041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Recruitment of mural cells to the vascular wall is essential for forming the vasculature as well as maintaining proper vascular functions. In recent years, zebrafish genetic tools for mural cell biology have improved substantially. Fluorescently labeled zebrafish mural cell reporter lines enable us to study, with higher spatiotemporal resolution than ever, the processes of mural cell development from their progenitors. Furthermore, recent phenotypic analysis of platelet-derived growth factor beta mutant zebrafish revealed well-conserved organotypic mural cell development and functions in vertebrates with the unique features of zebrafish. However, comprehensive reviews of zebrafish mural cells are lacking. Therefore, herein, we highlight recent advances in zebrafish mural cell tools. We also summarize the fundamental features of zebrafish mural cell development, especially at early stages, and functions.
Collapse
Affiliation(s)
- Koji Ando
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Tokyo 113 8602, Japan; (T.I.); (S.F.)
| | | | | |
Collapse
|
40
|
Yu W, Jin H, Sun W, Nan D, Deng J, Jia J, Yu Z, Huang Y. Connexin43 promotes angiogenesis through activating the HIF-1α/VEGF signaling pathway under chronic cerebral hypoperfusion. J Cereb Blood Flow Metab 2021; 41:2656-2675. [PMID: 33899559 PMCID: PMC8504949 DOI: 10.1177/0271678x211010354] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Chronic cerebral hypoperfusion, a major vascular contributor to vascular cognitive impairment and dementia, can exacerbate small vessel pathology. Connexin43, the most abundant gap junction protein in brain tissue, has been found to be critically involved in the pathological changes of vascular cognitive impairment and dementia caused by chronic cerebral hypoperfusion. However, the precise mechanisms underpinning its role are unclear. We established a mouse model via bilateral common carotid arteries stenosis on connexin43 heterozygous male mice and demonstrated that connexin43 improves brain blood flow recovery by mediating reparative angiogenesis under chronic cerebral hypoperfusion, which subsequently reduces the characteristic pathologies of vascular cognitive impairment and dementia including white matter lesions and irreversible neuronal injury. We additionally found that connexin43 mediates hypoxia inducible factor-1α expression and then activates the PKA signaling pathway to regulate vascular endothelial growth factor-induced angiogenesis. All the above findings were replicated in bEnd.3 cells treated with 375 µM CoCl2in vitro. These results suggest that connexin 43 could be instrumental in developing potential therapies for vascular cognitive impairment and dementia caused by chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Sun
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ding Nan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jingjing Jia
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zemou Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
41
|
van Lanen RH, Melchers S, Hoogland G, Schijns OE, Zandvoort MAV, Haeren RH, Rijkers K. Microvascular changes associated with epilepsy: A narrative review. J Cereb Blood Flow Metab 2021; 41:2492-2509. [PMID: 33866850 PMCID: PMC8504411 DOI: 10.1177/0271678x211010388] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier (BBB) is dysfunctional in temporal lobe epilepsy (TLE). In this regard, microvascular changes are likely present. The aim of this review is to provide an overview of the current knowledge on microvascular changes in epilepsy, and includes clinical and preclinical evidence of seizure induced angiogenesis, barriergenesis and microcirculatory alterations. Anatomical studies show increased microvascular density in the hippocampus, amygdala, and neocortex accompanied by BBB leakage in various rodent epilepsy models. In human TLE, a decrease in afferent vessels, morphologically abnormal vessels, and an increase in endothelial basement membranes have been observed. Both clinical and experimental evidence suggests that basement membrane changes, such as string vessels and protrusions, indicate and visualize a misbalance between endothelial cell proliferation and barriergenesis. Vascular endothelial growth factor (VEGF) appears to play a crucial role. Following an altered vascular anatomy, its physiological functioning is affected as expressed by neurovascular decoupling that subsequently leads to hypoperfusion, disrupted parenchymal homeostasis and potentially to seizures". Thus, epilepsy might be a condition characterized by disturbed cerebral microvasculature in which VEGF plays a pivotal role. Additional physiological data from patients is however required to validate findings from models and histological studies on patient biopsies.
Collapse
Affiliation(s)
- Rick Hgj van Lanen
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stan Melchers
- Faculty of Health Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Govert Hoogland
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Olaf Emg Schijns
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marc Amj van Zandvoort
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Molecular Cell Biology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Roel Hl Haeren
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Kim Rijkers
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
42
|
Nanoparticle shell structural cues drive in vitro transport properties, tissue distribution and brain accessibility in zebrafish. Biomaterials 2021; 277:121085. [PMID: 34461457 DOI: 10.1016/j.biomaterials.2021.121085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/26/2021] [Accepted: 08/23/2021] [Indexed: 01/14/2023]
Abstract
Zwitterion polymers with strong antifouling properties have been suggested as the prime alternative to polyethylene glycol (PEG) for drug nanocarriers surface coating. It is believed that PEG coating shortcomings, such as immune responses and incomplete protein repellency, could be overcome by zwitterionic polymers. However, no systematic study has been conducted so far to complete a comparative appraisal of PEG and zwitterionic-coating effects on nanoparticles (NPs) stealthness, cell uptake, cell barrier translocation and biodistribution in the context of nanocarriers brain targeting. Core-shell polymeric particles with identical cores and a shell of either PEG or poly(2-methacryloyloxyethyl phosphorylcholine (PMPC) were prepared by impinging jet mixer nanoprecipitation. NPs with similar size and surface potential were systematically compared using in vitro and in vivo assays. NPs behavior differences were rationalized based on their protein-particles interactions. PMPC-coated NPs were significantly more endocytosed by mouse macrophages or brain resident macrophages compared to PEGylated NPs but exhibited the remarkable ability to cross the blood-brain barrier in in vitro models. Nanoscale flow cytometry assays showed significantly more adsorbed proteins on PMPC-coated NPs than PEG-coated NPs. In vivo, distribution in zebrafish larvae, showed a strong propensity for PMPC-coated NPs to adhere to the vascular endothelium, while PEG-coated NPs were able to circulate for a longer time and escape the bloodstream to penetrate deep into the cerebral tissue. The stark differences between these two types of particles, besides their similarities in size and surface potential, points towards the paramount role of surface chemistry in controlling NPs fate likely via the formation of distinct protein corona for each coating.
Collapse
|
43
|
Umans RA, Ten Kate M, Pollock C, Sontheimer H. Fishing for Contact: Modeling Perivascular Glioma Invasion in the Zebrafish Brain. ACS Pharmacol Transl Sci 2021; 4:1295-1305. [PMID: 34423267 DOI: 10.1021/acsptsci.0c00129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly invasive, central nervous system (CNS) cancer for which there is no cure. Invading tumor cells evade treatment, limiting the efficacy of the current standard of care regimen. Understanding the underlying invasive behaviors that support tumor growth may allow for generation of novel GBM therapies. Zebrafish (Danio rerio) are attractive for genetics and live imaging and have, in recent years, emerged as a model system suitable for cancer biology research. While other groups have studied CNS tumors using zebrafish, few have concentrated on the invasive behaviors supporting the development of these diseases. Previous studies demonstrated that one of the main mechanisms of GBM invasion is perivascular invasion, i.e., single tumor cell migration along blood vessels. Here, we characterize phenotypes, methodology, and potential therapeutic avenues for utilizing zebrafish to model perivascular GBM invasion. Using patient-derived xenolines or an adherent cell line, we demonstrate tumor expansion within the zebrafish brain. Within 24-h postintracranial injection, D54-MG-tdTomato glioma cells produce fingerlike projections along the zebrafish brain vasculature. As few as 25 GBM cells were sufficient to promote single cell vessel co-option. Of note, these tumor-vessel interactions are CNS specific and do not occur on pre-existing blood vessels when injected into the animal's peripheral tissue. Tumor-vessel interactions increase over time and can be pharmacologically disrupted through inhibition of Wnt signaling. Therefore, zebrafish serve as a favorable model system to study perivascular glioma invasion, one of the deadly characteristics that make GBM so difficult to treat.
Collapse
Affiliation(s)
- Robyn A Umans
- Center for Glial Biology in Health, Disease, and Cancer, The Fralin Biomedical Research Institute at VTC, Roanoke, Virginia 24016, United States
| | - Mattie Ten Kate
- School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Carolyn Pollock
- School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Harald Sontheimer
- Center for Glial Biology in Health, Disease, and Cancer, The Fralin Biomedical Research Institute at VTC, Roanoke, Virginia 24016, United States.,School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| |
Collapse
|
44
|
Umans RA, Pollock C, Mills WA, Clark KC, Pan YA, Sontheimer H. Using Zebrafish to Elucidate Glial-Vascular Interactions During CNS Development. Front Cell Dev Biol 2021; 9:654338. [PMID: 34268301 PMCID: PMC8276133 DOI: 10.3389/fcell.2021.654338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022] Open
Abstract
An emerging area of interest in Neuroscience is the cellular relationship between glia and blood vessels, as many of the presumptive support roles of glia require an association with the vasculature. These interactions are best studied in vivo and great strides have been made using mice to longitudinally image glial-vascular interactions. However, these methods are cumbersome for developmental studies, which could benefit from a more accessible system. Zebrafish (Danio rerio) are genetically tractable vertebrates, and given their translucency, are readily amenable for daily live imaging studies. We set out to examine whether zebrafish glia have conserved traits with mammalian glia regarding their ability to interact with and maintain the developing brain vasculature. We utilized transgenic zebrafish strains in which oligodendrocyte transcription factor 2 (olig2) and glial fibrillary acidic protein (gfap) identify different glial populations in the zebrafish brain and document their corresponding relationship with brain blood vessels. Our results demonstrate that olig2+ and gfap+ zebrafish glia have distinct lineages and each interact with brain vessels as previously observed in mouse brain. Additionally, we manipulated these relationships through pharmacological and genetic approaches to distinguish the roles of these cell types during blood vessel development. olig2+ glia use blood vessels as a pathway during their migration and Wnt signaling inhibition decreases their single-cell vessel co-option. By contrast, the ablation of gfap+ glia at the beginning of CNS angiogenesis impairs vessel development through a reduction in Vascular endothelial growth factor (Vegf), supporting a role for gfap+ glia during new brain vessel formation in zebrafish. This data suggests that zebrafish glia, akin to mammalian glia, have different lineages that show diverse interactions with blood vessels, and are a suitable model for elucidating glial-vascular relationships during vertebrate brain development.
Collapse
Affiliation(s)
- Robyn A. Umans
- Glial Biology in Health, Disease, and Cancer Center, The Fralin Biomedical Research Institute at VTC, Roanoke, VA, United States
| | - Carolyn Pollock
- School of Neuroscience, Virginia Tech, Blacksburg, VA, United States
| | - William A. Mills
- Glial Biology in Health, Disease, and Cancer Center, The Fralin Biomedical Research Institute at VTC, Roanoke, VA, United States
| | - Kareem C. Clark
- Center for Neurobiology Research, The Fralin Biomedical Research Institute at VTC, Roanoke, VA, United States
| | - Y. Albert Pan
- Center for Neurobiology Research, The Fralin Biomedical Research Institute at VTC, Roanoke, VA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, The Fralin Biomedical Research Institute at VTC, Roanoke, VA, United States
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
45
|
Zhang T, Xu Z, Wen L, Lei D, Li S, Wang J, Huang J, Wang N, Durkan C, Liao X, Wang G. Cadmium-induced dysfunction of the blood-brain barrier depends on ROS-mediated inhibition of PTPase activity in zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2021; 412:125198. [PMID: 33550130 DOI: 10.1016/j.jhazmat.2021.125198] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/04/2021] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
Increasing evidence has demonstrated that cadmium accumulation in the blood increases the risk of neurological diseases. However, how cadmium breaks through the blood-brain barrier (BBB) and is transferred from the blood circulation into the central nervous system is still unclear. In this study, we examined the toxic effect of cadmium chloride (CdCl2) on the development and function of BBB in zebrafish. CdCl2 exposure induced cerebral hemorrhage, increased BBB permeability and promoted abnormal vascular formation by promoting VEGF production in zebrafish brain. Furthermore, in vivo and in vitro experiments showed that CdCl2 altered cell-cell junctional morphology by disrupting the proper localization of VE-cadherin and ZO-1. The potential mechanism involved in the inhibition of protein tyrosine phosphatase (PTPase) mediated by cadmium-induced ROS was confirmed with diphenylene iodonium (DPI), a ROS production inhibitor. Together, these data indicate that BBB is a critical target of cadmium toxicity and provide in vivo etiological evidence of cadmium-induced neurovascular disease in a zebrafish BBB model.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China; Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Lin Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Daoxi Lei
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Shuyu Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Jinxuan Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| | - Jinxia Huang
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB30FF, UK.
| | - Colm Durkan
- The Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB30FF, UK.
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
46
|
Hotz JM, Thomas JR, Katz EN, Robey RW, Horibata S, Gottesman MM. ATP-binding cassette transporters at the zebrafish blood-brain barrier and the potential utility of the zebrafish as an in vivo model. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:620-633. [PMID: 34308273 PMCID: PMC8297714 DOI: 10.20517/cdr.2021.35] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The brain is protected from toxins by a tightly regulated network of specialized cells, including endothelial cells, pericytes, astrocyes, and neurons, known collectively as the blood-brain barrier (BBB). This selectively permeable barrier permits only the most crucial molecules essential for brain function to enter and employs a number of different mechanisms to prevent the entry of potentially harmful toxins and pathogens. In addition to a physical barrier comprised of endothelial cells that form tight junctions to restrict paracellular transport, there is an active protective mechanism made up of energy-dependent transporters that efflux compounds back into the bloodstream. Two of these ATP-binding cassette (ABC) transporters are highly expressed at the BBB: P-glycoprotein (P-gp, encoded by the ABCB1 gene) and ABCG2 (encoded by the ABCG2 gene). Although a number of in vitro and in vivo systems have been developed to examine the role that ABC transporters play in keeping compounds out of the brain, all have inherent advantages and disadvantages. Zebrafish (Danio rerio) have become a model of interest for studies of the BBB due to the similarities between the zebrafish and mammalian BBB systems. In this review, we discuss what is known about ABC transporters in zebrafish and what information is still needed before the zebrafish can be recommended as a model to elucidate the role of ABC transporters at the BBB.
Collapse
Affiliation(s)
- Jordan M Hotz
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joanna R Thomas
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emily N Katz
- Zebrafish Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sachi Horibata
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Chico TJA, Kugler EC. Cerebrovascular development: mechanisms and experimental approaches. Cell Mol Life Sci 2021; 78:4377-4398. [PMID: 33688979 PMCID: PMC8164590 DOI: 10.1007/s00018-021-03790-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The cerebral vasculature plays a central role in human health and disease and possesses several unique anatomic, functional and molecular characteristics. Despite their importance, the mechanisms that determine cerebrovascular development are less well studied than other vascular territories. This is in part due to limitations of existing models and techniques for visualisation and manipulation of the cerebral vasculature. In this review we summarise the experimental approaches used to study the cerebral vessels and the mechanisms that contribute to their development.
Collapse
Affiliation(s)
- Timothy J A Chico
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| | - Elisabeth C Kugler
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| |
Collapse
|
48
|
Costa KCM, Brigante TAV, Fernandes GG, Scomparin DS, Scarante FF, de Oliveira DP, Campos AC. Zebrafish as a Translational Model: An Experimental Alternative to Study the Mechanisms Involved in Anosmia and Possible Neurodegenerative Aspects of COVID-19? eNeuro 2021; 8:ENEURO.0027-21.2021. [PMID: 33952614 PMCID: PMC8174008 DOI: 10.1523/eneuro.0027-21.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
The Coronavirus disease-2019 (COVID-19) presents a variability of clinical symptoms, ranging from asymptomatic to severe respiratory and systemic conditions. In a cohort of patients, the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2), beyond the classical respiratory manifestations, induces anosmia. Evidence has suggested SARS-CoV-2-induced anosmia can be the result of neurodegeneration of the olfactory pathway. Neurologic symptoms associated with COVID-19 have been reported; however, the precise mechanism and possible long-lasting effects remain poorly investigated. Preclinical models are valuable tools for describing and testing new possible treatments for neurologic disorders. In this way, the zebrafish (Danio rerio) organism model represents an attractive tool in the field of neuroscience, showing economic and logistic advantages besides genetic and physiologic similarities with mammalian, including the brain structure and functions. Besides, its external embryonic development, high availability of eggs, and fast development allows easy genetic manipulation and fast replications. In the present review, we suggest that the zebrafish model can be advantageous to investigate the neurologic features of COVID-19.
Collapse
Affiliation(s)
- Karla C M Costa
- Pharmacology of Neuroplasticity Laboratory, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil, 14049-900,
| | - Tamires A V Brigante
- Pharmacology of Neuroplasticity Laboratory, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil, 14049-900
| | - Gabriel G Fernandes
- Pharmacology of Neuroplasticity Laboratory, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil, 14049-900
| | - Davi S Scomparin
- Pharmacology of Neuroplasticity Laboratory, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil, 14049-900
| | - Franciele F Scarante
- Pharmacology of Neuroplasticity Laboratory, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil, 14049-900
| | - Danielle P de Oliveira
- EcoHumanTox Laboratory, Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Science of Ribeirão Preto, University of São Paulo, São Paulo, Brazil 14049-900
| | - Alline C Campos
- Pharmacology of Neuroplasticity Laboratory, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil, 14049-900
| |
Collapse
|
49
|
Rong J, He Y, Tang J, Qiao R, Lin S. "Fishing" nano-bio interactions at the key biological barriers. NANOSCALE 2021; 13:5954-5964. [PMID: 33734277 DOI: 10.1039/d1nr00328c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Understanding nano-bio interactions is pivotal to the safe implementation of nanotechnology for both biological and environmental applications. Zebrafish as a model organism provides unique opportunities to dissect nano-bio interactions occurring at different biological barriers. In this review, we focus on four key biological barriers, namely cell membrane, blood-brain barrier (BBB), skin and gill epithelia, and gastrointestinal tract (GIT), and highlight recent advancement achieved by using zebrafish to conduct both visualized observations and mechanistic investigations on a diversity of nano-bio interactions.
Collapse
Affiliation(s)
- Jinyu Rong
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China.
| | | | | | | | | |
Collapse
|
50
|
Girolamo F, de Trizio I, Errede M, Longo G, d'Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021; 18:14. [PMID: 33743764 PMCID: PMC7980348 DOI: 10.1186/s12987-021-00242-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Central nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches. ![]()
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Intensive Care Unit, Department of Intensive Care, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Molecular Biology Unit, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|