1
|
Gomez-Salinero JM, Redmond D, Rafii S. Microenvironmental determinants of endothelial cell heterogeneity. Nat Rev Mol Cell Biol 2025:10.1038/s41580-024-00825-w. [PMID: 39875728 DOI: 10.1038/s41580-024-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
During development, endothelial cells (ECs) undergo an extraordinary specialization by which generic capillary microcirculatory networks spanning from arteries to veins transform into patterned organotypic zonated blood vessels. These capillary ECs become specialized to support the cellular and metabolic demands of each specific organ, including supplying tissue-specific angiocrine factors that orchestrate organ development, maintenance of organ-specific functions and regeneration of injured adult organs. Here, we illustrate the mechanisms by which microenvironmental signals emanating from non-vascular niche cells induce generic ECs to acquire specific inter-organ and intra-organ functional attributes. We describe how perivascular, parenchymal and immune cells dictate vascular heterogeneity and capillary zonation, and how this system is maintained through tissue-specific signalling activated by vasculogenic and angiogenic factors and deposition of matrix components. We also discuss how perturbation of organotypic vascular niche cues lead to erasure of EC signatures, contributing to the pathogenesis of disease processes. We also describe approaches that use reconstitution of tissue-specific signatures of ECs to promote regeneration of damaged organs.
Collapse
Affiliation(s)
- Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration and Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Yuan Y, Liu H, Dai Z, He C, Qin S, Su Z. From Physiology to Pathology of Astrocytes: Highlighting Their Potential as Therapeutic Targets for CNS Injury. Neurosci Bull 2025; 41:131-154. [PMID: 39080102 PMCID: PMC11748647 DOI: 10.1007/s12264-024-01258-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 01/19/2025] Open
Abstract
In the mammalian central nervous system (CNS), astrocytes are the ubiquitous glial cells that have complex morphological and molecular characteristics. These fascinating cells play essential neurosupportive and homeostatic roles in the healthy CNS and undergo morphological, molecular, and functional changes to adopt so-called 'reactive' states in response to CNS injury or disease. In recent years, interest in astrocyte research has increased dramatically and some new biological features and roles of astrocytes in physiological and pathological conditions have been discovered thanks to technological advances. Here, we will review and discuss the well-established and emerging astroglial biology and functions, with emphasis on their potential as therapeutic targets for CNS injury, including traumatic and ischemic injury. This review article will highlight the importance of astrocytes in the neuropathological process and repair of CNS injury.
Collapse
Affiliation(s)
- Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Pain Medicine, School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Dai
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2024. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
4
|
Babu SR, Shekara HH, Sahoo AK, Harsha Vardhan PV, Thiruppathi N, Venkatesh MP. Intranasal nanoparticulate delivery systems for neurodegenerative disorders: a review. Ther Deliv 2023; 14:571-594. [PMID: 37691577 DOI: 10.4155/tde-2023-0019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 09/12/2023] Open
Abstract
Neurodegenerative diseases are a significant cause of mortality worldwide, and the blood-brain barrier (BBB) poses a significant challenge for drug delivery. An intranasal route is a prominent approach among the various methods to bypass the BBB. There are different pathways involved in intranasal drug delivery. The drawbacks of this method include mucociliary clearance, enzymatic degradation and poor drug permeation. Novel nanoformulations and intranasal drug-delivery devices offer promising solutions to overcome these challenges. Nanoformulations include polymeric nanoparticles, lipid-based nanoparticles, microspheres, liposomes and noisomes. Additionally, intranasal devices could be utilized to enhance drug-delivery efficacy. Therefore, intranasal drug-delivery systems show potential for treating neurodegenerative diseases through trigeminal or olfactory pathways, which can significantly improve patient outcomes.
Collapse
Affiliation(s)
- Someshbabu Ramesh Babu
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Harshith Hosahalli Shekara
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Ashish Kumar Sahoo
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Pyda Venkata Harsha Vardhan
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Nitheesh Thiruppathi
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Madhugiri Prakash Venkatesh
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Faculty of Pharmaceutical Sciences, UCSI University, Kaula Lampur, Malaysia
| |
Collapse
|
5
|
Kumar BS, Menon SC, Gayathri SR, Chakravarthy VS. The Influence of Neural Activity and Neural Cytoarchitecture on Cerebrovascular Arborization: A Computational Model. Front Neurosci 2022; 16:917196. [PMID: 35860300 PMCID: PMC9290769 DOI: 10.3389/fnins.2022.917196] [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] [Academic Contribution Register] [Received: 04/10/2022] [Accepted: 05/30/2022] [Indexed: 11/18/2022] Open
Abstract
Normal functioning of the brain relies on a continual and efficient delivery of energy by a vast network of cerebral blood vessels. The bidirectional coupling between neurons and blood vessels consists of vasodilatory energy demand signals from neurons to blood vessels, and the retrograde flow of energy substrates from the vessels to neurons, which fuel neural firing, growth and other housekeeping activities in the neurons. Recent works indicate that, in addition to the functional coupling observed in the adult brain, the interdependence between the neural and vascular networks begins at the embryonic stage, and continues into subsequent developmental stages. The proposed Vascular Arborization Model (VAM) captures the effect of neural cytoarchitecture and neural activity on vascular arborization. The VAM describes three important stages of vascular tree growth: (i) The prenatal growth phase, where the vascular arborization depends on the cytoarchitecture of neurons and non-neural cells, (ii) the post-natal growth phase during which the further arborization of the vasculature depends on neural activity in addition to neural cytoarchitecture, and (iii) the settling phase, where the fully grown vascular tree repositions its vascular branch points or nodes to ensure minimum path length and wire length. The vasculature growth depicted by VAM captures structural characteristics like vascular volume density, radii, mean distance to proximal neurons in the cortex. VAM-grown vasculature agrees with the experimental observation that the neural densities do not covary with the vascular density along the depth of the cortex but predicts a high correlation between neural areal density and microvascular density when compared over a global scale (across animals and regions). To explore the influence of neural activity on vascular arborization, the VAM was used to grow the vasculature in neonatal rat whisker barrel cortex under two conditions: (i) Control, where the whiskers were intact and (ii) Lesioned, where one row of whiskers was cauterized. The model captures a significant reduction in vascular branch density in lesioned animals compared to control animals, concurring with experimental observation.
Collapse
Affiliation(s)
- Bhadra S. Kumar
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Sarath C. Menon
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| | | | - V. Srinivasa Chakravarthy
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
- Center for Complex Systems and Dynamics, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
6
|
García-Salvador A, Domínguez-Monedero A, Gómez-Fernández P, García-Bilbao A, Carregal-Romero S, Castilla J, Goñi-de-Cerio F. Evaluation of the Influence of Astrocytes on In Vitro Blood-Brain Barrier Models. Altern Lab Anim 2020; 48:184-200. [PMID: 33136430 DOI: 10.1177/0261192920966954] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
In vitro blood-brain barrier (BBB) models are a useful tool to screen the permeability and toxicity of new drugs. Currently, many different in vitro BBB models coexist, but none stands out as being notably better than the rest. Therefore, there is still a need to evaluate the quality of BBB models under various conditions and assess their ability to mimic the in vivo situation. In this study, two brain endothelial cell lines (bEnd.3 and hCMEC/D3) and two epithelial-like cell lines (MDCKII and Caco-2) were selected for BBB modelling purposes. They were grown as monolayers of a single cell type, under the following conditions: in coculture with either primary or immortalised astrocytes; or in the presence of primary or immortalised astrocyte-derived conditioned media. A total of 20 different BBB models were established in this manner, in order to assess the effects of the astroglial components on the BBB phenotype in each case. To this end, six parameters were studied: the expression of selected tight junction proteins; the enzyme activities of alkaline phosphatase and of gamma glutamyl transpeptidase; the transendothelial/transepithelial electrical resistance (TEER); restriction in paracellular transport; and efflux transporter inhibition were each evaluated and correlated. The results showed that coculturing with either primary or immortalised astrocytes led to a general improvement in all parameters studied, evidencing the contribution of this cell type to effective BBB formation. Furthermore, the permeability coefficient (P e) of the tracer molecule, Lucifer Yellow, correlated with three of the six parameters studied. In addition, this study highlights the potential for the use of the Lucifer Yellow P e value as an indicator of barrier integrity in in vitro BBB models, which could be useful for screening the permeability of new drugs.
Collapse
Affiliation(s)
- Adrián García-Salvador
- 73049GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Zamudio, Bizkaia, Spain
| | - Alazne Domínguez-Monedero
- 73049GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Zamudio, Bizkaia, Spain
| | - Paloma Gómez-Fernández
- 73049GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Zamudio, Bizkaia, Spain
| | - Amaia García-Bilbao
- 73049GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Zamudio, Bizkaia, Spain
| | - Susana Carregal-Romero
- Molecular and Functional Biomarkers Group, 90216CIC biomaGUNE (BRTA), Donostia-San Sebastián, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Joaquín Castilla
- 73038CIC bioGUNE (BRTA), Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Felipe Goñi-de-Cerio
- 73049GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), Zamudio, Bizkaia, Spain
| |
Collapse
|
7
|
In Vitro Cell Models of the Human Blood-Brain Barrier: Demonstrating the Beneficial Influence of Shear Stress on Brain Microvascular Endothelial Cell Phenotype. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-1-4939-8946-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
|
8
|
Stevenson ME, Lensmire NA, Swain RA. Astrocytes and radial glia-like cells, but not neurons, display a nonapoptotic increase in caspase-3 expression following exercise. Brain Behav 2018; 8:e01110. [PMID: 30240148 PMCID: PMC6192401 DOI: 10.1002/brb3.1110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/01/2018] [Revised: 06/18/2018] [Accepted: 08/05/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Exercise induces plasticity in the hippocampus, which includes increases in neurogenesis, the proliferation of new neurons, and angiogenesis, the sprouting of new capillaries from preexisting blood vessels. Following exercise, astrocytes also undergo morphological changes that parallel the events occurring in the neurovascular system. Interestingly, there have also been reports of apoptosis in the hippocampus following aerobic exercise. This experiment aimed to identify which population of hippocampal cells undergoes apoptosis after an acute bout of exercise. METHODS Cleaved caspase-3, a terminal protein in the apoptotic cascade, was initially used to identify apoptotic cells in the hippocampus after rats completed an acute bout of exercise. Next, the proportion of immature neurons, adult neurons, astrocytes, or radial glia-like cells expressing cleaved caspase-3 was quantified. TUNEL staining was completed as a second measure of apoptosis. RESULTS Following exercise, cleaved caspase-3 expression was increased in the CA1 and DG regions of the hippocampus. Cleaved caspase-3 was not highly expressed in neuronal populations, and expression was not increased in these cells postexercise. Instead, cleaved caspase-3 was predominantly expressed in astrocytes. Following exercise, there was an increased number of cleaved caspase-3 positive astrocytes in DG and CA1, and cleaved caspase-3 positive radial glia-like cells located in the subgranular zone. To determine whether cleaved caspase-3 expression in these glial cells was associated with apoptosis, a TUNEL assay was completed. TUNEL staining was negligible in all groups and did not mirror the pattern of caspase-3 labeling. CONCLUSIONS Cleaved caspase-3 expression was detected largely in non-neuronal cell populations, and the pattern of cleaved caspase-3 expression did not match that of TUNEL. This suggests that after exercise, cleaved caspase-3 expression may serve a nonapoptotic role in these hippocampal astrocytes and radial glia-like cells. It will be important to identify the function of exercise-induced cleaved caspase-3 expression in the future experiments.
Collapse
Affiliation(s)
| | - Nicole A. Lensmire
- Department of PsychologyUniversity of Wisconsin‐MilwaukeeMilwaukeeWisconsin
| | - Rodney A. Swain
- Department of PsychologyUniversity of Wisconsin‐MilwaukeeMilwaukeeWisconsin
| |
Collapse
|
9
|
Harder DR, Rarick KR, Gebremedhin D, Cohen SS. Regulation of Cerebral Blood Flow: Response to Cytochrome P450 Lipid Metabolites. Compr Physiol 2018; 8:801-821. [PMID: 29687906 DOI: 10.1002/cphy.c170025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/07/2023]
Abstract
There have been numerous reviews related to the cerebral circulation. Most of these reviews are similar in many ways. In the present review, we thought it important to provide an overview of function with specific attention to details of cerebral arterial control related to brain homeostasis, maintenance of neuronal energy demands, and a unique perspective related to the role of astrocytes. A coming review in this series will discuss cerebral vascular development and unique properties of the neonatal circulation and developing brain, thus, many aspects of development are missing here. Similarly, a review of the response of the brain and cerebral circulation to heat stress has recently appeared in this series (8). By trying to make this review unique, some obvious topics were not discussed in lieu of others, which are from recent and provocative research such as endothelium-derived hyperpolarizing factor, circadian regulation of proteins effecting cerebral blood flow, and unique properties of the neurovascular unit. © 2018 American Physiological Society. Compr Physiol 8:801-821, 2018.
Collapse
Affiliation(s)
- David R Harder
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Clement J. Zablocki VA Medical Center, Milwaukee, Wisconsin, USA
| | - Kevin R Rarick
- Department of Pediatrics, Division of Critical Care, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Debebe Gebremedhin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Susan S Cohen
- Department of Pediatrics, Division of Neonatology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
10
|
|
11
|
Saadoun S, Papadopoulos MC. Role of membrane complement regulators in neuromyelitis optica. Mult Scler 2015; 21:1644-54. [PMID: 25698168 DOI: 10.1177/1352458515571446] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/16/2014] [Accepted: 01/13/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND It is unclear why AQP4-IgG primarily causes central nervous system lesions by activating complement, but generally spares peripheral AQP4-expressing organs. OBJECTIVES To determine whether peripheral AQP4-expressing cells are protected from complement-mediated damage by expressing complement regulators. METHODS Human tissue and cultured human cells were immunostained for aquaporin-4 (AQP4), CD46, CD55 and CD59. We also determined the vulnerability to AQP4-IgG and complement-mediated damage of astrocytes cultured alone or co-cultured with endothelial cells. RESULTS In normal brain, astrocyte end-feet express AQP4, but are devoid of CD46, CD55 and CD59. Immunoreactivity for CD46, CD55 and CD59 is not increased in or around neuromyelitis optica lesions. In kidney AQP4 is co-expressed with CD46 and CD55, in stomach AQP4 is co-expressed with CD46 and in skeletal muscle AQP4 is co-expressed with CD46. Astrocytes cultured alone co-express AQP4 and CD59 but, in astrocyte-endothelial co-cultures, AQP4 is found in cell processes devoid of CD59. Astrocytes co-cultured with endothelial cells are more vulnerable to AQP4-IgG and complement-mediated lysis than astrocytes cultured alone. CONCLUSIONS Complement regulators protect peripheral organs, but not the central nervous system, from AQP4-IgG and complement-mediated damage. Our findings may explain why neuromyelitis optica primarily damages the central nervous system, but spares peripheral organs.
Collapse
|
12
|
Chaitanya GV, Minagar A, Alexander JS. Neuronal and astrocytic interactions modulate brain endothelial properties during metabolic stresses of in vitro cerebral ischemia. Cell Commun Signal 2014; 12:7. [PMID: 24438487 PMCID: PMC3927849 DOI: 10.1186/1478-811x-12-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/17/2013] [Accepted: 11/25/2013] [Indexed: 01/25/2023] Open
Abstract
Neurovascular and gliovascular interactions significantly affect endothelial phenotype. Physiologically, brain endothelium attains several of its properties by its intimate association with neurons and astrocytes. However, during cerebrovascular pathologies such as cerebral ischemia, the uncoupling of neurovascular and gliovascular units can result in several phenotypical changes in brain endothelium. The role of neurovascular and gliovascular uncoupling in modulating brain endothelial properties during cerebral ischemia is not clear. Specifically, the roles of metabolic stresses involved in cerebral ischemia, including aglycemia, hypoxia and combined aglycemia and hypoxia (oxygen glucose deprivation and re-oxygenation, OGDR) in modulating neurovascular and gliovascular interactions are not known. The complex intimate interactions in neurovascular and gliovascular units are highly difficult to recapitulate in vitro. However, in the present study, we used a 3D co-culture model of brain endothelium with neurons and astrocytes in vitro reflecting an intimate neurovascular and gliovascular interactions in vivo. While the cellular signaling interactions in neurovascular and gliovascular units in vivo are much more complex than the 3D co-culture models in vitro, we were still able to observe several important phenotypical changes in brain endothelial properties by metabolically stressed neurons and astrocytes including changes in barrier, lymphocyte adhesive properties, endothelial cell adhesion molecule expression and in vitro angiogenic potential.
Collapse
Affiliation(s)
| | | | - Jonathan S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health-Shreveport, Louisiana 71103, USA.
| |
Collapse
|
13
|
Cantrill CA, Skinner RA, Rothwell NJ, Penny JI. An immortalised astrocyte cell line maintains the in vivo phenotype of a primary porcine in vitro blood-brain barrier model. Brain Res 2012; 1479:17-30. [PMID: 22940232 DOI: 10.1016/j.brainres.2012.08.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/15/2011] [Revised: 08/01/2012] [Accepted: 08/16/2012] [Indexed: 12/11/2022]
Abstract
Whilst it is well documented that all components of the neurovascular unit contribute to the restrictive nature of the blood-brain barrier (BBB), astrocytes have been identified as the cellular component most likely to play an essential role in maintaining the barrier properties. The aim of this study was to examine the impact of the rat astrocyte cell line, CTX-TNA2, on the structural and functional characteristics of an in vitro BBB and determine the capacity of this astrocyte cell line to maintain the BBB phenotype. Co-culture of the CTX-TNA2 cells with primary porcine brain endothelial cells produced an in vitro BBB model which retains key features of the in vivo BBB. High transendothelial electrical resistances, comparable to those reported in vivo, were obtained. Ultrastructural analysis revealed distinct intercellular tight junction protein complexes and immunocytochemistry confirmed expression of the tight junction proteins ZO-1 and occludin. Western blotting and fluorescent tracer assays confirmed expression and functional activity of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) efflux transporters. Studies employing Alexa-fluor 555-conjugated human transferrin revealed temperature-sensitive internalisation indicating the BBB model retains functional receptor-mediated transferrin uptake. The findings of this study indicate that a robust BBB model has been produced and this is the first report of the inductive capacity of the CTX-TNA2 cell line. Since this in vitro BBB model possesses many key characteristics of the BBB in vivo it has the potential to be a valuable tool for the study of biochemical and physiological processes associated with the BBB.
Collapse
Affiliation(s)
- Carina A Cantrill
- School of Pharmacy and Pharmaceutical Sciences, Stopford Building, University of Manchester, Manchester M13 9PT, UK.
| | | | | | | |
Collapse
|
14
|
Kernt M, Liegl RG, Rueping J, Neubauer AS, Haritoglou C, Lackerbauer CA, Eibl KH, Ulbig MW, Kampik A. Sorafenib protects human optic nerve head astrocytes from light-induced overexpression of vascular endothelial growth factor, platelet-derived growth factor, and placenta growth factor. Growth Factors 2010; 28:211-20. [PMID: 20166888 DOI: 10.3109/08977191003604505] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Growth factors, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and placenta growth factor (PlGF) are key players in the development of diabetic retinopathy, age-related macular degeneration, and other retinal neovascular diseases. Glial cells provide a significant source of retinal growth factor production under physiologic and pathologic conditions. Cumulative light exposure has been linked to increased retinal growth factor expression. Previous reports indicate that sorafenib, an oral multikinase inhibitor, might have a beneficial effect on retinal neovascularization. This study was designed to investigate the effects of sorafenib on light-induced overexpression of growth factors in human retinal glial cells. METHODS Primary human optic nerve head astrocytes (ONHAs) were exposed to white light and incubated with sorafenib. Viability, expression, and secretion of VEGF-A, PDGF-BB, and PlGF and their mRNA were determined by reverse transcription-polymerase chain reaction, immunohistochemistry, and enzyme-linked immunosorbent assay. RESULTS Light exposure decreased cell viability and increased VEGF-A, PDGF-BB, and PlGF expression and secretion. These light-induced effects were significantly reduced when cells were treated with sorafenib at a concentration of 1 microg/ml. CONCLUSION Sorafenib significantly reduced light-induced overexpression of VEGF-A, PDGF-BB, and PlGF in primary human ONHAs. Sorafenib has promising properties as a potential supportive treatment for retinal neovascularization.
Collapse
Affiliation(s)
- M Kernt
- Department of Ophthalmology, Ludwig-Maximilians University, 80336 Munich, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Tian W, Kyriakides TR. Matrix metalloproteinase-9 deficiency leads to prolonged foreign body response in the brain associated with increased IL-1beta levels and leakage of the blood-brain barrier. Matrix Biol 2009; 28:148-59. [PMID: 19264129 DOI: 10.1016/j.matbio.2009.02.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/28/2008] [Revised: 02/16/2009] [Accepted: 02/23/2009] [Indexed: 11/27/2022]
Abstract
Matrix metalloproteinases (MMPs) are enzymes with specificity towards extracellular matrix (ECM) components. MMPs, especially MMP-9, have been shown to degrade components of the basal lamina and disrupt the blood-brain barrier (BBB) and thus, contribute to neuroinflammation. In the present study we examined the role of MMP-9 in the foreign body response in the brain. Millipore filters of mixed cellulose ester were implanted into the brain cortex of wild type and MMP-9-null mice for a period of 2 d to 8 wks and the response was analyzed by histology and immunohistochemistry. We observed enhanced and prolonged neuroinflammation in MMP-9-null mice, evidenced by persistence of neutrophils, macrophages/microglia, and reactive astrocytes up to 8 wks post-implantation. In addition, blood vessel density around implants was increased in MMP-9-null mice and detection of mouse serum albumin (MSA) indicated that vessels were leaky. Immunohistochemical and western blot analyses indicated that this defect was associated with the absence of tight junction proteins zonula occludens-1 (ZO-1) and ZO-2 from vessels in proximity to implants. Analysis of brain sections and brain protein extracts revealed that the levels of the pro-inflammatory cytokine interleukin-1beta (IL-1beta), which is a substrate for MMP-9, were significantly higher in MMP-9-null mice at 8wks post-implantation. Collectively, our studies suggest that increased levels of IL-1beta and the delayed repair of BBB are associated with prolongation of the FBR in MMP-9-null mice.
Collapse
Affiliation(s)
- Weiming Tian
- Vascular Biology and Therapeutics Program, Departments of Pathology and Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| | | |
Collapse
|
16
|
Abstract
Neurons have long held the spotlight as the central players of the nervous system, but we must remember that we have equal numbers of astrocytes and neurons in the brain. Are these cells only filling up the space and passively nurturing the neurons, or do they also contribute to information transfer and processing? After several years of intense research since the pioneer discovery of astrocytic calcium waves and glutamate release onto neurons in vitro, the neuronal-glial studies have answered many questions thanks to technological advances. However, the definitive in vivo role of astrocytes remains to be addressed. In addition, it is becoming clear that diverse populations of astrocytes coexist with different molecular identities and specialized functions adjusted to their microenvironment, but do they all belong to the umbrella family of astrocytes? One population of astrocytes takes on a new function by displaying both support cell and stem cell characteristics in the neurogenic niches. Here, we define characteristics that classify a cell as an astrocyte under physiological conditions. We will also discuss the well-established and emerging functions of astrocytes with an emphasis on their roles on neuronal activity and as neural stem cells in adult neurogenic zones.
Collapse
|
17
|
Park JA, Lee HS, Ko KJ, Park SY, Kim JH, Choe G, Kweon HS, Song HS, Ahn JC, Yu YS, Kim KW. Meteorin regulates angiogenesis at the gliovascular interface. Glia 2008; 56:247-58. [PMID: 18059000 DOI: 10.1002/glia.20600] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/08/2022]
Abstract
Brain microvasculature requires a coordinated interaction between endothelial cells and astrocytes at the gliovascular interface. However, the role of the factors involved in that interaction and expressed by these cells is poorly understood. In this study, we demonstrate that Meteorin is highly expressed in astrocytes of the brain and retina during the late embryonic and postnatal stages of mouse development. Most notably, Meteorin is localized to the astrocyte endfeet that surround the blood vessels. To investigate the role of Meteorin in perivascular astrocytes, we depleted endogenous levels of Meteorin in cultured astrocytes using siRNA, and found that Meteorin attenuates angiogenic activity indirectly via astrocyte-derived thrombospondin-1/-2 (TSP-1/-2). Exogenous treatment of astrocytes with Meteorin protein also promotes astrocyte expression and secretion of TSP-1/-2. The conditioned media from the Meteorin-treated astrocytes attenuated angiogenic activity of microvascular endothelial cells. This activity was reversed by inhibiting the binding of TSP-1/-2 to its receptor. Furthermore, we found that TSP-1/-2 was co-localized with Meteorin in the developing brain. Therefore, our data strongly suggests that Meteorin is expressed and secreted by perivascular astrocytes and the secreted protein upregulates TSP-1/-2 to attenuate angiogenesis in the surrounding endothelial cells and to promote vascular maturation.
Collapse
Affiliation(s)
- Jeong Ae Park
- Department of Marine Biotechnology, College of Liberal Arts and Sciences, Anyang University, Incheon 417-833, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zozulya A, Weidenfeller C, Galla HJ. Pericyte–endothelial cell interaction increases MMP-9 secretion at the blood–brain barrier in vitro. Brain Res 2008; 1189:1-11. [DOI: 10.1016/j.brainres.2007.10.099] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/30/2007] [Revised: 10/26/2007] [Accepted: 10/26/2007] [Indexed: 10/22/2022]
|
19
|
Crivellato E, Nico B, Ribatti D. Contribution of endothelial cells to organogenesis: a modern reappraisal of an old Aristotelian concept. J Anat 2007; 211:415-27. [PMID: 17683480 PMCID: PMC2375830 DOI: 10.1111/j.1469-7580.2007.00790.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 04/27/2007] [Indexed: 01/02/2023] Open
Abstract
It is well established that many tissue-derived factors are involved in blood vessel formation, but evidence is now emerging that endothelial cells themselves represent a crucial source of instructive signals to non-vascular tissue cells during organ development. Thus, endothelial cell signalling is currently believed to promote fundamental cues for cell fate specification, embryo patterning, organ differentiation and postnatal tissue remodelling. This review article summarizes some of the recent advances in our understanding of the role of endothelial cells as effector cells in organ formation.
Collapse
Affiliation(s)
- E Crivellato
- Department of Medical and Morphological Research, Anatomy Section, University of Udine, Italy.
| | | | | |
Collapse
|
20
|
Sehara Y, Hayashi T, Deguchi K, Zhang H, Tsuchiya A, Yamashita T, Lukic V, Nagai M, Kamiya T, Abe K. Potentiation of neurogenesis and angiogenesis by G-CSF after focal cerebral ischemia in rats. Brain Res 2007; 1151:142-9. [PMID: 17459352 DOI: 10.1016/j.brainres.2007.01.149] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2006] [Revised: 01/09/2007] [Accepted: 01/10/2007] [Indexed: 01/03/2023]
Abstract
Recently, granulocyte colony-stimulating factor (G-CSF) is expected to demonstrate beneficial effects on cerebral ischemia. Here, we showed the potential benefit of G-CSF administration after transient middle cerebral artery occlusion (tMCAO). Adult male Wistar rats received vehicle or G-CSF (50 microg/kg) subcutaneously after reperfusion, and were treated with 5-bromodeoxyuridine (BrdU, 50 mg/kg) once daily by the intraperitoneal route for 3 days after tMCAO. Nissl-stained sections at 7 days after tMCAO showed significant reduction of the infarction area (31%, P<0.01). At 7 days after tMCAO, BrdU plus NeuN double-positive cells increased by 43.3% in the G-CSF-treated group (P<0.05), and BrdU-positive endothelial cells were increased 2.29 times in the G-CSF-treated group, to a level as high as that in the vehicle-treated group (P<0.01), in the periischemic area. Our results indicate that G-CSF caused potentiation of neuroprotection and neurogenesis and is expected to have practical therapeutic potential in treating individuals after ischemic brain injury.
Collapse
Affiliation(s)
- Yoshihide Sehara
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Siddharthan V, V. Kim Y, Liu S, Kim KS. Human astrocytes/astrocyte-conditioned medium and shear stress enhance the barrier properties of human brain microvascular endothelial cells. Brain Res 2007; 1147:39-50. [PMID: 17368578 PMCID: PMC2691862 DOI: 10.1016/j.brainres.2007.02.029] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/23/2005] [Revised: 01/12/2007] [Accepted: 02/02/2007] [Indexed: 11/20/2022]
Abstract
The blood-brain barrier (BBB) is a structural and functional barrier that regulates the passage of molecules into and out of the brain to maintain the neural microenvironment. We have previously developed the in vitro BBB model with human brain microvascular endothelial cells (HBMEC). However, in vivo HBMEC are shown to interact with astrocytes and also exposed to shear stress through blood flow. In an attempt to develop the BBB model to mimic the in vivo condition we constructed the flow-based in vitro BBB model using HBMEC and human fetal astrocytes (HFA). We also examined the effect of astrocyte-conditioned medium (ACM) in lieu of HFA to study the role of secreted factor(s) on the BBB properties. The tightness of HBMEC monolayer was assessed by the permeability of dextran and propidium iodide as well as by measuring the transendothelial electrical resistance (TEER). We showed that the HBMEC permeability was reduced and TEER was increased by non-contact, co-cultivation with HFA and ACM. The exposure of HBMEC to shear stress also exhibited decreased permeability. Moreover, HFA/ACM and shear flow exhibited additive effect of decreasing the permeability of HBMEC monolayer. In addition, we showed that the HBMEC expression of ZO-1 (tight junction protein) was increased by co-cultivation with ACM and in response to shear stress. These findings suggest that the non-contact co-cultivation with HFA helps maintain the barrier properties of HBMEC by secreting factor(s) into the medium. Our in vitro flow model system with the cells of human origin should be useful for studying the interactions between endothelial cells, glial cells, and secreted factor(s) as well as the role of shear stress in the barrier property of HBMEC.
Collapse
Affiliation(s)
- Venkatraman Siddharthan
- Division of Pediatric Infectious Diseases, Johns Hopkins University, School of Medicine, Baltimore, MD 21287
| | - Yuri V. Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University, School of Medicine, Baltimore, MD 21287
| | - Suyi Liu
- World Precision Instruments Inc., 175 Sarasota Center Blvd, Sarasota FL 34240 U.S.A
| | - Kwang Sik Kim
- Correspondence: Prof. Kwang Sik Kim, Division of Pediatric Infectious Diseases, Johns Hopkins University, School of Medicine, 600 North Wolfe Street, Park 256, Baltimore, MD 21287. , Phone: 410-614-3917, Fax: 410-614-1491
| |
Collapse
|
22
|
Abstract
The brain is critically dependent on oxygen and glucose supply for normal function. Various neurovascular control mechanisms assure that the blood supply of the brain is adequate to meet the energy needs of its components. Emerging evidence shows that neuronal activity can control microcirculation using astrocytes as a mediator. Astrocytes can sense neuronal activity and are involved in signal transmission. Synaptic activity triggers an increase in the intracellular calcium concentration [Ca(2+)]i of adjacent astrocytes, stimulating the release of adenosine triphosphate (ATP) and glutamate. The released ATP mediates the propagation of Ca(2+) waves between neighboring astrocytes, thereby recruiting them to mediate adequate cerebrovascular response to neuronal activation. Simultaneously, sodium-dependent glutamate uptake in astrocytes generates Na(+) waves and subsequently increases glucose uptake and metabolism that leads to the formation of lactate, which is then delivered to neurons as an energy substrate. Further, astrocytic Ca(2+) elevations can lead to secretion of vasodilatory substances from perivascular endfeet, such as epoxyeicosatrienoic acid (EETs), adenosine, nitric oxide (NO), and cyclooxygenase-2 (COX-2) metabolites, resulting in increased local blood flow. Thus, astrocytes by releasing vasoactive molecules mediate the neuron-astrocyte-endothelial signaling pathway and play a profound role in coupling blood flow to neuronal activity.
Collapse
Affiliation(s)
- Danica Jakovcevic
- Department of Physiology, Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | |
Collapse
|
23
|
Goldshmit Y, Galea MP, Bartlett PF, Turnley AM. EphA4 regulates central nervous system vascular formation. J Comp Neurol 2006; 497:864-75. [PMID: 16802330 DOI: 10.1002/cne.21029] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022]
Abstract
Molecules involved in axon guidance have recently also been shown to play a role in blood vessel guidance. To examine whether axon guidance molecules, such as the EphA4 receptor tyrosine kinase, might also play a role in development of the central nervous system (CNS) vasculature and repair following CNS injury, we examined wild-type and EphA4 null mutant (-/-) mice. EphA4-/- mice exhibited an abnormal CNS vascular structure in both the cerebral cortex and the spinal cord, with disorganized branching and a 30% smaller diameter. During development, EphA4 was expressed on endothelial cells. This pattern of expression was not maintained in the adult. After spinal cord injury in wild-type mice, expression of EphA4 was markedly up-regulated on activated astrocytes, many of which were tightly associated with blood vessels. In EphA4-/- spinal cord following injury, astrocytes were not as tightly associated with blood vessels as the wild-type astrocytes. In uninjured EphA4-/- mice, the blood-brain barrier (BBB) appeared normal, but it showed prolonged leakage following spinal cord injury. These results support a role for EphA4 in CNS vascular formation and guidance during development and an additional role in BBB repair.
Collapse
Affiliation(s)
- Yona Goldshmit
- Centre for Neuroscience, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | | | | | | |
Collapse
|
24
|
Petros TJ, Williams SE, Mason CA. Temporal regulation of EphA4 in astroglia during murine retinal and optic nerve development. Mol Cell Neurosci 2006; 32:49-66. [PMID: 16574431 DOI: 10.1016/j.mcn.2006.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/07/2005] [Revised: 02/06/2006] [Accepted: 02/16/2006] [Indexed: 01/03/2023] Open
Abstract
Eph receptors and their ephrin ligands play important roles in many aspects of visual system development. In this study, we characterized the spatial and temporal expression pattern of EphA4 in astrocyte precursor cell (APC) and astrocyte populations in the murine retina and optic nerve. EphA4 is expressed by immotile optic disc astrocyte precursor cells (ODAPS), but EphA4 is downregulated as these cells migrate into the retina. Surprisingly, mature astrocytes in the adult retina re-express EphA4. Within the optic nerve, EphA4 is expressed in specialized astrocytes that form a meshwork at the optic nerve head (ONH). Our in vitro and in vivo data indicate that EphA4 is dispensable for retinal ganglion cell (RGC) axon growth and projections through the chiasm. While optic stalk structure, APC proliferation and migration, retinal vascularization, and oligodendrocyte migration appear normal in EphA4 mutants, the expression of EphA4 in APCs and in the astrocyte meshwork at the ONH has implications for optic nerve pathologies.
Collapse
Affiliation(s)
- Timothy J Petros
- Center for Neurobiology and Behavior, Department of Pathology, Columbia University, College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | | | | |
Collapse
|
25
|
Li J, Ding YH, Rafols JA, Lai Q, McAllister JP, Ding Y. Increased astrocyte proliferation in rats after running exercise. Neurosci Lett 2005; 386:160-4. [PMID: 16024173 DOI: 10.1016/j.neulet.2005.06.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/31/2005] [Revised: 05/27/2005] [Accepted: 06/01/2005] [Indexed: 11/15/2022]
Abstract
The aim in this study was to investigate whether physical exercise could induce astroglial proliferation in the frontoparietal cortex and dorsolateral striatum where extensive angiogenesis had been found after exercise in previous studies. Adult male Sprague Dawley rats (n=48) were used in four experimental groups. Animals were exercised 30 min each day on a treadmill on which repetitive locomotor movement was required, for 0 (n=12), 3 (n=12) or 6 (n=12) weeks, as well as 3-week exercise plus 3-week rest (n=12). Brain tissues of the exercised and non-exercised rats were processed for glial fibrillary acidic protein (GFAP) immunocytochemistry (n=6 x 4) and Western blotting (n=6 x 4) to evaluate regional astrocyte proliferation in the frontoparietal cortex and dorsolateral striatum. By using GFAP immunocytochemistry and stereological methods, we compared the density of astrocytes in the animals with or without exercise. In comparison to non-exercised animals, a significant (p<0.01) increase in the number of astrocytes was observed in both cortex and striatum of rats exercised for 3 or 6 weeks. Our data also indicated that astrocytic density continued to increase up to 6 weeks either with an additional 3 weeks of exercise (p<0.01) or 3 weeks of rest (p<0.01). In addition, Western blotting analysis showed an obvious increase in GFAP protein from cortex and striatum of exercised animals. Astrocytosis after exercise, coupled with angiogenesis, is thought to provide strength to the neurovascular unit (a construct consisting of microvascular endothelium, astroglia, neurons and the extracellular matrix). Strengthening of this unit by exercise may protect blood-brain-barrier function following brain injury, such as that occurring after stroke.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurological Surgery, Wayne State University School of Medicine, Canfield, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
26
|
Yamamoto N, Hirabayashi Y, Amari M, Yamaguchi H, Romanov G, Van Nostrand WE, Yanagisawa K. Assembly of hereditary amyloid beta-protein variants in the presence of favorable gangliosides. FEBS Lett 2005; 579:2185-90. [PMID: 15811339 DOI: 10.1016/j.febslet.2005.03.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/08/2005] [Revised: 03/06/2005] [Accepted: 03/07/2005] [Indexed: 10/25/2022]
Abstract
The mechanisms underlying regional amyloid beta-protein (Abeta) deposition in brain remain unclear. Here we show that assembly of hereditary variant Dutch- and Italian-type Abetas, and Flemish-type Abeta was accelerated by GM3 ganglioside, and GD3 ganglioside, respectively. Notably, cerebrovascular smooth muscle cells, which compose the cerebral vessel wall at which the Dutch- and Italian-type Abetas deposit, exclusively express GM3 whereas GD3 is upregulated in the co-culture of endothelial cells and astrocytes, which forms the cerebrovascular basement membrane, the site of Flemish-type Abeta deposition. Our results suggest that regional Abeta deposition is induced by the local gangliosides in the brain.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Department of Alzheimer's Disease Research, National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, 36-3 Gengo, Morioka, Obu 474-8522, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhang C, Gehlbach P, Gongora C, Cano M, Fariss R, Hose S, Nath A, Green WR, Goldberg MF, Zigler JS, Sinha D. A potential role for β- and γ-crystallins in the vascular remodeling of the eye. Dev Dyn 2005; 234:36-47. [PMID: 16003775 DOI: 10.1002/dvdy.20494] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/09/2023] Open
Abstract
We demonstrate that expression of beta- and gamma-crystallins is associated with intraocular vessels during normal vascular development of the eye and also in the Nuc1 rat, a mutant in which the hyaloid vascular system fails to regress normally. Real-Time RT PCR, Western blot and metabolic labeling studies indicate an increased expression of beta- and gamma-crystallins in Nuc1 retina. The increased expression of crystallins was localized to the astrocytes surrounding the intraocular vessels. A similar pattern of crystallin expression was also observed in the retinal vessels during normal development. Cultured human astrocytes exposed to 3-nitropropionic acid, an established model of neuronal hypoxia, increased VEGF expression, as expected, but also increased expression of crystallins. Our data suggest that crystallins may function together with VEGF during vascular remodeling. Interestingly, in human PFV (persistent fetal vasculature) disease, where the hyaloid vasculature abnormally persists after birth, we show that astrocytes express both VEGF and crystallins.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Ophthalmology, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Garcia CM, Darland DC, Massingham LJ, D'Amore PA. Endothelial cell-astrocyte interactions and TGF beta are required for induction of blood-neural barrier properties. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 152:25-38. [PMID: 15283992 DOI: 10.1016/j.devbrainres.2004.05.008] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Accepted: 05/16/2004] [Indexed: 11/25/2022]
Abstract
We sought to establish a blood-neural barrier (BNB) model of astrocyte contact with endothelial cells (EC) to test the hypothesis that transforming growth factor beta (TGF beta) promotes an EC barrier-phenotype. Astrocyte-EC contact induced BNB properties in EC. Transendothelial resistance was augmented by direct contact between astrocytes-EC, but not by astrocyte-conditioned medium or astrocyte-EC coculture conditioned medium. Coculture of EC and astrocytes led to significant increase in endothelial occludin levels and junctional localization. EC gamma-glutamyl-transferase (GGT) activity was increased by direct contact with astrocytes, by conditioned medium from cocultures or by TGF beta1. Coculture inhibited EC proliferation with no effect on astrocyte proliferation. A neutralizing antibody to TGF beta decreased GGT activity in cocultures and increased cell number. Whereas total TGF beta was not significantly altered by coculture, activated TGF beta increased in astrocyte-EC cocultures. In summary, astrocyte-EC contact induces BNB characteristics in EC and locally activated TGF beta is responsible for part of the induction.
Collapse
Affiliation(s)
- Claudia M Garcia
- Biological and Biomedical Sciences Graduate Program, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
29
|
Virgintino D, Errede M, Robertson D, Capobianco C, Girolamo F, Vimercati A, Bertossi M, Roncali L. Immunolocalization of tight junction proteins in the adult and developing human brain. Histochem Cell Biol 2004. [PMID: 15221411 DOI: 10.1007/s00418‐004‐0665‐1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
The formation of endothelial tight junctions (TJs) is crucial in blood-brain barrier (BBB) differentiation, and the expression and targeting of TJ-associated proteins mark the beginning of BBB functions. Using confocal microscopy, this study analyzed endothelial TJs in adult human cerebral cortex and the fetal telencephalon and leptomeninges in order to compare the localization of two TJ-associated transmembrane proteins, occludin and claudin-5. In the arterioles and microvessels of adult brain, occludin and claudin-5 form continuous bands of endothelial immunoreactivity. During fetal development, occludin and claudin-5 immunoreactivity is first detected as a diffuse labeling of endothelial cytoplasm. Later, at 14 weeks, the immunosignal for both proteins shifts from the cytoplasm to the interface of adjacent endothelial cells, forming a linear, widely discontinuous pattern of immunoreactivity that achieves an adult-like appearance within a few weeks. These results demonstrate that occludin and claudin-5 expression is an early event in human brain development, followed shortly by assembly of both proteins at the junctional areas. This incremental process suggests more rapid establishment of the human BBB, consistent with its specific function of creating a suitable environment for neuron differentiation and neurite outgrowth during neocortical histogenesis.
Collapse
Affiliation(s)
- Daniela Virgintino
- Department of Human Anatomy and Histology, University of Bari School of Medicine, Bari, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Virgintino D, Errede M, Robertson D, Capobianco C, Girolamo F, Vimercati A, Bertossi M, Roncali L. Immunolocalization of tight junction proteins in the adult and developing human brain. Histochem Cell Biol 2004; 122:51-9. [PMID: 15221411 DOI: 10.1007/s00418-004-0665-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 05/24/2004] [Indexed: 01/16/2023]
Abstract
The formation of endothelial tight junctions (TJs) is crucial in blood-brain barrier (BBB) differentiation, and the expression and targeting of TJ-associated proteins mark the beginning of BBB functions. Using confocal microscopy, this study analyzed endothelial TJs in adult human cerebral cortex and the fetal telencephalon and leptomeninges in order to compare the localization of two TJ-associated transmembrane proteins, occludin and claudin-5. In the arterioles and microvessels of adult brain, occludin and claudin-5 form continuous bands of endothelial immunoreactivity. During fetal development, occludin and claudin-5 immunoreactivity is first detected as a diffuse labeling of endothelial cytoplasm. Later, at 14 weeks, the immunosignal for both proteins shifts from the cytoplasm to the interface of adjacent endothelial cells, forming a linear, widely discontinuous pattern of immunoreactivity that achieves an adult-like appearance within a few weeks. These results demonstrate that occludin and claudin-5 expression is an early event in human brain development, followed shortly by assembly of both proteins at the junctional areas. This incremental process suggests more rapid establishment of the human BBB, consistent with its specific function of creating a suitable environment for neuron differentiation and neurite outgrowth during neocortical histogenesis.
Collapse
Affiliation(s)
- Daniela Virgintino
- Department of Human Anatomy and Histology, University of Bari School of Medicine, Bari, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Dutt K, Sanford G, Harris-Hooker S, Brako L, Kumar R, Sroufe A, Melhado C. Three-dimensional model of angiogenesis: coculture of human retinal cells with bovine aortic endothelial cells in the NASA bioreactor. ACTA ACUST UNITED AC 2004; 9:893-908. [PMID: 14633374 DOI: 10.1089/107632703322495547] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022]
Abstract
Ocular angiogenesis is the leading cause of blindness and is associated with diabetic retinopathy and age-related macular degeneration. We describe, in this report, our preliminary studies using a horizontally rotating bioreactor (HRB), developed by the National Aeronautics and Space Administration (NASA), to explore growth and differentiation-associated events in the early phase of ocular angiogenesis. Human retinal (HRet) cells and bovine endothelial cells (ECs) were cocultured on laminin-coated Cytodex-3 microcarrier beads in an HRB for 1-36 days. Endothelial cells grown alone in the HRB remained cuboidal and were well differentiated. However, when HRet cells were cocultured with ECs, cordlike structures formed as early as 18-36 h and were positive for von Willebrand factor. In addition to the formation of cords and capillary-like structures, ECs showed the beginning of sprouts. The HRB seems not only to promote accelerated capillary formation, but also to enhance differentiation of retinal precursor cells. This leads to the formation of rosette-like structures (which may be aggregates of photoreceptors that were positive for rhodopsin). Upregulation of vascular endothelial growth factor and basic fibroblast growth factor was seen in retinal cells grown in the HRB as compared with monolayers and could be one of the factors responsible for accelerated capillary formation. Hence, the HRB promotes three-dimensional assembly and differentiation, possibly through promoting cell-to-cell interaction and/or secretion of growth and differentiation factors.
Collapse
Affiliation(s)
- Kamla Dutt
- Department of Pathology, Morehouse School of Medicine, Atlanta, Georgia 30310-1495, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Takuma K, Baba A, Matsuda T. Astrocyte apoptosis: implications for neuroprotection. Prog Neurobiol 2004; 72:111-27. [PMID: 15063528 DOI: 10.1016/j.pneurobio.2004.02.001] [Citation(s) in RCA: 344] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2003] [Accepted: 02/04/2004] [Indexed: 12/21/2022]
Abstract
Astrocytes, the most abundant glial cell types in the brain, provide metabolic and trophic support to neurons and modulate synaptic activity. Accordingly, impairment in these astrocyte functions can critically influence neuronal survival. Recent studies show that astrocyte apoptosis may contribute to pathogenesis of many acute and chronic neurodegenerative disorders, such as cerebral ischemia, Alzheimer's disease and Parkinson's disease. We found that incubation of cultured rat astrocytes in a Ca(2+)-containing medium after exposure to a Ca(2+)-free medium causes an increase in intracellular Ca(2+) concentration followed by apoptosis, and that NF-kappa B, reactive oxygen species, and enzymes such as calpain, xanthine oxidase, calcineurin and caspase-3 are involved in reperfusion-induced apoptosis. Furthermore, we demonstrated that heat shock protein, mitogen-activated protein/extracellular signal-regulated kinase, phosphatidylinositol-3 kinase and cyclic GMP phosphodiesterase are target molecules for anti-apoptotic drugs. This review summarizes (1) astrocytic functions in neuroprotection, (2) current evidence of astrocyte apoptosis in both in vitro and in vivo studies including its molecular pathways such as Ca(2+) overload, oxidative stress, NF-kappa B activation, mitochondrial dysfunction, endoplasmic reticulum stress, and protease activation, and (3) several drugs preventing astrocyte apoptosis. As a whole, this article provides new insights into the potential role of astrocytes as targets for neuroprotection. In addition, the advance in the knowledge of molecular mechanisms of astrocyte apoptosis may lead to the development of novel therapeutic strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Kazuhiro Takuma
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences and High Technology Research Center, Kobe Gakuin University, Kobe 651-2180, Japan
| | | | | |
Collapse
|
33
|
Hossain MA, Russell JC, Miknyoczki S, Ruggeri B, Lal B, Laterra J. Vascular endothelial growth factor mediates vasogenic edema in acute lead encephalopathy. Ann Neurol 2004; 55:660-7. [PMID: 15122706 DOI: 10.1002/ana.20065] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/08/2022]
Abstract
Brain injury from inorganic Pb(2+) is considered the most important environmental childhood health hazard worldwide. The microvasculature of the developing brain is uniquely susceptible to high level Pb(2+) toxicity (ie, Pb(2+) encephalopathy) characterized by cerebellar hemorrhage, increased blood-brain barrier permeability, and vasogenic edema. However, the specific molecular mediators of Pb(2+) encephalopathy have been elusive. We found that Pb(2+) induces vascular endothelial growth factor/vascular permeability factor (VEGF) in cultured astrocytes (J Biol Chem, 2000;275:27874-27882). The study presented here asks if VEGF dysregulation contributes mechanistically to Pb(2+) encephalopathy. Neonatal rats exposed to 4% Pb-carbonate develop the histopathological features of Pb(2+) encephalopathy seen in children. Cerebellar VEGF expression increased approximately twofold (p < 0.01) concurrent with the development of cerebellar microvascular hemorrhage, enhanced vascular permeability to serum albumin, and vasogenic cerebellar edema (p < 0.01). No change in VEGF expression occurred in cerebral cortex that does not develop these histopathological complications of acute Pb(2+) intoxication. Pb(2+) exposure increased phosphorylation of cerebellar Flk-1 VEGF receptors and the Flk-1 inhibitor CEP-3967 completely blocked cerebellar edema formation without affecting microhemorrhage formation or blood-brain barrier permeability. This establishes that Pb(2+)-induced vasogenic edema formation develops via a Flk-1-dependent mechanism and suggests that the vascular permeability caused by Pb(2+) is Flk-1 independent.
Collapse
Affiliation(s)
- Mir Ahamed Hossain
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
During the last decades a lot of attention has been focussed on mechanisms of glioma vascularization, particularly in terms of investigating vascular growth factors and receptors. Recently, these efforts resulted in various approaches for antiangiogenic treatment strategies using in vitro cell culture systems as well as experimental orthotopic and non-orthotopic brain tumors. These basic science and preclinical trials need an assortment of models, which should allow investigating a variety of questions. Several objectives concerning basic endothelial cell (EC) characteristics can adequately be studied in vitro using EC monolayer assays. Three-dimensional spheroid techniques respect the more complex cell-cell and cell-environment interplay within a 3-dimensional culture. Recent advances in molecular genetic techniques offer a wide access to the genome of EC. Using these micro array or chip methods differences between micro- and macromolecular EC as well as variations within the gene pool of different organ specific EC can be assessed. To optimize the imitation of the crucial interaction of human gliomas with host endothelial cells, immunological cells and extracellular matrix, animal models are mandatory. An essential rule is to utilize an orthotopic model, since tumor-host-interaction is organ specific. To avoid alloimmunogenic responses, it is desirable to use weak or non-immunogenic glioma grafts, which is best accomplished in a syngeneic model. However, since rat gliomas poorly resemble human glioma growth patterns, human glioma xenografting into immunocompromized animals should be considered. In vivo-monitoring techniques like videoscopy via a cranial window or magnetic resonance imaging (MRI) allow for functional studies and improve the validity of the model employed. Finally, it is essentially to recognize the limitations of each model considered and to select that model which seems to be most appropriate for the objectives to be investigated.
Collapse
Affiliation(s)
- Roland H Goldbrunner
- Department of Neurosurgery, Grosshadern Hospital, Ludwig-Maximilians, University of Munich, 81377 Munich, Germany
| | | | | |
Collapse
|
35
|
Abstract
OBJECTIVE To review recent advances in the field of endothelial cell heterogeneity, and to apply this knowledge to an understanding of site-specific vasculopathy, including acute lung injury. DATA SOURCES AND STUDY SELECTION Published research and review articles in the English language related to endothelial cell biology and endothelial cell heterogeneity. DATA EXTRACTION AND SYNTHESIS The results of published studies have been used to provide a perspective of endothelial cell phenotypes in health and disease. CONCLUSIONS The structure and function of endothelial cells are differentially regulated in space and time. Far from being a giant monopoly of homogeneous cells, the endothelium represents a consortium of smaller enterprises of cells located within blood vessels of different tissues. Although united in certain functions, each enterprise is uniquely adapted to meet the demands of the underlying tissue. The endothelium may also vary in its response to pathophysiologic stimuli and therefore contribute to the focal nature of vasculopathic disease states. In acute lung injury, the unique properties of the endothelium may conspire with systemic imbalances to localize pathology to the pulmonary vasculature.
Collapse
Affiliation(s)
- William C Aird
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| |
Collapse
|
36
|
Zhang C, Harder DR. Cerebral capillary endothelial cell mitogenesis and morphogenesis induced by astrocytic epoxyeicosatrienoic Acid. Stroke 2002; 33:2957-64. [PMID: 12468797 DOI: 10.1161/01.str.0000037787.07479.9a] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Background and Purpose- Epoxyeicosatrienoic acids (EETs) are products of cytochrome P450 epoxygenation of arachidonic acid. We have previously demonstrated that astrocyte-conditioned medium induced mitogenesis in brain capillary endothelial cells. The goals of the present studies are to further define the mechanism through which this can occur and to confirm that EETs are derived from astrocytes, through which astrocytic activity can regulate cerebral angiogenesis in response to neuronal activation. METHODS Astrocytes and cerebral capillary endothelial cells in primary cultures were cocultured to examine the interaction of the 2 cell types. We used multiple immunohistochemical techniques to characterize the multicellular nature of the capillaries, which is not simply an artifact related to the culture conditions. The mitogenic effect of EETs was determined by (3)H-thymidine incorporation and cell proliferation assay. Endothelial tube formation was examined in vitro and in vivo with the use of a reconstituted basement membrane (Matrigel) assay. RESULTS In cocultures of astrocytes and capillary endothelium, we observed morphological changes in both cell types such that each assumed certain physiological characteristics, ie, endothelial networks and astrocytes with "footlike" projections as well as intermittent gap junctions forming within the endothelial cells. EETs from astrocytes as well as synthetic EETs promoted mitogenesis of endothelial cells, a process sensitive to inhibition of tyrosine kinase with genistein. Treatments with exogenous EETs were sufficient for endothelial cells to differentiate into capillary-like structures in culture as well as in vivo in a Matrigel matrix. CONCLUSIONS The 2 major conclusions from these data are that astrocytes may play an important role in regulating angiogenesis in the brain and that cytochrome P450-derived EETs from astrocytes are mitogenic and angiogenic.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/metabolism
- 8,11,14-Eicosatrienoic Acid/pharmacology
- Animals
- Astrocytes/cytology
- Astrocytes/metabolism
- Brain/blood supply
- Capillaries/cytology
- Cell Differentiation/drug effects
- Cells, Cultured
- Coculture Techniques
- Culture Media, Conditioned/pharmacology
- Cytochrome P-450 Enzyme Inhibitors
- Cytochrome P-450 Enzyme System/metabolism
- Dose-Response Relationship, Drug
- Endothelial Growth Factors/pharmacology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Enzyme Inhibitors/pharmacology
- Intercellular Signaling Peptides and Proteins/pharmacology
- Lymphokines/pharmacology
- Mitosis/drug effects
- Neovascularization, Physiologic/drug effects
- Rats
- Thymidine/metabolism
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- Chenyang Zhang
- Cardiovascular Research Center, Department of Physiology, Medical College of Wisconsin, Milwaukee, USA
| | | |
Collapse
|
37
|
Velazquez OC, Snyder R, Liu ZJ, Fairman RM, Herlyn M. Fibroblast-dependent differentiation of human microvascular endothelial cells into capillary-like 3-dimensional networks. FASEB J 2002; 16:1316-8. [PMID: 12060671 DOI: 10.1096/fj.01-1011fje] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/11/2022]
Abstract
An in vitro model has been developed to study migration, survival, proliferation, and capillary-like differentiation of human microvascular endothelial cells (HMVECs) in an environment that avoids tumor promoters and complex matrices. HMVEC monolayers were plated, then induced to form three-dimensional, capillary-like networks by overlaying with human type I collagen followed by a second overlay of collagen with embedded fibroblasts. Detachment and migration of endothelial cells into the matrix was triggered within hours by the overlaying collagen, and the fibroblasts stimulated survival and formation of cords, vacuoles, tubes, and, after 4 to 5 days, capillary networks. The differentiation into branching capillary-like structures was dependent on direct fibroblast-endothelial cell contact and was not achieved when fibroblasts were replaced by seven types of melanoma cells, which included radial and vertical growth phase primary and metastatic stages. Vascular endothelial growth factor (VEGF), when overexpressed in fibroblasts, stimulated endothelial cell proliferation and migration, whereas angiopoietin-1 (Ang-1) had only motogenic effects. Neutralizing antibodies against VEGF and blocking antibodies for VEGF-receptor 2 (VEGFR2) significantly inhibited but not completely obliterated capillary network formation, suggesting that the VEGF signaling pathway is important but not exclusive and that other fibroblast-derived soluble factors and fibroblast-endothelial cell contact are essential for endothelial cell survival and differentiation.
Collapse
Affiliation(s)
- Omaida C Velazquez
- The Wistar Institute, University of Pennsylvania, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
38
|
Ramsauer M, Krause D, Dermietzel R. Angiogenesis of the blood-brain barrier in vitro and the function of cerebral pericytes. FASEB J 2002; 16:1274-6. [PMID: 12153997 DOI: 10.1096/fj.01-0814fje] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/11/2023]
Abstract
Cerebral pericytes constitute an essential component of the blood-brain barrier (BBB) and are involved in blood vessel assembly. Recently, we reported on the induction of a BBB-specific enzyme expressed by cerebral pericytes (pericytic aminopeptidase N/pAPN) in coculture with cerebral endothelial cells. We completed this in vitro BBB system by adding astrocytes to these mixed cultures of endothelial cells and pericytes. Under these triculture conditions, endothelial cells and pericytes reorganize into capillary-like structures (CLSs). Capillary formation can also be achieved by the application of transforming growth factor beta 1 (TGF-b1) in the culture medium of endothelial-pericyte cultures lacking astrocytes. In contrast to the effect achieved by astrocytes, pericytes did not assemble with endothelial cells. In both cases (application of astrocytes or TGF-b1), endothelial cells underwent apoptosis. However, endothelial cells that form CLSs in the presence of pericytes appeared to be resistant to induction of apoptosis. On the basis of these observations, we concluded that astrocytes have a profound influence on the morphogenetic events underlying the organization of the vessel wall; that the effect of TGF-b1 is different from the astrocytic effect because it lacks induction of endothelial-pericyte association; and that pericytes stabilize CLSs formed by endothelial cells in coculture with astrocytes.
Collapse
Affiliation(s)
- Markus Ramsauer
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-Universität Bochum, Bochum, Germany
| | | | | |
Collapse
|
39
|
Venetsanakos E, Mirza A, Fanton C, Romanov SR, Tlsty T, McMahon M. Induction of tubulogenesis in telomerase-immortalized human microvascular endothelial cells by glioblastoma cells. Exp Cell Res 2002; 273:21-33. [PMID: 11795943 DOI: 10.1006/excr.2001.5424] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022]
Abstract
To facilitate the study of human endothelial cells we have used a replication defective retrovirus encoding the catalytic subunit of telomerase (hTERT) to derive populations of telomerase-immortalized human microvascular endothelial (TIME) cells. Whereas parental HMVECs became senescent on average within 35-45 population doublings (PDs), TIME cells have continued to proliferate for at least 200 PDs. TIME cells express readily detectable telomerase activity but display only a modest increase in telomere length. Karyotypic analysis reveals the cells to have a normal complement of human chromosomes with no evidence of gross genetic abnormalities. Furthermore, TIME cells retain many of the characteristics of the primary endothelial cells from which they were derived. For example, they express a panel of characteristic endothelial cell surface marker proteins such as CD31/PECAM-1 and alpha(v)beta3-integrin. In addition, TIME cells express receptors for low-density lipoprotein (LDL) receptor as they are competent for receptor-mediated endocytosis of fluorescent acetylated LDL. Importantly, when plated on matrigel, TIME cells undergo tubule formation. Moreover, when cocultured in the presence of human glioma cells, but not primary human astrocytes, TIME cells are induced to form stable tubules. Detachment of TIME cells from extracellular matrix leads to a form of programmed cell death known as anoikis. Conditional activation of the protein kinase Akt (Akt:ER*) significantly inhibited the onset of TIME cell anoikis under these conditions. We believe that the ability of hTERT to immortalize primary human endothelial cells, and the fact that such cells retain the endothelial characteristics of the cells from which they were derived, will greatly facilitate the analysis of human endothelial cell biology in vitro.
Collapse
MESH Headings
- Apoptosis
- Blotting, Western
- Brain Neoplasms/blood supply
- Brain Neoplasms/pathology
- Cells, Cultured
- Coculture Techniques
- DNA-Binding Proteins
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Glioblastoma/blood supply
- Glioblastoma/pathology
- Humans
- In Situ Hybridization, Fluorescence
- Karyotyping
- Lipoproteins, LDL
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Protein Serine-Threonine Kinases
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- RNA, Messenger/metabolism
- Receptors, LDL/metabolism
- Receptors, Vitronectin/metabolism
- Retroviridae/genetics
- Telomerase/genetics
- Telomerase/metabolism
- Telomere/metabolism
Collapse
Affiliation(s)
- Eleni Venetsanakos
- Department of Neurological Surgery, Cancer Research Institute, San Francisco, California 94143, USA
| | | | | | | | | | | |
Collapse
|
40
|
Phillips PG, Birnby LM, Narendran A, Milonovich WL. Nitric oxide modulates capillary formation at the endothelial cell-tumor cell interface. Am J Physiol Lung Cell Mol Physiol 2001; 281:L278-90. [PMID: 11404271 DOI: 10.1152/ajplung.2001.281.1.l278] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide synthase expression has been documented in lung tumors, but a potential role for nitric oxide (NO) in induction of capillary formation remains to be elucidated. The purpose of this report was to characterize the direct effects of NO at the level of the tumor-endothelium interface with respect to angiogenesis. A Transwell two-compartment culture system, human endothelial cells (EC), and two human non-small cell lung cancer (CA) lines that constitutively produce NO were used to simulate the EC-tumor cell interface. Both histological types of lung CA, squamous and adenocarcinoma, induced baseline capillary formation by EC within 3 days. This process was inhibited by NO in the microenvironment because decreasing NO production with 100 microM aminoguanidine (AG) significantly increased capillary formation, whereas coincubation with 100 microM AG plus 400 microM L-arginine returned angiogenesis to baseline values. We demonstrate further that NO may exert its inhibitory effects by influencing matrix metalloproteinase expression/activity and tyrosine phosphorylation of proteins in the sprouting tips of nascent capillaries.
Collapse
Affiliation(s)
- P G Phillips
- Research Service, Samuel S. Stratton Veterans Affairs Medical Center, Albany Medical College, Albany, New York 12208, USA.
| | | | | | | |
Collapse
|
41
|
Braet K, Paemeleire K, D'herde K, Sanderson MJ, Leybaert L. Astrocyte-endothelial cell calcium signals conveyed by two signalling pathways. Eur J Neurosci 2001. [DOI: 10.1111/j.1460-9568.2001.01372.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
|
42
|
Nirmala C, Jasti SL, Sawaya R, Kyritsis AP, Konduri SD, Ali-Osman F, Rao JS, Mohanam S. Effects of radiation on the levels of MMP-2, MMP-9 and TIMP-1 during morphogenic glial-endothelial cell interactions. Int J Cancer 2000; 88:766-71. [PMID: 11072246 DOI: 10.1002/1097-0215(20001201)88:5<766::aid-ijc13>3.0.co;2-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/09/2022]
Abstract
Radiation-induced damage to the central nervous system (CNS) is believed to target glial or endothelial cells or both, although the pathophysiology of the process is poorly understood. We therefore used a coculture system, in which glioblastoma SNB19 cells induced bovine retinal endothelial (BRE) cells to form capillary-like structures, to examine the role of ionizing radiation in modulating the production of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinase-1 (TIMP-1). In particular, we irradiated both BRE cells and cocultures of BRE and SNB19 cells with a single dose of X-rays and then estimated the levels of MMP-2, MMP-9 and TIMP-1. Gelatin zymography revealed a continuous increase in the levels of MMP-2 and MMP-9 during capillary-like structure formation. Of note, the levels of both MMP-2 and MMP-9 were markedly higher in irradiated cocultures at 72 hr after irradiation than in untreated cocultures. Northern blot analysis also demonstrated an increased expression of MMP-9 mRNA in the irradiated cocultures. In addition, TIMP-1 mRNA and protein levels increased up to 48 hr in both irradiated and nonirradiated BRE cells and in nonirradiated cocultures, but there was a significant decrease in the TIMP-1 mRNA and protein levels in irradiated cocultures. It takes about 72 hr for capillaries to form in nonirradiated cocultures, but these capillary networks fail to form in endothelial cells in irradiated cocultures. These findings establish that radiation differentially affects the production of MMP-2, MMP-9 and TIMP-1 during glial-endothelial morphogenesis and suggest mechanisms by which microvessels in the CNS respond to radiation.
Collapse
Affiliation(s)
- C Nirmala
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
In the last two decades, much attention has been focussed on mechanisms of glioma vascularization including the investigation of growth factors and receptors involved. Recently, these efforts resulted in various approaches for antiangiogenic treatment of experimental brain tumors. These basic science and preclinical trials need an assortment of models, which should allow investigating a variety of questions. Several objectives concerning basic endothelial cell (EC) characteristics can adequately be studied in vitro using EC monolayer assays. Three-dimensional spheroid techniques respect the more complex cell-cell and cell-environment interplay within a three-dimensional culture. To optimize the imitation of the crucial interaction of human gliomas with host endothelial cells, immunological cells and extracellular matrix, animal models are mandatory. An essential rule is to utilize an orthotopic model, since tumor-host interaction is organ specific. To avoid alloimmunogenic responses, it is desirable to use weakly or not immunogenic glioma grafts, what is best accomplished in a syngeneic model. However, since rat gliomas poorly resemble human glioma growth patterns, human glioma xenografting into immunocompromized animals should be considered. In vivo monitoring techniques like videoscopy via a cranial window or magnetic resonance imaging (MRI) allow for functional studies and improve the validity of the model employed. Finally, it is essentially to recognize the limitations of each model considered and to select that model, which seems to be most appropriate for the objectives to be investigated.
Collapse
Affiliation(s)
- R H Goldbrunner
- Department of Neurosurgery, University of Wuerzburg, Germany
| | | | | | | |
Collapse
|
44
|
Abstract
1. The study of the blood-brain barrier and its various realms offers a myriad of opportunities for scientific exploration. This review focuses on two of these areas in particular: the induction of the blood-brain barrier and the molecular mechanisms underlying this developmental process. 2. The creation of the blood-brain barrier is considered a specific step in the differentiation of cerebral capillary endothelial cells, resulting in a number of biochemical and functional alterations. Although the specific endothelial properties which maintain the homeostasis in the central nervous system necessary for neuronal function have been well described, the inductive mechanisms which trigger blood-brain barrier establishment in capillary endothelial cells are unknown. 3. The timetable of blood-brain barrier formation is still a matter of debate, caused largely by the use of varying experimental systems and by the general difficulty of quantitatively measuring the degree of blood-brain barrier "tightness." However, there is a general consensus that a gradual formation of the blood-brain barrier starts shortly after intraneural neovascularization and that the neural microenvironment (neurons and/or astrocytes) plays a key role in inducing blood-brain barrier function in capillary endothelial cells. This view stems from numerous in vitro experiments using mostly cocultures of capillary endothelial cells and astrocytes and assays for easily measurable blood-brain barrier markers. In vivo, there are great difficulties in proving the inductive influence of the neuronal environment. Also dealt with in this article are brain tumors, the least understood in vivo systems, and the induction or noninduction of barrier function in the newly established tumor vascularization. 4. Finally, this review tries to elucidate the question concerning the nature of the inductive signal eliciting blood-brain barrier formation in the cerebral microvasculature.
Collapse
Affiliation(s)
- H C Bauer
- Institute für Molekularbiologie, Osterr. Akad. d. Wissenschaften, Salzburg, Austria.
| | | |
Collapse
|
45
|
Wolburg H, Liebner S, Reichenbach A, Gerhardt H. The pecten oculi of the chicken: a model system for vascular differentiation and barrier maturation. INTERNATIONAL REVIEW OF CYTOLOGY 1999; 187:111-59. [PMID: 10212979 DOI: 10.1016/s0074-7696(08)62417-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/13/2023]
Abstract
The pecten oculi is a convolute of blood vessels in the vitreous body of the avian eye. This structure is well known for more than a century, but its functions are still a matter of controversies. One of these functions must be the formation of a blood-retina barrier because there is no diffusion barrier for blood-borne compounds available between the pecten and the retina. Surprisingly, the blood-retina barrier characteristics of this organ have not been studied so far, although the pecten oculi may constitute a fascinating model of vascular differentiation and barrier maturation: Pectinate endothelial cells grow by angiogenesis from the ophthalmotemporal artery into the pecten primordium and consecutively gain barrier properties. The pectinate pigmented cells arise during development from retinal pigment epithelial cells and subsequently lose barrier properties. These inverse transdifferentiation processes may be triggered by the peculiar microenvironment in the vitreous body. In addition, the question is discussed whether the avascularity of the avian retina may be due to the specific metabolic activity of the pecten.
Collapse
Affiliation(s)
- H Wolburg
- Institute of Pathology, University of Tübingen, Germany
| | | | | | | |
Collapse
|
46
|
Bolton SJ, Anthony DC, Perry VH. Loss of the tight junction proteins occludin and zonula occludens-1 from cerebral vascular endothelium during neutrophil-induced blood-brain barrier breakdown in vivo. Neuroscience 1998; 86:1245-57. [PMID: 9697130 DOI: 10.1016/s0306-4522(98)00058-x] [Citation(s) in RCA: 271] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/08/2023]
Abstract
The tight junctions found between cerebral vascular endothelial cells form the basis of the blood-brain barrier. Breakdown of the blood-brain barrier is a feature of a variety of CNS pathologies that are characterized by extensive leucocyte recruitment, such as multiple sclerosis and stroke. The molecular mechanisms associated with opening of the blood-brain barrier and leucocyte recruitment in vivo are currently poorly understood. We have used an in vivo rat model to investigate the molecular response of the CNS endothelium to neutrophil adhesion and migration. Injection of interleukin-1 beta into the striatum of juvenile brains results in a neutrophil-dependent increase in vessel permeability at 4 h. Only a subset of blood vessels were associated with neutrophil recruitment. These particular vessels displayed an increase in phosphotyrosine staining, loss of the tight junctional proteins, occludin and zonula occludens-1, and apparent redistribution of the adherens junction protein vinculin. Examination of these vessels under the electron microscope indicated that the cell-cell adhesions in such vessels are morphologically different from normal junctions. This study provides the first direct evidence in vivo that leucocyte recruitment can trigger signal transduction cascades leading to junctional disorganization and blood-brain barrier breakdown. Our results have established an endothelial cell molecular profile associated with leucocyte-induced blood-brain barrier breakdown in vivo, and the relevance of different in vitro cell culture models may now be viewed more objectively.
Collapse
Affiliation(s)
- S J Bolton
- Department of Pharmacology, University of Oxford, U.K
| | | | | |
Collapse
|
47
|
Black AF, Berthod F, L'heureux N, Germain L, Auger FA. In vitro reconstruction of a human capillary-like network in a tissue-engineered skin equivalent. FASEB J 1998; 12:1331-40. [PMID: 9761776 DOI: 10.1096/fasebj.12.13.1331] [Citation(s) in RCA: 288] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/30/2022]
Abstract
For patients with extensive burns, wound coverage with an autologous in vitro reconstructed skin made of both dermis and epidermis should be the best alternative to split-thickness graft. Unfortunately, various obstacles have delayed the widespread use of composite skin substitutes. Insufficient vascularization has been proposed as the most likely reason for their unreliable survival. Our purpose was to develop a vascular-like network inside tissue-engineered skin in order to improve graft vascularization. To reach this aim, we fabricated a collagen biopolymer in which three human cell types keratinocytes, dermal fibroblasts, and umbilical vein endothelial cells were cocultured. We demonstrated that the endothelialized skin equivalent (ESE) promoted spontaneous formation of capillary-like structures in a highly differentiated extracellular matrix. Immunohistochemical analysis and transmission electron microscopy of the ESE showed characteristics associated with the microvasculature in vivo (von Willebrand factor, Weibel-Palade bodies, basement membrane material, and intercellular junctions). We have developed the first endothelialized human tissue-engineered skin in which a network of capillary-like tubes is formed. The transplantation of this ESE on human should accelerate graft revascularization by inosculation of its preexisting capillary-like network with the patient's own blood vessels, as it is observed with autografts. In addition, the ESE turns out to be a promising in vitro angiogenesis model.
Collapse
Affiliation(s)
- A F Black
- Laboratoire d'Organogénèse Expérimentale/LOEX, Centre Hospitalier Affilié, Pavillon Saint-Sacrement and Department of Surgery, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada G1S 4L8. HYPERLINKmailto:
| | | | | | | | | |
Collapse
|
48
|
|
49
|
Duport S, Robert F, Muller D, Grau G, Parisi L, Stoppini L. An in vitro blood-brain barrier model: cocultures between endothelial cells and organotypic brain slice cultures. Proc Natl Acad Sci U S A 1998; 95:1840-5. [PMID: 9465104 PMCID: PMC19200 DOI: 10.1073/pnas.95.4.1840] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/16/1997] [Accepted: 12/22/1997] [Indexed: 02/06/2023] Open
Abstract
This communication describes a novel in vitro blood-brain barrier (BBB) model: organotypic slice cultures from the central nervous system were overlaid on endothelial cell monolayers grown on permeable membranes. Morphological, electrophysiological, and microdialysis approaches were carried out to characterize and validate this model. After 10 days in coculture, morphological studies reveal the presence of tight junctions. Electrophysiological recordings of neuronal activity performed on organotypic cultures with or without an endothelial cell monolayer show that amplitude of evoked responses were comparable, indicating good viability of cocultures after 2 weeks. Perfusion of known BBB permeable or nonpermeable molecules was used to test the coculture tightness in conjunction with electrophysiological or microdialysis approaches: application of glutamate (Glu), which doesn't easily cross the BBB, triggers off rhythmic activity only in control cultures, whereas epileptogenic activity was observed in both control cultures and cocultures during perfusions with picrotoxin, a molecule that can diffuse through the BBB. Finally, the microdialysis technique was used to determine the permeability of molecules coming from the perfusion chamber: L-dopa, dopamine, and Glu were employed to assess the selective permeability of the coculture model. Thus, these results indicate that the in vitro model described possesses characteristics similar to those of the BBB in situ and that cocultures of organotypic slices and endothelial cell monolayers have potential as a powerful tool for studying biochemical mechanisms regulating BBB function and drug delivery to the central nervous system.
Collapse
Affiliation(s)
- S Duport
- Department of Pharmacology, Centre Médical Universitaire, CH-1211 Geneva 4, Switzerland
| | | | | | | | | | | |
Collapse
|
50
|
Sorensen M, Steenberg B, Knipp GT, Wang W, Steffansen B, Frokjaer S, Borchardt RT. The effect of beta-turn structure on the permeation of peptides across monolayers of bovine brain microvessel endothelial cells. Pharm Res 1997; 14:1341-8. [PMID: 9358545 DOI: 10.1023/a:1012104301773] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/05/2023]
Abstract
PURPOSE To investigate the effects of the beta-turn structure of a peptide on its permeation via the paracellular and transcellular routes across cultured bovine brain microvessel endothelial cell (BBMEC) monolayers, an in vitro model of the blood-brain barrier (BBB). METHODS The effective permeability coefficients (Peff) of the model peptides were determined across BBMEC monolayers. The dimensions of the aqueous pores in the tight junctions (TJs) of the BBMEC monolayers were determined using a series of hydrophilic permeants. This value and the molecular radius of each peptide were used to calculate the theoretical paracellular (PP*) and transcellular (PT*) permeability coefficients for each peptide. RESULTS A comparison of the theoretical PP* values with the observed Peff values was made for a series of model peptides. For the most hydrophobic peptides (Ac-PheProXaaIle-NH2 and Ac-PheProXaaIleVal-NH2; Xaa = Gly, Ile), it was concluded that the Gly-containing peptide of each pair more readily permeates BBMEC monolayers via the transcellular pathway than the Ile-containing analog. In addition, the Gly-containing peptides, which exhibit more beta-turn structure, were shown to be more lipophilic than the Ile-containing peptides as estimated by the log of their 1-octanol:HBSS partition coefficients (log Po/w). However, the three hydrophilic peptide pairs (Ac-TyrProXaaAspVal-NH2, Ac-TyrProXaaAsnVal-NH2, and Ac-TyrProXaaIleVal-NH2; Xaa = Gly, Ile) were found to permeate BBMEC monolayers predominantly via the paracellular pathway. No differences were observed in the Peff values of the hydrophilic peptides having higher beta-turn structures as compared to the peptides lacking these structural features. In addition, the Ile-containing peptides exhibited significantly higher log Po/w values than the Gly-containing hydrophilic peptides. CONCLUSIONS Hydrophobic peptides that exhibit significant beta-turn structure in solution are more lipophilic as measured by log Po/w, and more readily permeate BBMEC monolayers via the transcellular route than hydrophobic peptides that lack this type of solution structure. Similar secondary structural features in hydrophilic peptides do not appear to sufficiently alter the physicochemical properties of the peptides so as to alter their paracellular flux through BBMEC monolayers.
Collapse
Affiliation(s)
- M Sorensen
- Department of Pharmaceutics, Royal Danish School of Pharmacy, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|