brain pericytes
Recently Published Documents


TOTAL DOCUMENTS

131
(FIVE YEARS 51)

H-INDEX

31
(FIVE YEARS 6)

2022 ◽  
Vol 15 (1) ◽  
Author(s):  
Seo Young Kim ◽  
Jihye Choi ◽  
Junhee Roh ◽  
Chul Hoon Kim

AbstractIn the CNS, pericytes are important for maintaining the blood–brain barrier (BBB) and for controlling blood flow. Recently, several methods were suggested for the differentiation of human pluripotent stem cells (hPSCs) into brain mural cells, specifically pericytes or vascular smooth muscle cells (vSMCs). Unfortunately, identifying the pericytes from among such hPSC-derived mural cells has been challenging. This is due both to the lack of pericyte-specific markers and to the loss of defining anatomical information inherent to culture conditions. We therefore asked whether NeuroTrace 500/525, a newly developed dye that shows cell-specific uptake into pericytes in the mouse brain, can help identify human induced pluripotent stem cell (hiPSC)-derived brain pericyte-like cells. First, we found that NeuroTrace 500/525 specifically stains primary cultured human brain pericytes, confirming its specificity in vitro. Second, we found that NeuroTrace 500/525 specifically labels hiPSC-derived pericyte-like cells, but not endothelial cells or vSMCs derived from the same hiPSCs. Last, we found that neuroectoderm-derived vSMCs, which have pericyte-like features, also take up NeuroTrace 500/525. These data indicate NeuroTrace 500/525 is useful for identifying pericyte-like cells among hiPSC-derived brain mural cells.


Cells ◽  
2021 ◽  
Vol 11 (1) ◽  
pp. 133
Author(s):  
Camille Menaceur ◽  
Fabien Gosselet ◽  
Laurence Fenart ◽  
Julien Saint-Pol

The construction of the blood–brain barrier (BBB), which is a natural barrier for maintaining brain homeostasis, is the result of a meticulous organisation in space and time of cell–cell communication processes between the endothelial cells that carry the BBB phenotype, the brain pericytes, the glial cells (mainly the astrocytes), and the neurons. The importance of these communications for the establishment, maturation and maintenance of this unique phenotype had already been suggested in the pioneering work to identify and demonstrate the BBB. As for the history of the BBB, the evolution of analytical techniques has allowed knowledge to evolve on the cell–cell communication pathways involved, as well as on the role played by the cells constituting the neurovascular unit in the maintenance of the BBB phenotype, and more particularly the brain pericytes. This review summarises the key points of the history of the BBB, from its origin to the current knowledge of its physiology, as well as the cell–cell communication pathways identified so far during its development, maintenance, and pathophysiological alteration.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi199-vi199
Author(s):  
Cynthia Hajal ◽  
Yoojin Shin ◽  
Leanne Li ◽  
Jean Carlos Serrano ◽  
Tyler Jacks ◽  
...  

Abstract Although brain metastases are common in cancer patients, little is known about the mechanisms of extravasation across the blood-brain barrier (BBB), a key step in the metastatic cascade that regulates the entry of cancer cells into the brain parenchyma through its selective endothelial barrier. Progress in this area has been impeded by challenges in conducting high spatio-temporal resolution imaging in vivo and isolating factors and cellular interactions directly contributing to extravasation rather than cancer survival and proliferation in the brain tissue. To address these limitations, we engineered a three-dimensional in vitro BBB microvascular model with endothelial cells derived from induced pluripotent stem cells, brain pericytes, and astrocytes, into which we perfused cancer cells to recapitulate their circulation and extravasation at the BBB. With this platform, we revealed that astrocytes play a major role in promoting cancer cell transmigration via their secretion of C-C motif chemokine ligand 2 (CCL2). We found that this chemokine promoted the chemotaxis and chemokinesis of cancer cells via their C-C chemokine receptor type 2 (CCR2), with no significant changes in vascular permeability. These findings were validated in vivo, where CCR2-deficient cancer cells exhibited significantly reduced cancer cell arrest and transmigration in mouse brain capillaries. Our results attest to the translational value of our BBB-on-a-chip model and reveal that the CCL2-CCR2 astrocyte-cancer cell axis plays a fundamental role in extravasation and consequently metastasis to the brain.


2021 ◽  
pp. 105561
Author(s):  
Rayan Khaddaj-Mallat ◽  
Natija Aldib ◽  
Maxime Bernard ◽  
Anne-Sophie Paquette ◽  
Aymeric Ferreira ◽  
...  

2021 ◽  
Vol 22 (21) ◽  
pp. 11622
Author(s):  
Matteo Bocci ◽  
Clara Oudenaarden ◽  
Xavier Sàenz-Sardà ◽  
Joel Simrén ◽  
Arvid Edén ◽  
...  

A wide range of neurological manifestations have been associated with the development of COVID-19 following SARS-CoV-2 infection. However, the etiology of the neurological symptomatology is still largely unexplored. Here, we used state-of-the-art multiplexed immunostaining of human brains (n = 6 COVID-19, median age = 69.5 years; n = 7 control, median age = 68 years) and demonstrated that expression of the SARS-CoV-2 receptor ACE2 is restricted to a subset of neurovascular pericytes. Strikingly, neurological symptoms were exclusive to, and ubiquitous in, patients that exhibited moderate to high ACE2 expression in perivascular cells. Viral dsRNA was identified in the vascular wall and paralleled by perivascular inflammation, as signified by T cell and macrophage infiltration. Furthermore, fibrinogen leakage indicated compromised integrity of the blood–brain barrier. Notably, cerebrospinal fluid from additional 16 individuals (n = 8 COVID-19, median age = 67 years; n = 8 control, median age = 69.5 years) exhibited significantly lower levels of the pericyte marker PDGFRβ in SARS-CoV-2-infected cases, indicative of disrupted pericyte homeostasis. We conclude that pericyte infection by SARS-CoV-2 underlies virus entry into the privileged central nervous system space, as well as neurological symptomatology due to perivascular inflammation and a locally compromised blood–brain barrier.


Endocrinology ◽  
2021 ◽  
Author(s):  
Nele Warmke ◽  
Fiona Platt ◽  
Alexander F Bruns ◽  
Claire H Ozber ◽  
Natalie J Haywood ◽  
...  

Abstract Pericytes regulate vascular development, stability and quiescence; their dysfunction contributes to diabetic retinopathy. To explore the role of insulin receptors in pericyte biology, we created pericyte insulin receptor knockout mice (PIRKO) by crossing PDGFR β-Cre mice with insulin receptor (Insr) floxed mice. Their neonatal retinal vasculature exhibited peri-venous hypervascularity with venular dilatation, plus increased angiogenic sprouting in superficial and deep layers. Pericyte coverage of capillaries was unaltered in peri-venous and peri-arterial plexi and no differences in vascular regression or endothelial proliferation were apparent. Isolated brain pericytes from PIRKO had decreased angiopoietin-1 mRNA, whereas retinal and lung angiopoietin-2 mRNA was increased. Endothelial phospho-Tie2 staining was diminished and FoxO1 was more frequently nuclear localized in the peri-venous plexus of PIRKO, in keeping with reduced angiopoietin-Tie2 signaling. Silencing of Insr in human brain pericytes led to reduced insulin-stimulated angiopoietin-1 secretion, and conditioned media from these cells was less able to induce Tie2 phosphorylation in human endothelial cells. Hence, insulin signaling in pericytes promotes angiopoietin-1 secretion and endothelial Tie2 signaling and perturbation of this leads to excessive vascular sprouting and venous plexus abnormalities. This phenotype mimics elements of diabetic retinopathy, and future work should evaluate pericyte insulin signaling in this disease.


2021 ◽  
Vol 15 ◽  
Author(s):  
Elliott Hibbs ◽  
Seth Love ◽  
J. Scott Miners

Pericytes are vascular mural cells that contract and relax in response to vasoactive stimuli to regulate neurovascular coupling and cerebral blood flow. Pericytes are damaged and degenerate in Alzheimer’s disease (AD). We previously showed that the level of the regulatory vasoconstrictor, endothelin-1 (EDN1), is elevated in AD cerebral cortex and upregulated by amyloid-beta (Aβ). We have used electrical impedance analysis to monitor the contractile and proliferative response of cultured human fetal and adult brain-derived pericytes to EDN1 in real-time. EDN1 caused transient, dose-dependent contraction of fetal and adult brain pericytes that was mediated by EDN1 type A receptors and increased the subsequent proliferation of fetal but not adult cells. The contractile responses to EDN1 were weaker in the adult pericytes. The EDN1-mediated contractile response of fetal pericytes was unchanged after exposure to Aβ1–40 or Aβ1–42 (0.1–10 μM) for 1 h but both contraction and subsequent relaxation were significantly impaired upon exposure to Aβ for 24 h. These data suggest that chronic exposure to Aβ interferes with EDN1-mediated pericyte contractility, potentially contributing to neurovascular uncoupling and reduced cerebral blood flow in AD.


Author(s):  
Thanh Thuy Dan Pham ◽  
Shuin Park ◽  
Kamal Kolluri ◽  
Riki Kawaguchi ◽  
Lingjun Wang ◽  
...  

2021 ◽  
Vol 7 (34) ◽  
pp. eabi9119 ◽  
Author(s):  
Lena Neufeld ◽  
Eilam Yeini ◽  
Noa Reisman ◽  
Yael Shtilerman ◽  
Dikla Ben-Shushan ◽  
...  

Many drugs show promising results in laboratory research but eventually fail clinical trials. We hypothesize that one main reason for this translational gap is that current cancer models are inadequate. Most models lack the tumor-stroma interactions, which are essential for proper representation of cancer complexed biology. Therefore, we recapitulated the tumor heterogenic microenvironment by creating fibrin glioblastoma bioink consisting of patient-derived glioblastoma cells, astrocytes, and microglia. In addition, perfusable blood vessels were created using a sacrificial bioink coated with brain pericytes and endothelial cells. We observed similar growth curves, drug response, and genetic signature of glioblastoma cells grown in our 3D-bioink platform and in orthotopic cancer mouse models as opposed to 2D culture on rigid plastic plates. Our 3D-bioprinted model could be the basis for potentially replacing cell cultures and animal models as a powerful platform for rapid, reproducible, and robust target discovery; personalized therapy screening; and drug development.


Sign in / Sign up

Export Citation Format

Share Document