Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kavi Devraj is active.

Publication


Featured researches published by Kavi Devraj.


PLOS Biology | 2014

Extracellular vesicle-mediated transfer of genetic information between the hematopoietic system and the brain in response to inflammation.

Kirsten Ridder; Sascha Keller; Maria Dams; Anne-Kathleen Rupp; Jessica Schlaudraff; Domenico Del Turco; Julia Starmann; Jadranka Macas; Darja Karpova; Kavi Devraj; Candan Depboylu; Britta Landfried; Bernd Arnold; Karl H. Plate; Günter U. Höglinger; Holger Sültmann; Peter Altevogt; Stefan Momma

When stimulated by inflammation, peripheral blood cells signal directly to neurons in the brain via the transfer of functional RNA enclosed in extracellular vesicles.


Journal of Experimental Medicine | 2012

Endothelial Wnt/β-catenin signaling inhibits glioma angiogenesis and normalizes tumor blood vessels by inducing PDGF-B expression

Marco Reis; Cathrin J. Czupalla; Nicole Ziegler; Kavi Devraj; Jenny Zinke; Sascha Seidel; Rosario Heck; Sonja Thom; Jadranka Macas; Ernesto Bockamp; Marcus Fruttiger; Makoto M. Taketo; Stefanie Dimmeler; Karl H. Plate; Stefan Liebner

Wnt modulates glioma vascularization by regulating PDGF-B expression.


PLOS ONE | 2013

Wnt activation of immortalized brain endothelial cells as a tool for generating a standardized model of the blood brain barrier in vitro.

Roberta Paolinelli; Monica Corada; Luca Ferrarini; Kavi Devraj; Cédric Artus; Cathrin J. Czupalla; Noemi Rudini; Luigi Maddaluno; Eleanna Papa; Britta Engelhardt; Pierre Olivier Couraud; Stefan Liebner; Elisabetta Dejana

Reproducing the characteristics and the functional responses of the blood–brain barrier (BBB) in vitro represents an important task for the research community, and would be a critical biotechnological breakthrough. Pharmaceutical and biotechnology industries provide strong demand for inexpensive and easy-to-handle in vitro BBB models to screen novel drug candidates. Recently, it was shown that canonical Wnt signaling is responsible for the induction of the BBB properties in the neonatal brain microvasculature in vivo. In the present study, following on from earlier observations, we have developed a novel model of the BBB in vitro that may be suitable for large scale screening assays. This model is based on immortalized endothelial cell lines derived from murine and human brain, with no need for co-culture with astrocytes. To maintain the BBB endothelial cell properties, the cell lines are cultured in the presence of Wnt3a or drugs that stabilize β-catenin, or they are infected with a transcriptionally active form of β-catenin. Upon these treatments, the cell lines maintain expression of BBB-specific markers, which results in elevated transendothelial electrical resistance and reduced cell permeability. Importantly, these properties are retained for several passages in culture, and they can be reproduced and maintained in different laboratories over time. We conclude that the brain-derived endothelial cell lines that we have investigated gain their specialized characteristics upon activation of the canonical Wnt pathway. This model may be thus suitable to test the BBB permeability to chemicals or large molecular weight proteins, transmigration of inflammatory cells, treatments with cytokines, and genetic manipulation.


Methods of Molecular Biology | 2014

In vitro models of the blood-brain barrier.

Cathrin J. Czupalla; Stefan Liebner; Kavi Devraj

The blood-brain barrier (BBB) proper is composed of endothelial cells (ECs) of the cerebral microvasculature, which are interconnected by tight junctions (TJs) that in turn form a physical barrier restricting paracellular flux. Tight control of vascular permeability is essential for the homeostasis and functionality of the central nervous system (CNS). In vitro BBB models have been in use for decades and have been of great benefit in the process of investigating and understanding the cellular and molecular mechanisms underlying BBB establishment. BBB integrity changes can be addressed in vitro by determining cell monolayer permeability (Pe) to different solutes and measuring trans-endothelial electrical resistance (TEER).This chapter describes procedures that can be utilized for both freshly isolated mouse brain microvascular ECs (MBMECs) and murine or human brain EC lines (bEnd5 or hCMEC/D3), cultivated either as a single monolayer or in cocultivation with primary mouse astrocytes (ACs). It starts with detailed information on how to perform transwell cell culture, including coating of inserts and seeding of the ECs and ACs. Moreover, it encompasses instructions for electrical assessment of the in vitro BBB using the more recent cellZscope(®) device, which was traditionally performed with chopstick electrodes of voltohmmeter type (EVOM). From continuous impedance measurements, the cellZscope(®) device provides TEER (paracellular resistance) and cell membrane capacitance (Ccl-transcellular resistance), two independent measures of monolayer integrity. Additionally, this chapter provides guidance through subsequent experiments such as permeability analysis (Pe, flux), expression analysis (qRT-PCR and Western blotting), and localization analysis of BBB junction proteins (immunocytochemistry) using the same inserts subjected earlier to impedance analysis.As numerous diseases are associated with BBB breakdown, researchers aim to continuously improve and refine in vitro BBB models to mimic in vivo conditions as closely as possible. This chapter summarizes protocols with the intention to provide a collection of BBB in vitro assays that generate reproducible results not only with primary brain ECs but also with EC lines to open up the field for a broader spectrum of researchers who intend to investigate the BBB in vitro particularly aiming at therapeutic aspects.


Circulation | 2017

Long Noncoding RNA MANTIS Facilitates Endothelial Angiogenic Function

Matthias S. Leisegang; Christian Fork; Ivana Josipovic; Florian Martin Richter; Jens Preussner; Jiong Hu; Matthew J. Miller; Jeremy Epah; Patrick Hofmann; Stefan Günther; Franziska Moll; Chanil Valasarajan; Juliana Heidler; Yuliya Ponomareva; Thomas M. Freiman; Lars Maegdefessel; Karl H. Plate; Michel Mittelbronn; Shizuka Uchida; Carsten Künne; Konstantinos Stellos; Ralph T. Schermuly; Norbert Weissmann; Kavi Devraj; Ilka Wittig; Reinier A. Boon; Stefanie Dimmeler; Soni Savai Pullamsetti; Mario Looso; Francis J. Miller

Background: The angiogenic function of endothelial cells is regulated by numerous mechanisms, but the impact of long noncoding RNAs (lncRNAs) has hardly been studied. We set out to identify novel and functionally important endothelial lncRNAs. Methods: Epigenetically controlled lncRNAs in human umbilical vein endothelial cells were searched by exon-array analysis after knockdown of the histone demethylase JARID1B. Molecular mechanisms were investigated by RNA pulldown and immunoprecipitation, mass spectrometry, microarray, several knockdown approaches, CRISPR-Cas9, assay for transposase-accessible chromatin sequencing, and chromatin immunoprecipitation in human umbilical vein endothelial cells. Patient samples from lung and tumors were studied for MANTIS expression. Results: A search for epigenetically controlled endothelial lncRNAs yielded lncRNA n342419, here termed MANTIS, as the most strongly regulated lncRNA. Controlled by the histone demethylase JARID1B, MANTIS was downregulated in patients with idiopathic pulmonary arterial hypertension and in rats treated with monocrotaline, whereas it was upregulated in carotid arteries of Macaca fascicularis subjected to atherosclerosis regression diet, and in endothelial cells isolated from human glioblastoma patients. CRISPR/Cas9-mediated deletion or silencing of MANTIS with small interfering RNAs or GapmeRs inhibited angiogenic sprouting and alignment of endothelial cells in response to shear stress. Mechanistically, the nuclear-localized MANTIS lncRNA interacted with BRG1, the catalytic subunit of the switch/sucrose nonfermentable chromatin-remodeling complex. This interaction was required for nucleosome remodeling by keeping the ATPase function of BRG1 active. Thereby, the transcription of key endothelial genes such as SOX18, SMAD6, and COUP-TFII was regulated by ensuring efficient RNA polymerase II machinery binding. Conclusion: MANTIS is a differentially regulated novel lncRNA facilitating endothelial angiogenic function.


Acta Neuropathologica | 2016

Angiopoietin-2-induced blood-brain barrier compromise and increased stroke size are rescued by VE-PTP-dependent restoration of Tie2 signaling.

Stefanie Gurnik; Kavi Devraj; Jadranka Macas; Maiko Yamaji; Julia Starke; Alexander Scholz; Kathleen Sommer; Mariangela Di Tacchio; Rajkumar Vutukuri; Heike Beck; Michel Mittelbronn; Christian Foerch; Waltraud Pfeilschifter; Stefan Liebner; Kevin G. Peters; Karl H. Plate; Yvonne Reiss

The homeostasis of the central nervous system is maintained by the blood–brain barrier (BBB). Angiopoietins (Ang-1/Ang-2) act as antagonizing molecules to regulate angiogenesis, vascular stability, vascular permeability and lymphatic integrity. However, the precise role of angiopoietin/Tie2 signaling at the BBB remains unclear. We investigated the influence of Ang-2 on BBB permeability in wild-type and gain-of-function (GOF) mice and demonstrated an increase in permeability by Ang-2, both in vitro and in vivo. Expression analysis of brain endothelial cells from Ang-2 GOF mice showed a downregulation of tight/adherens junction molecules and increased caveolin-1, a vesicular permeability-related molecule. Immunohistochemistry revealed reduced pericyte coverage in Ang-2 GOF mice that was supported by electron microscopy analyses, which demonstrated defective intra-endothelial junctions with increased vesicles and decreased/disrupted glycocalyx. These results demonstrate that Ang-2 mediates permeability via paracellular and transcellular routes. In patients suffering from stroke, a cerebrovascular disorder associated with BBB disruption, Ang-2 levels were upregulated. In mice, Ang-2 GOF resulted in increased infarct sizes and vessel permeability upon experimental stroke, implicating a role of Ang-2 in stroke pathophysiology. Increased permeability and stroke size were rescued by activation of Tie2 signaling using a vascular endothelial protein tyrosine phosphatase inhibitor and were independent of VE-cadherin phosphorylation. We thus identified Ang-2 as an endothelial cell-derived regulator of BBB permeability. We postulate that novel therapeutics targeting Tie2 signaling could be of potential use for opening the BBB for increased CNS drug delivery or tighten it in neurological disorders associated with cerebrovascular leakage and brain edema.


Journal of Cerebral Blood Flow and Metabolism | 2016

BACE-1 is expressed in the blood-brain barrier endothelium and is upregulated in a murine model of Alzheimer's disease.

Kavi Devraj; Slobodan Poznanovic; Christoph Spahn; Gerhard P. Schwall; Patrick N. Harter; Michel Mittelbronn; Katia Antoniello; Paolo Paganetti; Andreas Muhs; Mike Heilemann; Richard A. Hawkins; André Schrattenholz; Stefan Liebner

Endothelial cells of the blood–brain barrier form a structural and functional barrier maintaining brain homeostasis via paracellular tight junctions and specific transporters such as P-glycoprotein. The blood–brain barrier is responsible for negligible bioavailability of many neuroprotective drugs. In Alzheimer’s disease, current treatment approaches include inhibitors of BACE-1 (β-site of amyloid precursor protein cleaving enzyme), a proteinase generating neurotoxic β-amyloid. It is known that BACE-1 is highly expressed in endosomes and membranes of neurons and glia. We now provide evidence that BACE-1 is expressed in blood–brain barrier endothelial cells of human, mouse, and bovine origin. We further show its predominant membrane localization by 3D-dSTORM super-resolution microscopy, and by biochemical fractionation that further shows an abluminal distribution of BACE-1 in brain microvessels. We confirm its functionality in processing APP in primary mouse brain endothelial cells. In an Alzheimer’s disease mouse model we show that BACE-1 is upregulated at the blood–brain barrier compared to healthy controls. We therefore suggest a critical role for BACE-1 at the blood–brain barrier in β-amyloid generation and in vascular aspects of Alzheimer’s disease, particularly in the development of cerebral amyloid angiopathy.


Nature | 2017

Inhibition of soluble epoxide hydrolase prevents diabetic retinopathy

Jiong Hu; Sarah Dziumbla; J Lin; Sofia Iris Bibli; Sven Zukunft; Julian De Mos; Khader Awwad; Timo Frömel; Andreas Jungmann; Kavi Devraj; Zhixing Cheng; Liya Wang; Sascha Fauser; Charles G. Eberhart; Akrit Sodhi; Bruce D. Hammock; Stefan Liebner; Oliver J. Müller; Clemens Glaubitz; Hp Hammes; Rüdiger Popp; Ingrid Fleming

Diabetic retinopathy is an important cause of blindness in adults, and is characterized by progressive loss of vascular cells and slow dissolution of inter-vascular junctions, which result in vascular leakage and retinal oedema. Later stages of the disease are characterized by inflammatory cell infiltration, tissue destruction and neovascularization. Here we identify soluble epoxide hydrolase (sEH) as a key enzyme that initiates pericyte loss and breakdown of endothelial barrier function by generating the diol 19,20-dihydroxydocosapentaenoic acid, derived from docosahexaenoic acid. The expression of sEH and the accumulation of 19,20-dihydroxydocosapentaenoic acid were increased in diabetic mouse retinas and in the retinas and vitreous humour of patients with diabetes. Mechanistically, the diol targeted the cell membrane to alter the localization of cholesterol-binding proteins, and prevented the association of presenilin 1 with N-cadherin and VE-cadherin, thereby compromising pericyte–endothelial cell interactions and inter-endothelial cell junctions. Treating diabetic mice with a specific sEH inhibitor prevented the pericyte loss and vascular permeability that are characteristic of non-proliferative diabetic retinopathy. Conversely, overexpression of sEH in the retinal Müller glial cells of non-diabetic mice resulted in similar vessel abnormalities to those seen in diabetic mice with retinopathy. Thus, increased expression of sEH is a key determinant in the pathogenesis of diabetic retinopathy, and inhibition of sEH can prevent progression of the disease.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Nucleoside Diphosphate Kinase B Regulates Angiogenesis Through Modulation of Vascular Endothelial Growth Factor Receptor Type 2 and Endothelial Adherens Junction Proteins

Yuxi Feng; Shalini Gross; Nadine M. Wolf; Vicki M. Butenschön; Yi Qiu; Kavi Devraj; Stefan Liebner; Jens Kroll; Edward Y. Skolnik; Hans-Peter Hammes; Thomas Wieland

Objective— Nucleoside diphosphate kinase B (NDPKB) participates in the activation of heterotrimeric and monomeric G proteins, which are pivotal mediators in angiogenic signaling. The role of NDPKB in angiogenesis has to date not been defined. Therefore, we analyzed the contribution of NDPKB to angiogenesis and its underlying mechanisms in well-characterized in vivo and in vitro models. Approach and Results— Zebrafish embryos were depleted of NDPKB by morpholino-mediated knockdown. These larvae displayed severe malformations specifically in vessels formed by angiogenesis. NDPKB-deficient (NDPKB−/−) mice were subjected to oxygen-induced retinopathy. In this model, the number of preretinal neovascularizations in NDPKB−/− mice was strongly reduced in comparison with wild-type littermates. In accordance, a delayed blood flow recovery was detected in the NDPKB−/− mice after hindlimb ligation. In in vitro studies, a small interfering RNA–mediated knockdown of NDPKB was performed in human umbilical endothelial cells. NDPKB depletion impaired vascular endothelial growth factor (VEGF)–induced sprouting and hampered the VEGF-induced spatial redistributions of the VEGF receptor type 2 and VE-cadherin at the plasma membrane. Concomitantly, NDPKB depletion increased the permeability of the human umbilical endothelial cell monolayer. Conclusions— This is the first report to show that NDPKB is required for VEGF-induced angiogenesis and contributes to the correct localization of VEGF receptor type 2 and VE-cadherin at the endothelial adherens junctions. Therefore, our data identify NDPKB as a novel molecular target to modulate VEGF-dependent angiogenesis.


Journal of Alzheimer's Disease | 2013

Increased Efflux of Amyloid-β Peptides through the Blood-Brain Barrier by Muscarinic Acetylcholine Receptor Inhibition Reduces Pathological Phenotypes in Mouse Models of Brain Amyloidosis

Paolo Paganetti; Katia Antoniello; Kavi Devraj; Nicolas Toni; Dairin Kieran; Rime Madani; Maria Pihlgren; Oskar Adolfsson; Wolfgang Froestl; André Schrattenholz; Stefan Liebner; Daniel Havas; Manfred Windisch; John R. Cirrito; Andrea Pfeifer; Andreas Muhs

The formation and accumulation of toxic amyloid-β peptides (Aβ) in the brain may drive the pathogenesis of Alzheimers disease. Accordingly, disease-modifying therapies for Alzheimers disease and related disorders could result from treatments regulating Aβ homeostasis. Examples are the inhibition of production, misfolding, and accumulation of Aβ or the enhancement of its clearance. Here we show that oral treatment with ACI-91 (Pirenzepine) dose-dependently reduced brain Aβ burden in AβPPPS1, hAβPPSL, and AβPP/PS1 transgenic mice. A possible mechanism of action of ACI-91 may occur through selective inhibition of muscarinic acetylcholine receptors (AChR) on endothelial cells of brain microvessels and enhanced Aβ peptide clearance across the blood-brain barrier. One month treatment with ACI-91 increased the clearance of intrathecally-injected Aβ in plaque-bearing mice. ACI-91 also accelerated the clearance of brain-injected Aβ in blood and peripheral tissues by favoring its urinal excretion. A single oral dose of ACI-91 reduced the half-life of interstitial Aβ peptide in pre-plaque mhAβPP/PS1d mice. By extending our studies to an in vitro model, we showed that muscarinic AChR inhibition by ACI-91 and Darifenacin augmented the capacity of differentiated endothelial monolayers for active transport of Aβ peptide. Finally, ACI-91 was found to consistently affect, in vitro and in vivo, the expression of endothelial cell genes involved in Aβ transport across the Blood Brain Brain (BBB). Thus increased Aβ clearance through the BBB may contribute to reduced Aβ burden and associated phenotypes. Inhibition of muscarinic AChR restricted to the periphery may present a therapeutic advantage as it avoids adverse central cholinergic effects.

Collaboration


Dive into the Kavi Devraj's collaboration.

Top Co-Authors

Avatar

Stefan Liebner

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Karl H. Plate

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jadranka Macas

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Rajkumar Vutukuri

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Stefanie Dimmeler

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ingrid Fleming

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Jiong Hu

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Michel Mittelbronn

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge