Network


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

Hotspot


Dive into the research topics where Aysar Alhussaini is active.

Publication


Featured researches published by Aysar Alhussaini.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2012

The monocrotaline model of pulmonary hypertension in perspective

Jose Gomez-Arroyo; Laszlo Farkas; Aysar Alhussaini; Daniela Farkas; Donatas Kraskauskas; Norbert F. Voelkel; Harm J. Bogaard

Severe forms of pulmonary arterial hypertension (PAH) are characterized by various degrees of remodeling of the pulmonary arterial vessels, which increases the pulmonary vascular resistance and right ventricular afterload, thus contributing to the development of right ventricle dysfunction and failure. Recent years have seen advances in the understanding of the pathobiology of PAH; however, many important questions remain unanswered. Elucidating the pathobiology of PAH continues to be critical to design new effective therapeutic strategies, and appropriate animal models of PAH are necessary to achieve the task. Although the monocrotaline rat model of PAH has contributed to a better understanding of vascular remodeling in pulmonary hypertension, we question the validity of this model as a preclinically relevant model of severe plexogenic PAH. Here we review pertinent publications that either have been forgotten or ignored, and we reexamine the monocrotaline model in the context of human forms of PAH.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2011

p53 Gene deficiency promotes hypoxia-induced pulmonary hypertension and vascular remodeling in mice

Shiro Mizuno; H.J. Bogaard; Donatas Kraskauskas; Aysar Alhussaini; Jose Gomez-Arroyo; Norbert F. Voelkel; Takeshi Ishizaki

Chronic hypoxia induces pulmonary arterial remodeling, resulting in pulmonary hypertension and right ventricular hypertrophy. Hypoxia has been implicated as a physiological stimulus for p53 induction and hypoxia-inducible factor-1α (HIF-1α). However, the subcellular interactions between hypoxic exposure and expression of p53 and HIF-1α remain unclear. To examine the role of p53 and HIF-1α expression on hypoxia-induced pulmonary arterial remodeling, wild-type (WT) and p53 knockout (p53KO) mice were exposed to either normoxia or hypoxia for 8 wk. Following chronic hypoxia, both genotypes demonstrated elevated right ventricular pressures, right ventricular hypertrophy as measured by the ratio of the right ventricle to the left ventricle plus septum weights, and vascular remodeling. However, the right ventricular systolic pressures, the ratio of the right ventricle to the left ventricle plus septum weights, and the medial wall thickness of small vessels were significantly greater in the p53KO mice than in the WT mice. The p53KO mice had lower levels of p21 and miR34a expression, and higher levels of HIF-1α, VEGF, and PDGF expression than WT mice following chronic hypoxic exposure. This was associated with a higher proliferating cell nuclear antigen expression of pulmonary artery in p53KO mice. We conclude that p53 plays a critical role in the mitigation of hypoxia-induced small pulmonary arterial remodeling. By interacting with p21 and HIF-1α, p53 may suppress hypoxic pulmonary arterial remodeling and pulmonary arterial smooth muscle cell proliferation under hypoxia.


Chest | 2012

MicroRNA-199a-5p Is Associated With Hypoxia-Inducible Factor-1α Expression in Lungs From Patients With COPD

Shiro Mizuno; Harm J. Bogaard; Jose Gomez-Arroyo; Aysar Alhussaini; Donatas Kraskauskas; Carlyne D. Cool; Norbert F. Voelkel

BACKGROUND MicroRNAs (miRNAs) are small noncoding RNAs that silence target gene expression posttranscriptionally, and their impact on gene expression has been reported in various diseases. It has been reported that the expression of the hypoxia-inducible factor-1α (HIF-1α) is reduced and that of p53 is increased in lungs from patients with COPD. However, the role of miRNAs associated with these genes in lungs from patients with COPD is unknown. METHODS Lung tissue samples from 55 patients were included in this study. Total RNA, miRNA, and protein were extracted from lung tissues and used for reverse transcriptase polymerase chain reaction and Western blot analysis. Cell culture experiments were performed using cultured human pulmonary microvascular endothelial cells (HPMVECs). RESULTS miR-34a and miR-199a-5p expressions were increased, and the phosphorylation of AKT was decreased in the lung tissue samples of patients with COPD. The miR-199a-5p expression was correlated with HIF-1α protein expression in the lungs of patients with COPD. Transfection of HPMVECs with the miR-199a-5p precursor gene decreased HIF-1α protein expression, and transfection with the miR-34a precursor gene increased miR-199a-5p expression. CONCLUSIONS These data suggest that miR-34a and miR-199a-5p contribute to the pathogenesis of COPD, and these miRNAs may also affect the HIF-1α-dependent lung structure maintenance program.


PLOS ONE | 2012

Copper Deficiency Induced Emphysema Is Associated with Focal Adhesion Kinase Inactivation

Shiro Mizuno; Masanori Yasuo; Harm J. Bogaard; Donatas Kraskauskas; Aysar Alhussaini; Jose Gomez-Arroyo; Daniela Farkas; Laszlo Farkas; Norbert F. Voelkel

Background Copper is an important regulator of hypoxia inducible factor 1 alpha (HIF-1α) dependent vascular endothelial growth factor (VEGF) expression, and is also required for the activity of lysyl oxidase (LOX) to effect matrix protein cross-linking. Cell detachment from the extracellular matrix can induce apoptosis (anoikis) via inactivation of focal adhesion kinase (FAK). Methodology To examine the molecular mechanisms whereby copper depletion causes the destruction of the normal alveolar architecture via anoikis, Male Sprague-Dawley rats were fed a copper deficient diet for 6 weeks while being treated with the copper chelator, tetrathiomolybdate. Other groups of rats were treated with the inhibitor of auto-phosphorylation of FAK, 1,2,4,5-benzenetetraamine tetrahydrochloride (1,2,4,5-BT) or FAK small interfering RNA (siRNA). Principal Findings Copper depletion caused emphysematous changes, decreased HIF-1α activity, and downregulated VEGF expression in the rat lungs. Cleaved caspase-3, caspase-8 and Bcl-2 interacting mediator of cell death (Bim) expression was increased, and the phosphorylation of FAK was decreased in copper depleted rat lungs. Administration of 1,2,4,5-BT and FAK siRNA caused emphysematous lung destruction associated with increased expression of cleaved capase-3, caspase-8 and Bim. Conclusions These data indicate that copper-dependent mechanisms contribute to the pathogenesis of emphysema, which may be associated with decreased HIF-1α and FAK activity in the lung.


PLOS ONE | 2014

CXCR4 Inhibition Ameliorates Severe Obliterative Pulmonary Hypertension and Accumulation of C-Kit+ Cells in Rats

Daniela Farkas; Donatas Kraskauskas; Jennifer I. Drake; Aysar Alhussaini; Vita Kraskauskiene; Harm J. Bogaard; Carlyne D. Cool; Norbert F. Voelkel; Laszlo Farkas

Successful curative treatment of severe pulmonary arterial hypertension with luminal obliteration will require a thorough understanding of the mechanism underlying the development and progression of pulmonary vascular lesions. But the cells that obliterate the pulmonary arterial lumen in severe pulmonary arterial hypertension are incompletely characterized. The goal of our study was to evaluate whether inhibition of CXC chemokine receptor 4 will prevent the accumulation of c-kit+ cells and severe pulmonary arterial hypertension. We detected c-kit+ cells expressing endothelial (von Willebrand Factor) or smooth muscle cell/myofibroblast (α-smooth muscle actin) markers in pulmonary arterial lesions of SU5416/chronic hypoxia rats. We found increased expression of CXC chemokine ligand 12 in the lung tissue of SU5416/chronic hypoxia rats. In our prevention study, AMD3100, an inhibitor of the CXC chemokine ligand 12 receptor, CXC chemokine receptor 4, only moderately decreased pulmonary arterial obliteration and pulmonary hypertension in SU5416/chronic hypoxia animals. AMD3100 treatment reduced the number of proliferating c-kit+ α-smooth muscle actin+ cells and pulmonary arterial muscularization and did not affect c-kit+ von Willebrand Factor+ cell numbers. Both c-kit+ cell types expressed CXC chemokine receptor 4. In conclusion, our data demonstrate that in the SU5416/chronic hypoxia model of severe pulmonary hypertension, the CXC chemokine receptor 4-expressing c-kit+ α-smooth muscle actin+ cells contribute to pulmonary arterial muscularization. In contrast, vascular lumen obliteration by c-kit+ von Willebrand Factor+ cells is largely independent of CXC chemokine receptor 4.


american thoracic society international conference | 2012

Inflammation Is Required For The Initiation Of Angioproliferative PAH

Laszlo Farkas; Donatas Kraskauskas; Daniela Farkas; Jose Gomez-Arroyo; Aysar Alhussaini; Ramesh Natarajan; Norbert F. Voelkel


Circulation | 2013

Abstract 9814: Nf-?B Inhibition Prevents Vascular Obliteration and Improves Immune Regulation in Severe Pulmonary Arterial Hypertension

Daniela Farkas; Aysar Alhussaini; Donatas Kraskauskas; Vita Kraskauskiene; Mark R. Nicolls; Norbert F. Voelkel; Ramesh Natarajan; Laszlo Farkas


american thoracic society international conference | 2012

Anti-Proliferative Treatment With Tetrahydromolybdate (TTM) In The SU5416/Chronic Hypoxia Model Restores The Endothelial Monolayer

Laszlo Farkas; Daniela Farkas; Aysar Alhussaini; Donatas Kraskauskas; Harm J. Bogaard; Norbert F. Voelkel


american thoracic society international conference | 2012

Inhibition Of Focal Adhesion Kinase Causes Emphysema In Rat Lung

Shiro Mizuno; Harm J. Bogaard; Donatas Kraskauskas; Aysar Alhussaini; Jose Gomez-Arroyo; Laszlo Farkas; Norbert F. Voelkel


European Respiratory Journal | 2012

Contribution of stem-like cells to angioproliferative pulmonary arterial hypertension in the SU5416/chronic hypoxia model

Laszlo Farkas; Daniela Farkas; Donatas Kraskauskas; Aysar Alhussaini; Vita Kraskauskiene; Harm J. Bogaard; Norbert F. Voelkel

Collaboration


Dive into the Aysar Alhussaini's collaboration.

Top Co-Authors

Avatar

Donatas Kraskauskas

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Norbert F. Voelkel

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Jose Gomez-Arroyo

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Harm J. Bogaard

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Laszlo Farkas

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Shiro Mizuno

Kanazawa Medical University

View shared research outputs
Top Co-Authors

Avatar

Daniela Farkas

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Carlyne D. Cool

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Vita Kraskauskiene

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Ramesh Natarajan

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge