Network


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

Hotspot


Dive into the research topics where Hairu Chen is active.

Publication


Featured researches published by Hairu Chen.


Circulation Research | 2004

Paradoxical cAMP-Induced Lung Endothelial Hyperpermeability Revealed by Pseudomonas aeruginosa ExoY

Sarah Sayner; Dara W. Frank; Judy A. King; Hairu Chen; John VandeWaa; Troy Stevens

Mammalian transmembrane adenylyl cyclases synthesize a restricted plasmalemmal cAMP pool that is intensely endothelial barrier protective. Bacteria have devised mechanisms of transferring eukaryotic factor–dependent adenylyl cyclases into mammalian cells. Pseudomonas aeruginosa ExoY is one such enzyme that catalyzes cytosolic cAMP synthesis, with unknown function. Pseudomonas aeruginosa genetically modified to introduce only the ExoY toxin elevated cAMP 800-fold in pulmonary microvascular endothelial cells over 4 hours, whereas a catalytically deficient (ExoYK81M) strain did not increase cAMP. ExoY-derived cAMP was localized to a cytosolic microdomain not regulated by phosphodiesterase activity. In contrast to the barrier-enhancing actions of plasmalemmal cAMP, the ExoY cytosolic cAMP pool induced endothelial gap formation and increased the filtration coefficient in the isolated perfused lung. These findings collectively illustrate a previously unrecognized mechanism of hyperpermeability induced by rises in cytosolic cAMP.


Circulation Research | 2005

Essential Role of a Ca2+-Selective, Store-Operated Current (ISOC) in Endothelial Cell Permeability. Determinants of the Vascular Leak Site

Songwei Wu; Eugene A. Cioffi; Diego F. Alvarez; Sarah Sayner; Hairu Chen; Donna L. Cioffi; Judy A. King; Judy Creighton; Mary I. Townsley; Steven R. Goodman; Troy Stevens

Store-operated calcium (SOC) entry is sufficient to disrupt the extra-alveolar, but not the alveolar, endothelial cell barrier. Mechanism(s) underlying such insensitivity to transitions in cytosolic calcium ([Ca2+]i) in microvascular endothelial cells are unknown. Depletion of stored Ca2+ activates a larger SOC entry response in extra-alveolar (pulmonary artery; PAECs) than alveolar (pulmonary microvascular; PMVECs) endothelial cells. In vivo permeation studies revealed that Ca2+ store depletion activates similar nonselective cationic conductances in PAECs and PMVECs, while only PAECs possess the calcium-selective, store-operated Ca2+ entry current, ISOC. Pretreatment with the type 4 phosphodiesterase inhibitor, rolipram, abolished thapsigargin-activated ISOC in PAECs, and revealed ISOC in PMVECs. Rolipram pretreatment shifted the thapsigargin-induced fluid leak site from extra-alveolar to alveolar vessels in the intact pulmonary circulation. Thus, our results indicate ISOC provides a [Ca2+]i source that is needed to disrupt the endothelial cell barrier, and demonstrate that intracellular events controlling ISOC activation coordinate the site-specific vascular response to inflammation.


Circulation Research | 2012

Orai1 Determines Calcium Selectivity of an Endogenous TRPC Heterotetramer Channel

Donna L. Cioffi; Songwei Wu; Hairu Chen; Mikhail Alexeyev; Claudette M. St. Croix; Bruce R. Pitt; Stefan Uhlig; Troy Stevens

Rationale: Canonical transient receptor potential 4 (TRPC4) contributes to the molecular composition of a channel encoding for a calcium selective store-operated current, ISOC, whereas Orai1 critically comprises a channel encoding for the highly selective calcium release activated calcium current, ICRAC. However, Orai1 may interact with TRPC proteins and influence their activation and permeation characteristics. Endothelium expresses both TRPC4 and Orai1, and it remains unclear as to whether Orai1 interacts with TRPC4 and contributes to calcium permeation through the TPRC4 channel. Objective: We tested the hypothesis that Orai1 interacts with TRPC4 and contributes to the channels selective calcium permeation important for endothelial barrier function. Methods and Results: A novel method to purify the endogenous TRPC4 channel and probe for functional interactions was developed, using TRPC4 binding to protein 4.1 as bait. Isolated channel complexes were conjugated to anti-TRPC protein antibodies labeled with cy3-cy5 pairs. Förster Resonance Energy Transfer among labeled subunits revealed the endogenous protein alignment. One TRPC1 and at least 2 TRPC4 subunits constituted the endogenous channel (TRPC1/4). Orai1 interacted with TRPC4. Conditional Orai1 knockdown reduced the probability for TRPC1/4 channel activation and converted it from a calcium-selective to a nonselective channel, an effect that was rescued on Orai1 reexpression. Loss of Orai1 improved endothelial cell barrier function. Conclusion: Orai1 interacts with TRPC4 in the endogenous channel complex, where it controls TRPC1/4 activation and channel permeation characteristics, including calcium selectivity, important for control of endothelial cell barrier function.


Journal of Biological Chemistry | 2007

Microtubule Motors Regulate ISOC Activation Necessary to Increase Endothelial Cell Permeability

Songwei Wu; Hairu Chen; Mikhail Alexeyev; Judy A. King; Timothy M. Moore; Troy Stevens; Ronald Balczon

Calcium store depletion activates multiple ion channels, including calcium-selective and nonselective channels. Endothelial cells express TRPC1 and TRPC4 proteins that contribute to a calcium-selective store-operated current, ISOC. Whereas thapsigargin activates the ISOC in pulmonary artery endothelial cells (PAECs), it does not activate ISOC in pulmonary microvascular endothelial cells (PMVECs), despite inducing a significant rise in global cytosolic calcium. Endoplasmic reticulum exhibits retrograde distribution in PMVECs when compared with PAECs. We therefore sought to determine whether endoplasmic reticulum-to-plasma membrane coupling represents an important determinant of ISOC activation in PAECs and PMVECs. Endoplasmic reticulum organization is controlled by microtubules, because nocodozole induced microtubule disassembly and caused retrograde endoplasmic reticulum collapse in PMVECs. In PMVECs, rolipram treatment produced anterograde endoplasmic reticulum distribution and revealed a thapsigargin-activated ISOC that was abolished by nocodozole and taxol. Microtubule motors control organelle distribution along microtubule tracks, with the dynein motor causing retrograde movement and the kinesin motor causing anterograde movement. Dynamitin expression reduces dynein motor function inducing anterograde endoplasmic reticulum transport, which allows for direct activation of ISOC by thapsigargin in PMVECs. In contrast, expression of dominant negative kinesin light chain reduces kinesin motor function and induces retrograde endoplasmic reticulum transport; dominant negative kinesin light chain expression prevented the direct activation of ISOC by thapsigargin in PAECs. ISOC activation is an important step leading to disruption of cell-cell adhesion and increased macromolecular permeability. Thus, microtubule motor function plays an essential role in activating cytosolic calcium transitions through the membrane ISOC channel leading to endothelial barrier disruption.


Molecular Cancer | 2010

Mechanisms of transcriptional regulation and prognostic significance of activated leukocyte cell adhesion molecule in cancer.

Judy A. King; Fang Tan; Flaubert Mbeunkui; Zachariah Chambers; Sarah Cantrell; Hairu Chen; Diego F. Alvarez; Lalita A. Shevde; Solomon F. Ofori-Acquah

BackgroundActivated leukocyte cell adhesion molecule (ALCAM) is implicated in the prognosis of multiple cancers with low level expression associated with metastasis and early death in breast cancer. Despite this significance, mechanisms that regulate ALCAM gene expression and ALCAMs role in adhesion of pre-metastatic circulating tumor cells have not been defined. We studied ALCAM expression in 20 tumor cell lines by real-time PCR, western blot and immunochemistry. Epigenetic alterations of the ALCAM promoter were assessed using methylation-specific PCR and bisulfite sequencing. ALCAMs role in adhesion of tumor cells to the vascular wall was studied in isolated perfused lungs.ResultsA common site for transcription initiation of the ALCAM gene was identified and the ALCAM promoter sequenced. The promoter contains multiple cis-active elements including a functional p65 NF-κB motif, and it harbors an extensive array of CpG residues highly methylated exclusively in ALCAM-negative tumor cells. These CpG residues were modestly demethylated after 5-aza-2-deoxycytidine treatment. Restoration of high-level ALCAM expression using an ALCAM cDNA increased clustering of MDA-MB-435 tumor cells perfused through the pulmonary vasculature of ventilated rat lungs. Anti-ALCAM antibodies reduced the number of intravascular tumor cell clusters.ConclusionOur data suggests that loss of ALCAM expression, due in part to DNA methylation of extensive segments of the promoter, significantly impairs the ability of circulating tumor cells to adhere to each other, and may therefore promote metastasis. These findings offer insight into the mechanisms for down-regulation of ALCAM gene expression in tumor cells, and for the positive prognostic value of high-level ALCAM in breast cancer.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2010

α1G T-type calcium channel selectively regulates P-selectin surface expression in pulmonary capillary endothelium

Chun Zhou; Hairu Chen; Judy A. King; Hassan Sellak; Wolfgang M. Kuebler; Jun Yin; Mary I. Townsley; Hee-Sup Shin; Songwei Wu

Regulated P-selectin surface expression provides a rapid measure for endothelial transition to a proinflammatory phenotype. In general, P-selectin surface expression results from Weibel-Palade body (WPb) exocytosis. Yet, it is unclear whether pulmonary capillary endothelium possesses WPbs or regulated P-selectin surface expression and, if so, how inflammatory stimuli initiate exocytosis. We used immunohistochemistry, immunofluorescence labeling, ultrastructural assessment, and an isolated perfused lung model to demonstrate that capillary endothelium lacks WPbs but possesses P-selectin. Thrombin stimulated P-selectin surface expression in both extra-alveolar vessel and alveolar capillary endothelium. Only in capillaries was the thrombin-stimulated P-selectin surface expression considerably mitigated by pharmacologic blockade of the T-type channel or genetic knockout of the T-type channel alpha(1G)-subunit. Depolarization of endothelial plasma membrane via high K(+) perfusion capable of eliciting cytosolic Ca(2+) transients also provoked P-selectin surface expression in alveolar capillaries that was abolished by T-type channel blockade or alpha(1G) knockout. Our findings reveal an intracellular WPb-independent P-selectin pool in pulmonary capillary endothelium, where the regulated P-selectin surface expression is triggered by Ca(2+) transients evoked through activation of the alpha(1G) T-type channel.


Circulation Research | 2005

Essential Role of a Ca 2+ -Selective, Store-Operated Current ( I SOC ) in Endothelial Cell Permeability

Songwei Wu; Eugene A. Cioffi; Diego F. Alvarez; Sarah Sayner; Hairu Chen; Donna L. Cioffi; Judy A. King; Judy Creighton; Mary I. Townsley; Steven R. Goodman; Troy Stevens

Store-operated calcium (SOC) entry is sufficient to disrupt the extra-alveolar, but not the alveolar, endothelial cell barrier. Mechanism(s) underlying such insensitivity to transitions in cytosolic calcium ([Ca2+]i) in microvascular endothelial cells are unknown. Depletion of stored Ca2+ activates a larger SOC entry response in extra-alveolar (pulmonary artery; PAECs) than alveolar (pulmonary microvascular; PMVECs) endothelial cells. In vivo permeation studies revealed that Ca2+ store depletion activates similar nonselective cationic conductances in PAECs and PMVECs, while only PAECs possess the calcium-selective, store-operated Ca2+ entry current, ISOC. Pretreatment with the type 4 phosphodiesterase inhibitor, rolipram, abolished thapsigargin-activated ISOC in PAECs, and revealed ISOC in PMVECs. Rolipram pretreatment shifted the thapsigargin-induced fluid leak site from extra-alveolar to alveolar vessels in the intact pulmonary circulation. Thus, our results indicate ISOC provides a [Ca2+]i source that is needed to disrupt the endothelial cell barrier, and demonstrate that intracellular events controlling ISOC activation coordinate the site-specific vascular response to inflammation.


Circulation Research | 2005

Essential Role of a Ca2+-Selective, Store-Operated Current (ISOC) in Endothelial Cell Permeability

Songwei Wu; Eugene A. Cioffi; Diego F. Alvarez; Sarah Sayner; Hairu Chen; Donna L. Cioffi; Judy A. King; Judy Creighton; Mary I. Townsley; Steven R. Goodman; Troy Stevens

Store-operated calcium (SOC) entry is sufficient to disrupt the extra-alveolar, but not the alveolar, endothelial cell barrier. Mechanism(s) underlying such insensitivity to transitions in cytosolic calcium ([Ca2+]i) in microvascular endothelial cells are unknown. Depletion of stored Ca2+ activates a larger SOC entry response in extra-alveolar (pulmonary artery; PAECs) than alveolar (pulmonary microvascular; PMVECs) endothelial cells. In vivo permeation studies revealed that Ca2+ store depletion activates similar nonselective cationic conductances in PAECs and PMVECs, while only PAECs possess the calcium-selective, store-operated Ca2+ entry current, ISOC. Pretreatment with the type 4 phosphodiesterase inhibitor, rolipram, abolished thapsigargin-activated ISOC in PAECs, and revealed ISOC in PMVECs. Rolipram pretreatment shifted the thapsigargin-induced fluid leak site from extra-alveolar to alveolar vessels in the intact pulmonary circulation. Thus, our results indicate ISOC provides a [Ca2+]i source that is needed to disrupt the endothelial cell barrier, and demonstrate that intracellular events controlling ISOC activation coordinate the site-specific vascular response to inflammation.


Microscopy and Microanalysis | 2008

Lung Endothelial Cells Express ALCAM on Released Exosomes/Microparticles

Judy A. King; E Syklawer; Hairu Chen; J Resmondo; F McDonald; Troy Stevens; Lalita A. Shevde; S Ofori-Acquah; Timothy M. Moore; Natalie N. Bauer


Microscopy Today | 2009

Microparticles/Exosomes: Isolation and TEM Analysis

Natalie N. Bauer; Jyoti Rai; Hairu Chen; Lillianne G. Harris; Lalita A. Shevde; Timothy M. Moore; Judy A. King

Collaboration


Dive into the Hairu Chen's collaboration.

Top Co-Authors

Avatar

Judy A. King

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Songwei Wu

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Troy Stevens

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Donna L. Cioffi

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Mary I. Townsley

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Diego F. Alvarez

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Timothy M. Moore

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Lalita A. Shevde

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Mikhail Alexeyev

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Sarah Sayner

University of South Alabama

View shared research outputs
Researchain Logo
Decentralizing Knowledge