Network


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

Hotspot


Dive into the research topics where Mack H. Wu is active.

Publication


Featured researches published by Mack H. Wu.


Expert Reviews in Molecular Medicine | 2009

Molecular mechanisms of endothelial hyperpermeability: implications in inflammation

Puneet Kumar; Qiang Shen; Christopher D. Pivetti; Eugene S. Lee; Mack H. Wu; Sarah Y. Yuan

Endothelial hyperpermeability is a significant problem in vascular inflammation associated with trauma, ischaemia-reperfusion injury, sepsis, adult respiratory distress syndrome, diabetes, thrombosis and cancer. An important mechanism underlying this process is increased paracellular leakage of plasma fluid and protein. Inflammatory stimuli such as histamine, thrombin, vascular endothelial growth factor and activated neutrophils can cause dissociation of cell-cell junctions between endothelial cells as well as cytoskeleton contraction, leading to a widened intercellular space that facilitates transendothelial flux. Such structural changes initiate with agonist-receptor binding, followed by activation of intracellular signalling molecules including calcium, protein kinase C, tyrosine kinases, myosin light chain kinase, and small Rho-GTPases; these kinases and GTPases then phosphorylate or alter the conformation of different subcellular components that control cell-cell adhesion, resulting in paracellular hypermeability. Targeting key signalling molecules that mediate endothelial-junction-cytoskeleton dissociation demonstrates a therapeutic potential to improve vascular barrier function during inflammatory injury.


Cardiovascular Research | 2010

Myosin Light Chain Kinase in Microvascular Endothelial Barrier Function

Qiang Shen; Robert R. Rigor; Christopher D. Pivetti; Mack H. Wu; Sarah Y. Yuan

Microvascular barrier dysfunction is implicated in the initiation and progression of inflammation, posttraumatic complications, sepsis, ischaemia-reperfusion injury, atherosclerosis, and diabetes. Under physiological conditions, a precise equilibrium between endothelial cell-cell adhesion and actin-myosin-based centripetal tension tightly controls the semi-permeability of microvascular barriers. Myosin light chain kinase (MLCK) plays an important role in maintaining the equilibrium by phosphorylating myosin light chain (MLC), thereby inducing actomyosin contractility and weakening endothelial cell-cell adhesion. MLCK is activated by numerous physiological factors and inflammatory or angiogenic mediators, causing vascular hyperpermeability. In this review, we discuss experimental evidence supporting the crucial role of MLCK in the hyperpermeability response to key cell signalling events during inflammation. At the cellular level, in vitro studies of cultured endothelial monolayers treated with MLCK inhibitors or transfected with specific inhibiting peptides have demonstrated that induction of endothelial MLCK activity is necessary for hyperpermeability. Ex vivo studies of live microvessels, enabled by development of the isolated, perfused venule method, support the importance of MLCK in endothelial permeability regulation in an environment that more closely resembles in vivo tissues. Finally, the role of MLCK in vascular hyperpermeability has been confirmed with in vivo studies of animal disease models and the use of transgenic MLCK210 knockout mice. These approaches provide a more complete view of the role of MLCK in vascular barrier dysfunction.


Microcirculation | 2006

Rho and ROCK Signaling in VEGF-Induced Microvascular Endothelial Hyperpermeability

Hengrui Sun; Jerome W. Breslin; Jun Zhu; Sarah Y. Yuan; Mack H. Wu

Objectives: Vascular endothelial growth factor (VEGF) plays an important role in the regulation of microvascular permeability under various physiological and pathological conditions. The authors tested the hypothesis that the small GTPase Rho and its downstream effector ROCK (Rho‐associated coiled‐coil‐containing protein kinase) mediate VEGF‐induced increases in venular permeability. They also investigated myosin light chain (MLC) phosphorylation and actin polymerization, two well‐characterized targets of the Rho‐ROCK pathway that are implicated in the regulation of endothelial barrier function.


Circulation Research | 2002

Myosin Light Chain Phosphorylation in Neutrophil-Stimulated Coronary Microvascular Leakage

Sarah Y. Yuan; Mack H. Wu; Elena E. Ustinova; Mingzhang Guo; John H. Tinsley; Primal de Lanerolle; Wenjuan Xu

Neutrophil-induced coronary microvascular leakage represents an important pathophysiological consequence of ischemic and inflammatory heart diseases. The precise mechanism by which neutrophils regulate endothelial barrier function remains to be established. The aim of this study was to examine the microvascular endothelial response to neutrophil activation with a focus on myosin light chain kinase (MLCK)-mediated myosin light chain (MLC) phosphorylation, a regulatory process that controls cell contraction. The apparent permeability coefficient of albumin (Pa) was measured in intact isolated porcine coronary venules. Incubation of the vessels with C5a-activated neutrophils induced a time- and concentration-dependent increase in Pa. The hyperpermeability response was significantly attenuated during inhibition of endothelial MLC phosphorylation with the selective MLCK inhibitor ML-7 and transfection of a specific MLCK-inhibiting peptide. In contrast, transfection of constitutively active MLCK elevated Pa, which was abolished by ML-7. In addition to the vessel study, albumin transendothelial flux was measured in cultured bovine coronary venular endothelial monolayers, which displayed a hyperpermeability response to neutrophils and MLCK in a pattern similar to that in venules. Importantly, neutrophil stimulation caused MLC phosphorylation in endothelial cells in a time course closely correlated with that of the hyperpermeability response. Consistently, the MLCK inhibitors abolished neutrophil-induced MLC phosphorylation. Furthermore, immunohistochemical observation of neutrophil-stimulated endothelial cells revealed an increased staining for phosphorylated MLC in association with contractile stress fiber formation and intercellular gap development. Taken together, the results suggest that endothelial MLCK activation and MLC phosphorylation play an important role in mediating endothelial barrier dysfunction during neutrophil activation.


The Journal of Physiology | 2005

Endothelial focal adhesions and barrier function.

Mack H. Wu

Focal adhesions composed of integrins provide an important structural basis for anchoring the endothelial lining to its surrounding matrices in the vascular wall. Complex molecular reactions occur at the endothelial cell–matrix contact sites in response to physical and chemical stress present in the circulatory system. Recent experimental evidence points to the importance of focal adhesions in the regulation of microvascular barrier function. On one hand, the adhesive interaction between integrins and their extracellular ligands is essential to the maintenance of endothelial barrier properties, and interruption of integrin–matrix binding leads to leaky microvessels. On the other hand, focal adhesion assembly and activation serve as important signalling events in modulating endothelial permeability under stimulatory conditions in the presence of angiogenic factors, inflammatory mediators, or physical forces. The focal responses show distinctive patterns with different temporal characteristics, whereas focal adhesion kinase (FAK) plays a central role in initiating and integrating various signalling pathways that ultimately affect the barrier function. The molecular basis of focal adhesion‐dependent microvascular permeability is currently under investigation, and advances in the technologies of computerized quantitative microscopy and intact microvessel imaging should aid the establishment of a functional significance for focal adhesions in the physiological regulation of microvascular permeability.


Circulation | 2011

Nonmuscle Myosin Light-Chain Kinase Deficiency Attenuates Atherosclerosis in Apolipoprotein E–Deficient Mice via Reduced Endothelial Barrier Dysfunction and Monocyte Migration

Chongxiu Sun; Mack H. Wu; Sarah Y. Yuan

Background— Endothelial dysfunction and monocyte migration are key events in the pathogenesis of atherosclerosis. Nonmuscle myosin light-chain kinase (nmMLCK), the predominant MLCK isoform in endothelial cells, has been shown to contribute to vascular inflammation by altering endothelial barrier function. However, its impact on atherogenesis remains unknown. Methods and Results— We investigated the role of nmMLCK in the development of atherosclerotic lesions in apolipoprotein E–deficient (apoE−/−) mice fed an atherogenic diet for 12 weeks. Histopathological examination demonstrated that nmMLCK deficiency (apoE−/−nmmlck−/−) reduced the size of aortic lesions by 53%, lipid contents by 44%, and macrophage deposition by 40%. Western blotting and reverse-transcription polymerase chain reaction revealed the expression of nmMLCK in aortic endothelial cells and peripheral blood monocytes. Measurements of transendothelial electric resistance indicated that nmMLCK deficiency attenuated endothelial barrier dysfunction caused by thrombin, oxidized low-density lipoprotein, and tumor necrosis factor &agr;. In monocytes, nmMLCK deficiency reduced their migration in response to the chemokine monocyte chemoattractant protein-1. Further mechanistic studies showed that nmMLCK acted through both myosin light chain phosphorylation-coupled and -uncoupled pathways; the latter involved Rous sacracoma virus homolog genes-encoded tyrosine kinases (Src) signaling. Moreover, depletion of Src via gene silencing, site-specific mutagenesis, or pharmacological inhibition of Src greatly attenuated nmMLCK-dependent endothelial barrier dysfunction and monocyte migration. Conclusions— Nonmuscle myosin light-chain kinase contributes to atherosclerosis by regulating endothelial barrier function and monocyte migration via mechanisms involving not only kinase-mediated MLC phosphorylation but also Src activation.


Journal of Bacteriology | 2007

VapC-1 of Nontypeable Haemophilus influenzae Is a Ribonuclease

Dayle A. Daines; Mack H. Wu; Sarah Y. Yuan

Nontypeable Haemophilus influenzae (NTHi) organisms are obligate parasites of the human upper respiratory tract that can exist as commensals or pathogens. Toxin-antitoxin (TA) loci are highly conserved gene pairs that encode both a toxin and antitoxin moiety. Seven TA gene families have been identified to date, and NTHi carries two alleles of the vapBC family. Here, we have characterized the function of one of the NTHi alleles, vapBC-1. The gene pair is transcribed as an operon in two NTHi clinical isolates, and promoter fusions display an inverse relationship to culture density. The antitoxin VapB-1 forms homomultimers both in vitro and in vivo. The expression of the toxin VapC-1 conferred growth inhibition to an Escherichia coli expression strain and was successfully purified only when cloned in tandem with its cognate antitoxin. Using total RNA isolated from both E. coli and NTHi, we show for the first time that VapC-1 is an RNase that is active on free RNA but does not degrade DNA in vitro. Preincubation of the purified toxin and antitoxin together results in the formation of a protein complex that abrogates the activity of the toxin. We conclude that the NTHi vapBC-1 gene pair functions as a classical TA locus and that the induction of VapC-1 RNase activity leads to growth inhibition via the mechanism of mRNA cleavage.


Microcirculation | 2007

Microvascular Permeability in Diabetes and Insulin Resistance

Sarah Y. Yuan; Jerome W. Breslin; Rachel M. Perrin; Nathalie Gaudreault; Mingzhang Guo; Hamed Kargozaran; Mack H. Wu

ABSTRACT


Cardiovascular Research | 2010

ADAM15 regulates endothelial permeability and neutrophil migration via Src/ERK1/2 signalling

Chongxiu Sun; Mack H. Wu; Mingzhang Guo; Mark L. Day; Eugene S. Lee; Sarah Y. Yuan

AIMS Endothelial barrier dysfunction is a key event in the pathogenesis of vascular diseases associated with inflammation. ADAM (a disintegrin and metalloprotease) 15 has been shown to contribute to the development of vascular inflammation. However, its role in regulating endothelial barrier function is unknown. The aim of this study was to examine the effect of ADAM15 on endothelial permeability and its underlying mechanisms. METHODS AND RESULTS By measuring albumin transendothelial flux and transendothelial electric resistance in cultured human umbilical vein endothelial cell monolayers, we found that depletion of ADAM15 expression via siRNA decreased endothelial permeability and attenuated thrombin-induced barrier dysfunction. In contrast, endothelial cells overexpressing either wild-type or catalytically dead mutant ADAM15 displayed a higher basal permeability and augmented hyperpermeability in response to thrombin. In addition, ADAM15 knockdown inhibited whereas ADAM15 overexpression promoted neutrophil transendothelial migration. Further molecular assays revealed that ADAM15 did not cleave vascular endothelial-cadherin or cause its degradation. However, overexpression of ADAM15 promoted extracellular signal-regulated kinase (ERK)1/2 phosphorylation in both non-stimulated and thrombin-stimulated endothelial cells in a protease activity-independent manner. Pharmacological inhibition of Src kinase or ERK activation reversed ADAM15-induced hyperpermeability and neutrophil transmigration. CONCLUSION The data provide evidence for a novel function of ADAM15 in regulating endothelial barrier properties. The mechanisms of ADAM15-induced hyperpermeability involve Src/ERK1/2 signalling independent of junction molecule shedding.


Medicinal Research Reviews | 2013

Myosin Light Chain Kinase Signaling in Endothelial Barrier Dysfunction

Robert R. Rigor; Qiang Shen; Christopher D. Pivetti; Mack H. Wu; Sarah Y. Yuan

Microvascular barrier dysfunction is a serious problem that occurs in many inflammatory conditions, including sepsis, trauma, ischemia–reperfusion injury, cardiovascular disease, and diabetes. Barrier dysfunction permits extravasation of serum components into the surrounding tissue, leading to edema formation and organ failure. The basis for microvascular barrier dysfunction is hyperpermeability at endothelial cell–cell junctions. Endothelial hyperpermeability is increased by actomyosin contractile activity in response to phosphorylation of myosin light chain by myosin light chain kinase (MLCK). MLCK‐dependent endothelial hyperpermeability occurs in response to inflammatory mediators (e.g., activated neutrophils, thrombin, histamine, tumor necrosis factor alpha, etc.), through multiple cell signaling pathways and signaling molecules (e.g., Ca++, protein kinase C, Src kinase, nitric oxide synthase, etc.). Other signaling molecules protect against MLCK‐dependent hyperpermeability (e.g., sphingosine‐1‐phosphate or cAMP). In addition, individual MLCK isoforms play specific roles in endothelial barrier dysfunction, suggesting that isoform‐specific inhibitors could be useful for treating inflammatory disorders and preventing multiple organ failure. Because endothelial barrier dysfunction depends upon signaling through MLCK in many instances, MLCK‐dependent signaling comprises multiple potential therapeutic targets for preventing edema formation and multiple organ failure. The following review is a discussion of MLCK‐dependent mechanisms and cell signaling events that mediate endothelial hyperpermeability.

Collaboration


Dive into the Mack H. Wu's collaboration.

Top Co-Authors

Avatar

Sarah Y. Yuan

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Mingzhang Guo

University of California

View shared research outputs
Top Co-Authors

Avatar

Jerome W. Breslin

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Qiang Shen

University of California

View shared research outputs
Top Co-Authors

Avatar

Richard S. Beard

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Eugene S. Lee

University of California

View shared research outputs
Top Co-Authors

Avatar

Ricci J. Haines

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chongxiu Sun

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge