Network


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

Hotspot


Dive into the research topics where Steven K. Huang is active.

Publication


Featured researches published by Steven K. Huang.


The FASEB Journal | 2009

Prostaglandin E2 induces fibroblast apoptosis by modulating multiple survival pathways

Steven K. Huang; Eric S. White; Scott H. Wettlaufer; Heather Grifka; Cory M. Hogaboam; Victor J. Thannickal; Jeffrey C. Horowitz; Marc Peters-Golden

Although the lipid mediator prostaglandin E2 (PGE2) exerts antifibrotic effects by inhibiting multiple fibroblast functions, its ability to regulate fibroblast survival is unknown. Here, we examined the effects of this prostanoid on apoptosis and apoptosis pathways in normal and fibrotic lung fibroblasts. As compared to medium alone, 24 h of treatment with PGE2 increased apoptosis of normal lung fibroblasts in a dose‐dependent manner (EC50~50 nM), as measured by annexin V staining, caspase 3 activity, cleavage of poly‐ADP‐ribose polymerase, and single‐stranded DNA levels. PGE2 also potentiated apoptosis elicited by Fas ligand plus cycloheximide. These proapoptotic actions were dependent on signaling through the EP2/EP4 receptors and by downstream activation of both caspases 8 and 9. Silencing and gene deletion of PTEN demonstrated that the effects of PGE2 involved decreased activity of the prosurvival molecule Akt. PGE2 also down‐regulated expression of survivin, an inhibitor of apoptosis, and increased expression of Fas. Fibroblasts from patients with pulmonary fibrosis exhibited resistance to the apoptotic effects of PGE2. These findings show for the first time that, in contrast to its effects on many other cell types, PGE2 promotes apoptosis in lung fibroblasts through diverse pathways. They provide another dimension by which PGE2 may inhibit, and perhaps even reverse, fibrogenesis in patients with interstitial lung disease.—Huang, S. K., White, E. S., Wettlaufer, S. H., Grifka, H., Hogaboam, C. M., Thannickal, V. J., Horowitz, J. C., Peters‐Golden, M. Prostaglandin E2 induces fibroblast apoptosis by modulating multiple survival pathways. FASEB J. 23, 4317–4326 (2009). www.fasebj.org


Journal of Clinical Investigation | 2010

The antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice

Kristy A. Bauman; Scott H. Wettlaufer; Katsuhide Okunishi; Kevin M. Vannella; Joshua S. Stoolman; Steven K. Huang; Anthony J. Courey; Eric S. White; Cory M. Hogaboam; Richard Simon; Galen B. Toews; Thomas H. Sisson; Bethany B. Moore; Marc Peters-Golden

Plasminogen activation to plasmin protects from lung fibrosis, but the mechanism underlying this antifibrotic effect remains unclear. We found that mice lacking plasminogen activation inhibitor-1 (PAI-1), which are protected from bleomycin-induced pulmonary fibrosis, exhibit lung overproduction of the antifibrotic lipid mediator prostaglandin E2 (PGE2). Plasminogen activation upregulated PGE2 synthesis in alveolar epithelial cells, lung fibroblasts, and lung fibrocytes from saline- and bleomycin-treated mice, as well as in normal fetal and adult primary human lung fibroblasts. This response was exaggerated in cells from Pai1-/- mice. Although enhanced PGE2 formation required the generation of plasmin, it was independent of proteinase-activated receptor 1 (PAR-1) and instead reflected proteolytic activation and release of HGF with subsequent induction of COX-2. That the HGF/COX-2/PGE2 axis mediates in vivo protection from fibrosis in Pai1-/- mice was demonstrated by experiments showing that a selective inhibitor of the HGF receptor c-Met increased lung collagen to WT levels while reducing COX-2 protein and PGE2 levels. Of clinical interest, fibroblasts from patients with idiopathic pulmonary fibrosis were found to be defective in their ability to induce COX-2 and, therefore, unable to upregulate PGE2 synthesis in response to plasmin or HGF. These studies demonstrate crosstalk between plasminogen activation and PGE2 generation in the lung and provide a mechanism for the well-known antifibrotic actions of the fibrinolytic pathway.


Cell Death and Disease | 2013

Histone modifications are responsible for decreased Fas expression and apoptosis resistance in fibrotic lung fibroblasts

Steven K. Huang; Anne M. Scruggs; J Donaghy; Jeffrey C. Horowitz; Zbigniew Zasłona; S Przybranowski; Eric S. White; Marc Peters-Golden

Although the recruitment of fibroblasts to areas of injury is critical for wound healing, their subsequent apoptosis is necessary in order to prevent excessive scarring. Fibroproliferative diseases, such as pulmonary fibrosis, are often characterized by fibroblast resistance to apoptosis, but the mechanism(s) for this resistance remains elusive. Here, we employed a murine model of pulmonary fibrosis and cells from patients with idiopathic pulmonary fibrosis (IPF) to explore epigenetic mechanisms that may be responsible for the decreased expression of Fas, a cell surface death receptor whose expression has been observed to be decreased in pulmonary fibrosis. Murine pulmonary fibrosis was elicited by intratracheal injection of bleomycin. Fibroblasts cultured from bleomycin-treated mice exhibited decreased Fas expression and resistance to Fas-mediated apoptosis compared with cells from saline-treated control mice. Although there were no differences in DNA methylation, the Fas promoter in fibroblasts from bleomycin-treated mice exhibited decreased histone acetylation and increased histone 3 lysine 9 trimethylation (H3K9Me3). This was associated with increased histone deacetylase (HDAC)-2 and HDAC4 expression. Treatment with HDAC inhibitors increased Fas expression and restored susceptibility to Fas-mediated apoptosis. Fibroblasts from patients with IPF likewise exhibited decreased histone acetylation and increased H3K9Me3 at the Fas promoter and increased their expression of Fas in the presence of an HDAC inhibitor. These findings demonstrate the critical role of histone modifications in the development of fibroblast resistance to apoptosis in both a murine model and in patients with pulmonary fibrosis and suggest novel approaches to therapy for progressive fibroproliferative disorders.


Chest | 2008

Eicosanoid lipid mediators in fibrotic lung diseases: ready for prime time?

Steven K. Huang; Marc Peters-Golden

Recognition of a pivotal role for eicosanoids in both normal and pathologic fibroproliferation is long overdue. These lipid mediators have the ability to regulate all cell types and nearly all pathways relevant to fibrotic lung disorders. Abnormal fibroproliferation is characterized by an excess of profibrotic leukotrienes and a deficiency of antifibrotic prostaglandins. The relevance of an eicosanoid imbalance is pertinent to diseases involving the parenchymal, airway, and vascular compartments of the lung, and is supported by studies conducted both in humans and animal models. Given the lack of effective alternatives, and the existing and emerging options for therapeutic targeting of eicosanoids, such treatments are ready for prime time.


American Journal of Respiratory Cell and Molecular Biology | 2013

Reversal of Myofibroblast Differentiation by Prostaglandin E2

Garth Garrison; Steven K. Huang; Katsuhide Okunishi; Jacob P. Scott; Loka Raghu Kumar Penke; Anne M. Scruggs; Marc Peters-Golden

Differentiation of fibroblasts into α-smooth muscle actin (SMA)-expressing myofibroblasts represents a critical step in the pathogenesis of fibrotic disorders, and is generally regarded as irreversible. Prostaglandin E2 (PGE2) has been shown to prevent multiple aspects of fibroblast activation, including the differentiation of fibroblasts to myofibroblasts. Here, we investigated its ability to reverse this differentiated phenotype. Fetal and adult lung fibroblasts were induced to differentiate into myofibroblasts by 24-hour culture with transforming growth factor (TGF)-β1 or endothelin-1. Cells were then treated without or with PGE2 for various intervals and assessed for α-SMA expression. In the absence of PGE2 treatment, α-SMA expression induced by TGF-β1 was persistent and stable for up to 8 days. By contrast, PGE2 treatment effected a dose-dependent decrease in α-SMA and collagen I expression that was observed 2 days after PGE2 addition, peaked at 3 days, and persisted through 8 days in culture. This effect was not explained by an increase in myofibroblast apoptosis, and indeed, reintroduction of TGF-β1 2 days after addition of PGE2 prompted dedifferentiated fibroblasts to re-express α-SMA, indicating redifferentiation to myofibroblasts. This effect of PGE2 was associated with inhibition of focal adhesion kinase signaling, and a focal adhesion kinase inhibitor was also capable of reversing myofibroblast phenotype. These data unambiguously demonstrate reversal of established myofibroblast differentiation. Because many patients have established or even advanced fibrosis by the time they seek medical attention, this capacity of PGE2 has the potential to be harnessed for therapy of late-stage fibrotic disorders.


Chest | 2008

Translating Basic Research into Clinical PracticeEicosanoid Lipid Mediators in Fibrotic Lung Diseases: Ready for Prime Time?

Steven K. Huang; Marc Peters-Golden

Recognition of a pivotal role for eicosanoids in both normal and pathologic fibroproliferation is long overdue. These lipid mediators have the ability to regulate all cell types and nearly all pathways relevant to fibrotic lung disorders. Abnormal fibroproliferation is characterized by an excess of profibrotic leukotrienes and a deficiency of antifibrotic prostaglandins. The relevance of an eicosanoid imbalance is pertinent to diseases involving the parenchymal, airway, and vascular compartments of the lung, and is supported by studies conducted both in humans and animal models. Given the lack of effective alternatives, and the existing and emerging options for therapeutic targeting of eicosanoids, such treatments are ready for prime time.


The FASEB Journal | 2012

Prostaglandin E2 increases fibroblast gene-specific and global DNA methylation via increased DNA methyltransferase expression

Steven K. Huang; Anne M. Scruggs; Jake Donaghy; Richard C. McEachin; Aaron S. Fisher; Bruce C. Richardson; Marc Peters-Golden

Although alterations in DNA methylation patterns have been associated with specific diseases and environmental exposures, the mediators and signaling pathways that direct these changes remain understudied. The bioactive lipid mediator prostaglandin E2 (PGE2) has been shown to exert a myriad of effects on cell survival, proliferation, and differentiation. Here, we report that PGE2 also signals to increase global DNA methylation and DNA methylation machinery in fibro‐blasts. HumanMethylation27 BeadChip array analysis of primary fetal (IMR‐90) and adult lung fibroblasts identified multiple genes that were hypermethylated in response to PGE2. PGE2, compared with nontreated controls, increased expression and activity (EC50~107 M) of one specific isoform of DNA methyltransferase, DNMT3a. Silencing of DNMT3a negated the ability of PGE2 to increase DNMT activity. The increase in DNMT3a expression was mediated by PGE2 signaling via its E prostanoid 2 receptor and the second messenger cAMP. PGE2, compared with the untreated control, increased the expression and activity of Sp1 and Sp3 (EC50~3×107 M), transcription factors known to increase DNMT3a expression, and inhibition of these transcription factors abrogated the PGE2 increase of DNMT3a expression. These findings were specific to fibroblasts, as PGE2 decreased DNMT1 and DNMT3a expression in RAW macrophages. Taken together, these findings establish that DNA methylation is regulated by a ubiquitous bioactive endogenous mediator. Given that PGE2 biosynthesis is modulated by environmental toxins, various disease states, and commonly used pharmacological agents, these findings uncover a novel mechanism by which alterations in DNA methylation patterns may arise in association with disease and certain environmental exposures.—Huang, S. K., Scruggs, A. M., Donaghy, J., McEachin, R. C., Fisher, A. S., Richardson, B. C., Peters‐Golden, M. Prostaglandin E2 increases fibroblast gene‐specific and global DNA methylation via increased DNA methyltransferase expression. FASEB J. 26, 3703–3714 (2012). www.fasebj.org


PLOS ONE | 2014

Lung Fibroblasts from Patients with Idiopathic Pulmonary Fibrosis Exhibit Genome-Wide Differences in DNA Methylation Compared to Fibroblasts from Nonfibrotic Lung

Steven K. Huang; Anne M. Scruggs; Richard C. McEachin; Eric S. White; Marc Peters-Golden

Excessive fibroproliferation is a central hallmark of idiopathic pulmonary fibrosis (IPF), a chronic, progressive disorder that results in impaired gas exchange and respiratory failure. Fibroblasts are the key effector cells in IPF, and aberrant expression of multiple genes contributes to their excessive fibroproliferative phenotype. DNA methylation changes are critical to the development of many diseases, but the DNA methylome of IPF fibroblasts has never been characterized. Here, we utilized the HumanMethylation 27 array, which assays the DNA methylation level of 27,568 CpG sites across the genome, to compare the DNA methylation patterns of IPF fibroblasts (n = 6) with those of nonfibrotic patient controls (n = 3) and commercially available normal lung fibroblast cell lines (n = 3). We found that multiple CpG sites across the genome are differentially methylated (as defined by P value less than 0.05 and fold change greater than 2) in IPF fibroblasts compared to fibroblasts from nonfibrotic controls. These methylation differences occurred both in genes recognized to be important in fibroproliferation and extracellular matrix generation, as well as in genes not previously recognized to participate in those processes (including organ morphogenesis and potassium ion channels). We used bisulfite sequencing to independently verify DNA methylation differences in 3 genes (CDKN2B, CARD10, and MGMT); these methylation changes corresponded with differences in gene expression at the mRNA and protein level. These differences in DNA methylation were stable throughout multiple cell passages. DNA methylation differences may thus help to explain a proportion of the differences in gene expression previously observed in studies of IPF fibroblasts. Moreover, significant variability in DNA methylation was observed among individual IPF cell lines, suggesting that differences in DNA methylation may contribute to fibroblast heterogeneity among patients with IPF. These results demonstrate that IPF fibroblasts exhibit global differences in DNA methylation that may contribute to the excessive fibroproliferation associated with this disease.


Journal of Biological Chemistry | 2014

Prostaglandin E2 Inhibits α-Smooth Muscle Actin Transcription during Myofibroblast Differentiation via Distinct Mechanisms of Modulation of Serum Response Factor and Myocardin-related Transcription Factor-A

Loka Raghu Kumar Penke; Steven K. Huang; Eric S. White; Marc Peters-Golden

Background: PGE2 inhibits TGF-β1-induced myofibroblast differentiation, but the mechanism is incompletely understood. Results: PGE2 inhibits α-SMA transcription in human lung fibroblasts by preventing both up-regulation of SRF expression and nuclear translocation of MRTF-A. Conclusion: PGE2 blocks myofibroblast differentiation by targeting two critical determinants of contractile gene expression. Significance: These actions provide a mechanistic basis for therapeutic targeting of lung fibrosis. Differentiation of lung fibroblasts into contractile protein-expressing myofibroblasts by transforming growth factor-β1 (TGF-β1) is a critical event in the pathogenesis of pulmonary fibrosis. Transcription of the contractile protein α-smooth muscle actin (α-SMA) is mediated by the transcription factor serum-response factor (SRF) along with its co-activator, myocardin-related transcription factor-A (MRTF-A). The endogenous lipid mediator prostaglandin E2 (PGE2) exerts anti-fibrotic effects, including the inhibition of myofibroblast differentiation. However, the mechanism by which PGE2 inhibits α-SMA expression is incompletely understood. Here, we show in normal lung fibroblasts that PGE2 reduced the nuclear accumulation of MRTF-A·SRF complexes and consequently inhibited α-SMA promoter activation. It did so both by independently inhibiting SRF gene expression and nuclear import of MRTF-A. We identified that p38 MAPK is critical for TGF-β1-induced SRF gene expression and that PGE2 inhibition of SRF expression is associated with its ability to inhibit p38 activation. Its inhibition of MRTF-A import occurs via activation of cofilin 1 and inactivation of vasodilator-stimulated phosphoprotein. Similar effects of PGE2 on SRF gene expression were observed in fibroblasts from the lungs of patients with idiopathic pulmonary fibrosis. Thus, PGE2 is the first substance described to prevent myofibroblast differentiation by disrupting, via distinct mechanisms, the actions of both SRF and MRTF-A.


American Journal of Respiratory Cell and Molecular Biology | 2013

X-linked inhibitor of apoptosis regulates lung fibroblast resistance to Fas-mediated apoptosis.

Iyabode O. Ajayi; Thomas H. Sisson; Peter D. Higgins; Adam J. Booth; Rommel L. Sagana; Steven K. Huang; Eric S. White; Jessie E. King; Bethany B. Moore; Jeffrey C. Horowitz

The accumulation of apoptosis-resistant fibroblasts within fibroblastic foci is a characteristic feature of idiopathic pulmonary fibrosis (IPF), but the mechanisms underlying apoptosis resistance remain unclear. A role for the inhibitor of apoptosis (IAP) protein family member X-linked inhibitor of apoptosis (XIAP) has been suggested by prior studies showing that (1) XIAP is localized to fibroblastic foci in IPF tissue and (2) prostaglandin E₂ suppresses XIAP expression while increasing fibroblast susceptibility to apoptosis. Based on these observations, we hypothesized that XIAP would be regulated by the profibrotic mediators transforming growth factor (TGF)β-1 and endothelin (ET)-1 and that increased XIAP would contribute to apoptosis resistance in IPF fibroblasts. To address these hypotheses, we examined XIAP expression in normal and IPF fibroblasts at baseline and in normal fibroblasts after treatment with TGF-β1 or ET-1. The role of XIAP in the regulation of fibroblast susceptibility to Fas-mediated apoptosis was examined using functional XIAP antagonists and siRNA silencing. In concordance with prior reports, fibroblasts from IPF lung tissue had increased resistance to apoptosis compared with normal lung fibroblasts. Compared with normal fibroblasts, IPF fibroblasts had significantly but heterogeneously increased basal XIAP expression. Additionally, TGF-β1 and ET-1 induced XIAP protein expression in normal fibroblasts. Inhibition or silencing of XIAP enhanced the sensitivity of lung fibroblasts to Fas-mediated apoptosis without causing apoptosis in the absence of Fas activation. Collectively, these findings support a mechanistic role for XIAP in the apoptosis-resistant phenotype of IPF fibroblasts.

Collaboration


Dive into the Steven K. Huang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cory M. Hogaboam

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge