Network


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

Hotspot


Dive into the research topics where Steven C. Wu is active.

Publication


Featured researches published by Steven C. Wu.


Journal of Biological Chemistry | 2007

Protein kinase C ζ: A novel regulator of both phosphorylation and de-phosphorylation of cardiac sarcomeric proteins

Steven C. Wu; R. John Solaro

Our experiments investigated associations of specific isoforms of protein kinase C (PKC) with individual proteins in the cardiac troponin complex. Troponin I (cTnI) associated with PKCϵ and ζ and troponin T (cTnT) associated with PKC α, δ, and ϵ. Based on its association with cTnI, we hypothesized that PKCζ is a major regulator of myofilament protein phosphorylation. To test this, we infected adult cardiac myocytes with adenoviral constructs containing DsRed monomer-tagged wild type (WT) and the following constitutively active forms of PKCζ: the pseudo-substrate region (A119E), 3′-phospho-inositide-dependent kinase-1 (T410E), and auto-phosphorylation (T560E). The A119E and T410E mutants displayed increased localization to the Z-discs compared with WT and T560E. Immunoprecipitations were performed in myocytes expressing PKCζ using PKC phospho-motif antibodies to determine the phosphophorylation of cTnI, cTnT, tropomyosin, myosin-binding protein C, and desmin. We did not find serine (Ser) phosphorylation of cTnI or cTnT. However, we observed a significant decrease in threonine (Thr) phosphorylation of cTnI and cTnT notably by PKCζ T560E. Ser phosphorylation of tropomyosin was increased by all three active mutants of PKCζ. Ser/Thr phosphorylation of myosin-binding protein C increased primarily by PKCζ A119E. Both PKCζ A119E and T410E mutants increased desmin Ser/Thr phosphorylation. To explain the apparent Thr dephosphorylation of cTnI and cTnT, we hypothesized that PKCζ exists as a complex with p21-activated kinase-1 (Pak1) and protein phosphatase 2A (PP2A), and this was confirmed by immunoprecipitation Western blot. Our data demonstrate that PKCζ is a novel regulator of myofilament protein phosphorylation.


Journal of Biological Chemistry | 2000

Purification and Characterization of β-Adrenergic Receptor mRNA-binding Proteins

Burns C. Blaxall; Amy C. Pellett; Steven C. Wu; Aldo Pende; J. David Port

β-Adrenergic receptors (β-ARs), like other G-protein-coupled receptors, can undergo post-transciptional regulation at the level of mRNA stability. In particular, the human β1- and β2-ARs and the hamster β2-AR mRNA undergo β-agonist-mediated destabilization. By UV cross-linking, we have previously described an ∼M r 36,000 mRNA-binding protein, βARB, that binds to A/C+U-rich nucleotide regions within 3′-untranslated regions. Further, we have demonstrated previously that βARB is immunologically distinct from AUF1/heterogeneous nuclear ribonucleoprotein (hnRNP) D, another mRNA-binding protein associated with destabilization of A+U-rich mRNAs (Pende, A., Tremmel, K. D., DeMaria, C. T., Blaxall, B. C., Minobe, W., Sherman, J. A., Bisognano, J., Bristow, M. R., Brewer, G., and Port, J. D. (1996) J. Biol. Chem. 271, 8493–8501). In this report, we describe the peptide composition of βARB. Mass spectrometric analysis of an ∼M r36,000 band isolated from ribosomal salt wash proteins revealed the presence of two mRNA-binding proteins, hnRNP A1, and the elav-like protein, HuR, both of which are known to bind to A+U-rich nucleotide regions. By immunoprecipitation, HuR appears to be the biologically dominant RNA binding component of βARB. Although hnRNP A1 and HuR can both be immunoprecipitated from ribosomal salt wash proteins, the composition of βARB (HuR alone versus HuR and hnRNP A1) appears to be dependent on the mRNA probe used. The exact role of HuR and hnRNP A1 in the regulation of β-AR mRNA stability remains to be determined.


Journal of Biological Chemistry | 1997

Binding of src-like Kinases to the β-Subunit of the Interleukin-3 Receptor

Elizabeth A. Burton; Seija Hunter; Steven C. Wu; Steven M. Anderson

We have previously shown that stimulation of 32D cl3 cells with interleukin (IL)-3 results in the activation of three src-like tyrosine kinases, fyn, hck, and lyn. The β subunit of the IL-3 receptor co-immunoprecipitated with hck in lysates of both unstimulated and IL-3-stimulated cells; however, the β subunit did not precipitate with either fyn or lyn. The association of these three kinases with the β subunit of the IL-3 receptor was further investigated using bacterial fusion proteins encoding the unique, SH3, and SH2 domains of these three kinases. Fusion proteins of both hck and fyn bound to a 150-kDa tyrosine-phosphorylated protein present in lysates of IL-3-stimulated cells. This protein was identified as the β subunit of the IL-3 receptor by immunoblotting with an anti-β antibody. Glutathione S-transferase (GST) fusion proteins containing the SH2 domain of hck bound to the β subunit although the amount of β subunit that bound to the SH2 domain alone was only 30% of that which bound to the fusion protein containing the unique, SH3, and SH2 domains. This indicates that the SH2 domain is one of the motifs involved in binding hck to the β subunit. A GST fusion protein encoding a 236-amino acid region of the cytoplasmic tail of the β subunit, which contained four tyrosine residues, bound to hck and fyn. Binding to both proteins was dramatically increased when the GST-β fusion protein was tyrosine-phosphorylated. Far Western blot analysis was used to demonstrate the binding of the unique, SH3, and SH2 domains of hck to this 236-amino acid region of the β subunit; tyrosine phosphorylation of this protein increased the binding of both the unique region and the SH2 domain probes. These data indicate that binding of hck to the β subunit is mediated by both phosphotyrosine-dependent and -independent mechanisms.


Cellular Signalling | 2012

Nuclear localization drives α1-adrenergic receptor oligomerization and signaling in cardiac myocytes

Casey D. Wright; Steven C. Wu; Erika F. Dahl; Alan J. Sazama; Timothy D. O'Connell

Conventional models of G-protein coupled receptor (GPCR) signaling describe cell surface receptors binding to external ligands, such as hormones or circulating peptides, to induce intracellular signaling and a physiologic response. However, recent studies identify new paradigms indicating that GPCRs localize to and signal at the nucleus and that GPCR oligomers can influence receptor function. Previously, we reported that endogenous α1-adrenergic receptors (α1-ARs) localize to and signal at the nuclei in adult cardiac myocytes. In this study, we examined the mechanisms behind α1-AR nuclear localization and how nuclear localization impacted receptor function. We verified that endogenous α1-ARs localized to the nuclear membrane of intact nuclei isolated from wild-type adult cardiac myocytes. Next, we identified and disrupted putative nuclear localization sequences in both the α1A- and α1B-adrenergic receptors, which led to mis-localization of α1-ARs in cultured adult cardiac myocytes. Using these mutants, we demonstrated that nuclear localization was required for α1-signaling in adult cardiac myocytes. We also found that the nuclear export inhibitor leptomycin B inhibited α1-AR signaling, indicating α1-AR signaling must arise in the nucleus in adult cardiac myocytes. Finally, we found that co-localization of the α1-subtypes at the nuclei in adult cardiac myocytes facilitated the formation of receptor oligomers that could affect receptor signaling. In summary, our data indicate that α1-AR nuclear localization can drive the formation of receptor oligomers and regulate signaling in adult cardiac myocytes.


Molecular and Cellular Biochemistry | 2002

Correlation between intrinsic mRNA stability and the affinity of AUF1 (hnRNP D) and HuR for A+U-rich mRNAs

Burns C. Blaxall; Aldo Pende; Steven C. Wu; J. David Port

Presence of A+U-rich elements (AREs) within 3′-untranslated regions (3′UTRs) of numerous mRNAs has been associated with rapid mRNA turnover; however, the interaction of specific factors with AREs is also associated with mRNA stabilization. Recently, two ARE binding proteins with putative mRNA destabilizing (AUF1) and stabilizing (HuR) properties have been described. However, no direct comparison of AUF1 and HuR binding properties has been made. Therefore, we examined the relative affinities of p37AUF1 and HuR for a diverse set of ARE-containing mRNAs encoding β-adrenergic receptors, a proto-oncogene, and a cytokine. We find that high-affinity AUF1 binding appears to require elements beyond primary nucleotide sequence. In contrast, binding of HuR appears considerably less constrained. As a functional correlate, we determined the ability of these specific mRNA sequences to affect the stability of chimeric β-globin mRNA constructs. Although the relative affinity of AUF1 and HuR are generally predictive of mRNA stability, we find that certain mRNA sequences do not conform to these generalizations.


Circulation-heart Failure | 2015

Derivation and High Engraftment of Patient-Specific Cardiomyocyte-Sheet Using Induced Pluripotent Stem Cells Generated From Adult Cardiac Fibroblast

Liying Zhang; Jing Guo; Pengyuan Zhang; Qiang Xiong; Steven C. Wu; Lily Xia; Samit S. Roy; Jakub Tolar; Timothy D. O’Connell; Michael Kyba; K. Liao; Jianyi Zhang

Background—Induced pluripotent stem cells (iPSCs) can be differentiated into potentially unlimited lineages of cell types for use in autologous cell therapy. However, the efficiency of the differentiation procedure and subsequent function of the iPSC-derived cells may be influenced by epigenetic factors that the iPSCs retain from their tissues of origin; thus, iPSC-derived cells may be more effective for treatment of myocardial injury if the iPSCs were engineered from cardiac-lineage cells, rather than dermal fibroblasts. Methods and Results—We show that human cardiac iPSCs (hciPSCs) can be generated from cardiac fibroblasts and subsequently differentiated into exceptionally pure (>92%) sheets of cardiomyocytes (CMs). The hciPSCs passed through all the normal stages of differentiation before assuming a CM identity. When using the fibrin gel–enhanced delivery of hciPSC-CM sheets at the site of injury in infarcted mouse hearts, the engraftment rate was 31.91%±5.75% at Day 28 post transplantation. The hciPSC-CM in the sheet also appeared to develop a more mature, structurally aligned phenotype 28 days after transplantation and was associated with significant improvements in cardiac function, vascularity, and reduction in apoptosis. Conclusions—These data strongly support the potential of hciPSC-CM sheet transplantation for the treatment of heart with acute myocardial infarction.


Journal of Biological Chemistry | 2007

Identification of a Region of Troponin I Important in Signaling Cross-bridge-dependent Activation of Cardiac Myofilaments

Patti L. Engel; Tomoyoshi Kobayashi; Brandon J. Biesiadecki; Jonathan P. Davis; Svetlana B. Tikunova; Steven C. Wu; R. John Solaro

Force generating strong cross-bridges are required to fully activate cardiac thin filaments, but the molecular signaling mechanism remains unclear. Evidence demonstrating differential extents of cross-bridge-dependent activation of force, especially at acidic pH, in myofilaments in which slow skeletal troponin I (ssTnI) replaced cardiac TnI (cTnI) indicates the significance of a His in ssTnI that is an homologous Ala in cTnI. We compared cross-bridge-dependent activation in myofilaments regulated by cTnI, ssTnI, cTnI(A66H), or ssTnI(H34A). A drop from pH 7.0 to 6.5 induced enhanced cross-bridge-dependent activation in cTnI myofilaments, but depressed activation in cTnI(A66H) myofilaments. This same drop in pH depressed cross-bridge-dependent activation in both ssTnI myofilaments and ssTnI(H34A) myofilaments. Compared with controls, cTnI(A66H) myofilaments were desensitized to Ca2+, whereas there was no difference in the Ca2+-force relationship between ssTnI and ssTnI(H34A) myofilaments. The mutations in cTnI and ssTnI did not affect Ca2+ dissociation rates from cTnC at pH 7.0 or 6.5. However, at pH 6.5, cTnI(A66H) had lower affinity for cTnT than cTnI. We also probed cross-bridge-dependent activation in myofilaments regulated by cTnI(Q56A). Myofilaments containing cTnI(Q56A) demonstrated cross-bridge-dependent activation that was similar to controls containing cTnI at pH 7.0 and an enhanced cross-bridge-dependent activation at pH 6.5. We conclude that a localized N-terminal region of TnI comprised of amino acids 33–80, which interacts with C-terminal regions of cTnC and cTnT, is of particular significance in transducing signaling of thin filament activation by strong cross-bridges.


Journal of Molecular and Cellular Cardiology | 2010

An Association Between Gene Expression and Better Survival in Female Mice Following Myocardial Infarction

Quanhai Chen; Roy Williams; Chastity L. Healy; Casey D. Wright; Steven C. Wu; Timothy D. O'Connell

Following myocardial infarction, the prognosis for females is better than males. Estrogen is thought to be protective, but clinical trials with hormone replacement failed to show protection. Here, we sought to identify novel mechanisms that might explain this sex-based difference. By diverging from the traditional focus on sex hormones, we employed a conceptually novel approach to this question by using a non-biased approach to measure global changes in gene expression following infarction. We hypothesized that specific gene programs are initiated in the heart following infarction that might account for this sex-based difference. We induced small, medium, and large infarcts in male and female mice and measured changes in gene expression by microarray following infarction. Regardless of infarct size, survival was better in females, while mortality occurred 3-10 days following infarction in males. Two days following infarction, males developed significant ventricular dilation, the best predictor of mortality in humans. Three days following infarction, we measured gene expression by microarray, comparing male versus female and sham versus surgery/infarction. In general, our results indicate a higher relative level of gene induction in females versus males and identified programs for angiogenesis, extracellular matrix remodeling, and immune response. This pattern of gene expression was linked to less pathologic remodeling in female hearts, including increased capillary density and decreased fibrosis. In summary, our results suggest an association between improved survival and less pathologic remodeling and the relative induction of gene expression in females following myocardial infarction.


Journal of the American Heart Association | 2014

Nuclear Localization of α1A-Adrenergic Receptors Is Required for Signaling in Cardiac Myocytes: An “Inside-Out” α1-AR Signaling Pathway

Steven C. Wu; Erika F. Dahl; Casey D. Wright; Andrew L Cypher; Chastity L. Healy; Timothy D. O'Connell

Background Recent studies indicate that α1‐adrenergic receptors (α1‐ARs) are cardioprotective by preventing cardiac myocyte death and augmenting contractility in heart failure. Although G‐protein‐coupled receptors are assumed to localize to and signal at the plasma membrane, we previously demonstrated that endogenous α1‐ARs localize to the nuclei in adult cardiac myocytes. However, the functional consequence of this nuclear localization remains unclear. Here, we attempted to reconcile nuclear localization of α1‐ARs with their physiologic function by examining α1‐AR‐induced contractility in adult cardiac myocytes. Methods and Results By measuring shortening in unloaded, cultured adult cardiac myocytes, we found that the α1A‐subtype regulated contractility through phosphorylation of cardiac troponin I (cTnI) at the protein kinase C (PKC) site, threonine 144. Reconstitution of an α1A‐subtype nuclear localization mutant in cardiac myocytes lacking α1‐ARs failed to rescue nuclear α1A‐mediated phosphorylation of cTnI and myocyte contractility. Leptomycin B, the nuclear export inhibitor, also blocked α1A‐mediated phosphorylation of cTnI. These data indicate that α1‐AR signaling originates in the nucleus. Consistent with these observations, we localized the α1A‐subtype to the inner nuclear membrane, identified PKCα, δ, and ε in the nucleus, and found that α1‐ARs activate PKCδ in nuclei isolated from adult cardiac myocytes. Finally, we found that a PKCδ nuclear localization mutant blunted α1‐induced phosphorylation of cTnI. Conclusions Together, our data identify a novel, “inside‐out” nuclear α1A‐subtype/PKCδ/cTnI‐signaling pathway that regulates contractile function in adult cardiac myocytes. Importantly, these data help resolve the discrepancy between nuclear localization of α1‐ARs and α1‐AR‐mediated physiologic function.


Journal of Cardiovascular Pharmacology | 2014

Nuclear compartmentalization of α1-adrenergic receptor signaling in adult cardiac myocytes.

Steven C. Wu; OʼConnell Td

Abstract: Although convention dictates that G protein-coupled receptors localize to and signal at the plasma membrane, accumulating evidence suggests that G protein-coupled receptors localize to and signal at intracellular membranes, most notably the nucleus. In fact, there is now significant evidence indicating that endogenous alpha-1 adrenergic receptors (&agr;1-ARs) localize to and signal at the nuclei in adult cardiac myocytes. Cumulatively, the data suggest that &agr;1-ARs localize to the inner nuclear membrane, activate intranuclear signaling, and regulate physiologic function in adult cardiac myocytes. Although &agr;1-ARs signal through G&agr;q, unlike other Gq-coupled receptors, &agr;1-ARs mediate important cardioprotective functions including adaptive/physiologic hypertrophy, protection from cell death (survival signaling), positive inotropy, and preconditioning. Also unlike other Gq-coupled receptors, most, if not all, functional &agr;1-ARs localize to the nuclei in adult cardiac myocytes, as opposed to the sarcolemma. Together, &agr;1-AR nuclear localization and cardioprotection might suggest a novel model for compartmentalization of Gq-coupled receptor signaling in which nuclear Gq-coupled receptor signaling is cardioprotective.

Collaboration


Dive into the Steven C. Wu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chastity L. Healy

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Casey D. Wright

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aldo Pende

Anschutz Medical Campus

View shared research outputs
Top Co-Authors

Avatar

Andrew L Cypher

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Burns C. Blaxall

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gregory C. Shearer

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge