Network


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

Hotspot


Dive into the research topics where H. S. Wiley is active.

Publication


Featured researches published by H. S. Wiley.


Journal of Biological Chemistry | 1999

ErbB-2 amplification inhibits down-regulation and induces constitutive activation of both ErbB-2 and epidermal growth factor receptors.

Worthylake R; Lee K. Opresko; H. S. Wiley

ErbB-2/HER2 is an important signaling partner for the epidermal growth factor receptor (EGFR). Overexpression of erbB-2 is also associated with poor prognosis in breast cancer. To investigate how erbB-2 amplification affects its interactions with the EGFR, we used a human mammary epithelial cell system in which erbB-2 expression was increased 7–20-fold by gene transfection. We found that amplification of erbB-2 caused constitutive activation of erbB-2 as well as ligand-independent activation of the EGFR. Overexpression of erbB-2 strongly inhibited erbB-2 down-regulation following transactivation by EGFR. Significantly, down-regulation of activated EGFR was also inhibited by erbB-2 amplification, resulting in enhanced ligand-dependent activation of the EGFR. The rate of EGFR endocytosis was not affected by erbB-2 overexpression, but the rate of lysosomal targeting was significantly reduced. In addition, erbB-2 overexpression promoted rapid recycling of activated EGFR back to the cell surface and decreased ligand dissociation from the EGFR. Our data suggest that overexpression of erbB-2 inhibits both its down-regulation and that of the EGFR. The net effect is increased signaling through the EGFR system.


Journal of Biological Chemistry | 1999

Internalized Epidermal Growth Factor Receptors Participate in the Activation of p21 ras in Fibroblasts

Jason M. Haugh; Huang Ac; H. S. Wiley; Alan Wells; Douglas A. Lauffenburger

Regulated activation of the highly conserved Ras GTPase is a central event in the stimulation of cell proliferation, motility, and differentiation elicited by receptor tyrosine kinases, such as the epidermal growth factor receptor (EGFR). In fibroblasts, this involves formation and membrane localization of Shc·Grb2·Sos complexes, which increases the rate of Ras guanine nucleotide exchange. In order to control Ras-mediated cell responses, this activity is regulated by receptor down-regulation and a feedback loop involving the dual specificity kinase mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK). We investigated the role of EGFR endocytosis in the regulation of Ras activation. Of fundamental interest is whether activated receptors in endosomes can participate in the stimulation of Ras guanine nucleotide exchange, because the constitutive membrane localization of Ras may affect its compartmentalization. By exploiting the differences in postendocytic signaling of two EGFR ligands, epidermal growth factor and transforming growth factor-α, we found that activated EGFR located at the cell surface and in internal compartments contribute equally to the membrane recruitment and tyrosine phosphorylation of Shc in NR6 fibroblasts expressing wild-type EGFR. Importantly, both the rate of Ras-specific guanine nucleotide exchange and the level of Ras-GTP were depressed to near basal values on the time scale of receptor trafficking. Using the selective MEK inhibitor PD098059, we were able to block the feedback desensitization pathway and maintain activation of Ras. Under these conditions, the generation of Ras-GTP was not significantly affected by the subcellular location of activated EGFR. In conjunction with our previous analysis of the phospholipase C pathway in the same cell line, this suggests a selective continuation of specific signaling activities and cessation of others upon receptor endocytosis.


Journal of Cell Science | 2007

EGF-receptor-mediated mammary epithelial cell migration is driven by sustained ERK signaling from autocrine stimulation.

Elizabeth J. Joslin; Lee K. Opresko; Alan Wells; H. S. Wiley; Douglas A. Lauffenburger

EGF family ligands are synthesized as membrane-anchored precursors whose proteolytic release yields mature diffusible factors that can activate cell surface receptors in autocrine or paracrine mode. Expression of these ligands is altered in pathological states and in physiological processes, such as development and tissue regeneration. Despite the widely documented biological importance of autocrine EGF signaling, quantitative relationships between protease-mediated ligand release and consequent cell behavior have not been rigorously investigated. We thus explored the relationship between autocrine EGF release rates and cell behavioral responses along with activation of ERK, a key downstream signal, by expressing chimeric ligand precursors and modulating their proteolytic shedding using a metalloprotease inhibitor in human mammary epithelial cells. We found that ERK activation increased monotonically with increasing ligand release rate despite concomitant downregulation of EGF receptor levels. Cell migration speed was directly related to ligand release rate and proportional to steady-state phospho-ERK levels. Moreover, migration speed was significantly greater for autocrine stimulation compared with exogenous stimulation, even at comparable phospho-ERK levels. By contrast, cell proliferation rates were approximately equivalent at all ligand release rates and were similar regardless of whether the ligand was presented endogenously or exogenously. Thus, in our mammary epithelial cell system, migration and proliferation are differentially sensitive to the mode of EGF ligand presentation.


Cell | 1980

Differential postendocytotic compartmentation in Xenopus oocytes is mediated by a specifically bound ligand

L. Opresko; H. S. Wiley; R.A. Wallace

Xenopus laevis oocytes were used as a model system to study the intracellular fate of proteins incorporated by endocytosis. We found that the intracellular stability and compartmentation of proteins incorporated by receptor-mediated endocytosis differed substantially from that of proteins incorporated by nonspecific endocytosis. After its uptake, the specifically sequestered yolk precursor protein vitellogenin was converted to the yolk proteins lipovitellin and phosvitin which were stable with time (up to 13 days in culture). In contrast, nonspecifically incorporated albumin (125I-labeled or 3H-labeled bovine serum albumin) was rapidly degraded. To determine whether the differential stability of these proteins was related to their entry into different postendocytotic compartments, we examined the intracellular transfer pathways taken by these proteins. The transfer of vitellogenin from coated vesicles to the yolk platelets was found to involve a secondary compartment formed by the fusion of the incoming endosomes. This compartment, termed transitional yolk bodies (TYB), underwent a progressive condensation until it attained its terminal density (1.21 g/cm3) after approximately 1.5 hr. The fusion of the TYB with the yolk platelets then occurred coincidentally with the time at which vitellogenin was proteolytically processed into the yolk proteins within the TYB. When the proteolytic cleavage of vitellogenin was blocked there was no fusion of the two compartments. In contrast, we found that albumin incorporated in the absence of vitellogenin was directly transferred from endosomes to yolk platelets without the formation of, or fusion with a secondary compartment. However, when oocytes were exposed simultaneously to albumin and vitellogenin both proteins followed identical routes of compartmentation (that of vitellogenin) with no evidence of direct transfer of either protein to the yolk platelets. These results suggest that the incorporation of a specifically bound ligand can result in the formation of a unique intracellular compartment. Moreover, since yolk platelets were able to fuse only with vesicles lacking occupied receptors (in the case of albumin alone) or with a compartment in which the specific ligand had been proteolytically cleaved and presumably released from its receptor (in the case of VTG), we suggest that occupied receptors can act as a transmembrane signal which directs the postendocytotic compartmentation of proteins.


British Journal of Cancer | 1999

Amphiregulin acts as an autocrine growth factor in two human polarizing colon cancer lines that exhibit domain selective EGF receptor mitogenesis

Lars Damstrup; Scott K. Kuwada; Peter J. Dempsey; Christa L. Brown; Hawkey Cj; Poulsen Hs; H. S. Wiley; Robert J. Coffey

SummaryColonic enterocytes, like many epithelial cells in vivo, are polarized with functionally distinct apical and basolateral membrane domains. The aims of this study were to characterize the endogenous epidermal growth factor (EGF)-like ligands expressed in two polarizing colon cancer cell lines, HCA-7 Colony 29 (HCA-7) and Caco-2, and to examine the effects of cell polarity on EGF receptor-mediated mitogenesis. HCA-7 and Caco-2 cells were grown on plastic, or as a polarized monolayer on Transwell filters. Cell proliferation was measured by 3H-thymidine incorporation and EGF receptor (EGFR) binding was assessed by Scatchard analysis. EGFR ligand expression was determined by Northern blot analysis, reverse transcription polymerase chain reaction, metabolic labelling and confocal microscopy. We found that amphiregulin (AR) was the most abundant EGFR ligand expressed in HCA-7 and Caco-2 cells. AR was localized to the basolateral surface and detected in basolateral-conditioned medium. Basolateral administration of neutralizing AR antibodies significantly reduced basal DNA replication. A single class of high-affinity EGFRs was detected in the basolateral compartment, whereas the apical compartment of polarized cells, and cells cultured on plastic, displayed two classes of receptor affinity. Basolateral administration of transforming growth factor alpha (TGF-α) or an EGFR neutralizing antibody also resulted in a dose-dependent stimulation or attenuation, respectively, of DNA replication. However, no mitogenic response was observed when these agents were added to the apical compartment or to confluent cells cultured on plastic. We conclude that amphiregulin acts as an autocrine growth factor in HCA-7 and Caco-2 cells, and EGFR ligand-induced proliferation is influenced by cellular polarity.


Oncogene | 2014

Autocrine HBEGF expression promotes breast cancer intravasation, metastasis and macrophage-independent invasion in vivo

Zhen Ni Zhou; Ved P. Sharma; Bt Beaty; Minna Roh-Johnson; Ea Peterson; N. van Rooijen; Pa Kenny; H. S. Wiley; John Condeelis; Jeffrey E. Segall

Increased expression of HBEGF in estrogen receptor-negative breast tumors is correlated with enhanced metastasis to distant organ sites and more rapid disease recurrence upon removal of the primary tumor. Our previous work has demonstrated a paracrine loop between breast cancer cells and macrophages in which the tumor cells are capable of stimulating macrophages through the secretion of colony-stimulating factor-1 while the tumor-associated macrophages (TAMs), in turn, aid in tumor cell invasion by secreting epidermal growth factor. To determine how the autocrine expression of epidermal growth factor receptor (EGFR) ligands by carcinoma cells would affect this paracrine loop mechanism, and in particular whether tumor cell invasion depends on spatial ligand gradients generated by TAMs, we generated cell lines with increased HBEGF expression. We found that autocrine HBEGF expression enhanced in vivo intravasation and metastasis and resulted in a novel phenomenon in which macrophages were no longer required for in vivo invasion of breast cancer cells. In vitro studies revealed that expression of HBEGF enhanced invadopodium formation, thus providing a mechanism for cell autonomous invasion. The increased invadopodium formation was directly dependent on EGFR signaling, as demonstrated by a rapid decrease in invadopodia upon inhibition of autocrine HBEGF/EGFR signaling as well as inhibition of signaling downstream of EGFR activation. HBEGF expression also resulted in enhanced invadopodium function via upregulation of matrix metalloprotease 2 (MMP2) and MMP9 expression levels. We conclude that high levels of HBEGF expression can short-circuit the tumor cell/macrophage paracrine invasion loop, resulting in enhanced tumor invasion that is independent of macrophage signaling.


Journal of Biological Chemistry | 2008

Multiple Mechanisms Are Responsible for Transactivation of the Epidermal Growth Factor Receptor in Mammary Epithelial Cells

Karin D. Rodland; Nikki Bollinger; Danielle L. Ippolito; Lee K. Opresko; Robert J. Coffey; Richard C. Zangar; H. S. Wiley

The number of distinct signaling pathways that can transactivate the epidermal growth factor receptor (EGFR) in a single cell type is unclear. Using a single strain of human mammary epithelial cells, we found that a wide variety of agonists, such as lysophosphatidic acid (LPA), uridine triphosphate, growth hormone, vascular endothelial growth factor, insulin-like growth factor-1 (IGF-1), and tumor necrosis factor-α, require EGFR activity to induce ERK phosphorylation. In contrast, hepatocyte growth factor can stimulate ERK phosphorylation independent of the EGFR. EGFR transactivation also correlated with an increase in cell proliferation and could be inhibited with metalloprotease inhibitors. However, there were significant differences with respect to transactivation kinetics and sensitivity to different inhibitors. In particular, IGF-1 displayed relatively slow transactivation kinetics and was resistant to inhibition by the selective ADAM-17 inhibitor WAY-022 compared with LPA-induced transactivation. Studies using anti-ligand antibodies showed that IGF-1 transactivation required amphiregulin production, whereas LPA was dependent on multiple ligands. Direct measurement of ligand shedding confirmed that LPA treatment stimulated shedding of multiple EGFR ligands, but paradoxically, IGF-1 had little effect on the shedding rate of any ligand, including amphiregulin. Instead, IGF-1 appeared to work by enhancing EGFR activation of Ras in response to constitutively produced amphiregulin. This enhancement of EGFR signaling was independent of both receptor phosphorylation and PI-3-kinase activity, suggestive of a novel mechanism. Our studies demonstrate that within a single cell type, the EGFR autocrine system can couple multiple signaling pathways to ERK activation and that this modulation of EGFR autocrine signaling can be accomplished at multiple regulatory steps.


Journal of Biological Chemistry | 1997

Structural aspects of the epidermal growth factor receptor required for transmodulation of erbB-2/neu.

Worthylake R; H. S. Wiley

The epidermal growth factor receptor (EGF-R) is known to transmodulate the activity and level of the erbB-2/neu protein in several epithelial cell lines. We therefore determined which structural features of the EGF-R were important in transmodulating erbB-2. We found that the addition of EGF to nontransformed epithelial cells resulted in down-regulation of erbB-2 with the same kinetics and similar extent as the EGF-R. By using cells expressing a series of EGF-R modified by site-directed mutagenesis, we found that EGF-R tyrosine kinase activity was not necessary for down-regulation of erbB-2, but receptor sequences between 899 and 958 in the EGF-R were required. To determine whether transmodulation was associated with activation of erbB-2, tyrosine phosphorylation of erbB-2 was determined following addition of EGF. Again, phosphorylation of erbB-2 following EGF addition did not require the intrinsic tyrosine kinase activity of the EGF-R, but did require sequences between 899 and 958. To determine the localization of EGF-R and erbB-2 following EGF addition, the relative distribution of the two receptors was evaluated by fluorescence microscopy. Surprisingly, the majority of erbB-2 was found in small cytoplasmic vesicles, whereas the EGF-R was predominantly found on the cell surface. Addition of EGF resulted in a redistribution and consequent colocalization of both receptors in endosomal and lysosomal structures. We conclude that activation and transmodulation of erbB-2 does not require intrinsic tyrosine kinase activity of the EGF-R, but does require sequences in the EGF-R which regulate its trafficking.


Biotechnology and Bioengineering | 1998

Escape of autocrine ligands into extracellular medium: Experimental test of theoretical model predictions

G. T. Oehrtman; H. S. Wiley; Douglas A. Lauffenburger

We have developed an experimental system for testing mathematical model predictions concerning escape of autocrine ligands into the extracellular bulk medium. This system employs anti-receptor blocking antibodies against the epidermal growth factor receptor (EGFR)/transforming growth factor alpha (TGFalpha) receptor/ligand pair. TGFalpha was expressed under the control of a tetracycline-repressed promoter, together with a constitutively expressed human EGFR in B82 mouse fibroblast cells. This expression system allowed us to vary TGFalpha synthesis rates over a roughly 300-fold range by adjusting tetracycline concentration. TGFalpha accumulation in the extracellular bulk medium was then measured as a function of cell density, TGFalpha synthesis rate, and anti-EGFR blocking antibody concentration. Consistent with model predictions, amounts of ligand in the medium on a per cell basis were found to diminish as cell density was increased but with reduced dependence on cell density at higher ligand synthesis rates. Similarly consistent with model predictions, higher ligand synthesis rates also decreased the effect of anti-receptor blocking antibodies. Our investigation has established that we can successfully analyze and understand autocrine ligand secretion behavior from the basis of our theoretical model.


Reproduction Nutrition Development | 1980

Growth of anuran oocytes in serum-supplemented medium

Robin A. Wallace; Ziva Misulovin; H. S. Wiley

Appropriately-sized oocytes from Xenopus laevis can be grown in vitro in vitellogenin-containing serum for up to 2 weeks. The source of serum appears to be unimportant. Rates of oocyte growth are somewhat better than those achieved in the presence of vitellogenin alone. Rana pipiens oocytes grow about twice as fast as X. laevis oocytes under identical conditions. The oocyte culture conditions described appear to be applicable to amphibian oocytes in general.

Collaboration


Dive into the H. S. Wiley's collaboration.

Top Co-Authors

Avatar

Douglas A. Lauffenburger

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gordon N. Gill

University of California

View shared research outputs
Top Co-Authors

Avatar

Alan Wells

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

K A Lund

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Robert J. Coffey

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robin A. Wallace

Oak Ridge National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Cheri S. Lazar

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge