Network


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

Hotspot


Dive into the research topics where Angela R. Hess is active.

Publication


Featured researches published by Angela R. Hess.


American Journal of Pathology | 1999

Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry.

Andrew J. Maniotis; Robert Folberg; Angela R. Hess; Elisabeth A. Seftor; Lynn M.G. Gardner; Jacob Pe'er; Jeffrey M. Trent; Paul S. Meltzer; Mary J.C. Hendrix

Tissue sections from aggressive human intraocular (uveal) and metastatic cutaneous melanomas generally lack evidence of significant necrosis and contain patterned networks of interconnected loops of extracellular matrix. The matrix that forms these loops or networks may be solid or hollow. Red blood cells have been detected within the hollow channel components of this patterned matrix histologically, and these vascular channel networks have been detected in human tumors angiographically. Endothelial cells were not identified within these matrix-embedded channels by light microscopy, by transmission electron microscopy, or by using an immunohistochemical panel of endothelial cell markers (Factor VIII-related antigen, Ulex, CD31, CD34, and KDR[Flk-1]). Highly invasive primary and metastatic human melanoma cells formed patterned solid and hollow matrix channels (seen in tissue sections of aggressive primary and metastatic human melanomas) in three-dimensional cultures containing Matrigel or dilute Type I collagen, without endothelial cells or fibroblasts. These tumor cell-generated patterned channels conducted dye, highlighting looping patterns visualized angiographically in human tumors. Neither normal melanocytes nor poorly invasive melanoma cells generated these patterned channels in vitro under identical culture conditions, even after the addition of conditioned medium from metastatic pattern-forming melanoma cells, soluble growth factors, or regimes of hypoxia. Highly invasive and metastatic human melanoma cells, but not poorly invasive melanoma cells, contracted and remodeled floating hydrated gels, providing a biomechanical explanation for the generation of microvessels in vitro. cDNA microarray analysis of highly invasive versus poorly invasive melanoma tumor cells confirmed a genetic reversion to a pluripotent embryonic-like genotype in the highly aggressive melanoma cells. These observations strongly suggest that aggressive melanoma cells may generate vascular channels that facilitate tumor perfusion independent of tumor angiogenesis.


Nature Medicine | 2006

Embryonic and tumorigenic pathways converge via Nodal signaling: role in melanoma aggressiveness

Jolanta M. Topczewska; Lynne-Marie Postovit; Naira V. Margaryan; Anthony Sam; Angela R. Hess; William W. Wheaton; Brian J. Nickoloff; Jacek Topczewski; Mary J.C. Hendrix

Bidirectional cellular communication is integral to both cancer progression and embryological development. In addition, aggressive tumor cells are phenotypically plastic, sharing many properties with embryonic cells. Owing to the similarities between these two types of cells, the developing zebrafish can be used as a biosensor for tumor-derived signals. Using this system, we show that aggressive melanoma cells secrete Nodal (a potent embryonic morphogen) and consequently can induce ectopic formation of the embryonic axis. We further show that Nodal is present in human metastatic tumors, but not in normal skin, and thus may be involved in melanoma pathogenesis. Inhibition of Nodal signaling reduces melanoma cell invasiveness, colony formation and tumorigenicity. Nodal inhibition also promotes the reversion of melanoma cells toward a melanocytic phenotype. These data suggest that Nodal signaling has a key role in melanoma cell plasticity and tumorigenicity, thereby providing a previously unknown molecular target for regulating tumor progression.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Expression and functional significance of VE-cadherin in aggressive human melanoma cells: Role in vasculogenic mimicry

Mary J.C. Hendrix; Elisabeth A. Seftor; Paul S. Meltzer; Lynn M.G. Gardner; Angela R. Hess; Dawn A. Kirschmann; Gina C. Schatteman; Richard E.B. Seftor

We recently have introduced the term vasculogenic mimicry to describe the unique ability of aggressive melanoma tumor cells to form tubular structures and patterned networks in three-dimensional culture, which “mimics” embryonic vasculogenic networks formed by differentiating endothelial cells. In the current study, we address the biological significance of several endothelial-associated molecules (revealed by microarray analysis) with respect to expression and function in highly aggressive and poorly aggressive human cutaneous melanoma cell lines (established from the same patient). In a comparative analysis, CD31 was not expressed by any of the melanoma cell lines, whereas TIE-1 (tyrosine kinase with Ig and epidermal growth factor homology domains-1) was strongly expressed in the highly aggressive tumor cells with a low level of expression in one of the poorly aggressive cell lines. Vascular endothelial (VE)-cadherin was exclusively expressed by highly aggressive melanoma cells and was undetectable in the poorly aggressive tumor cells, suggesting the possibility of a vasculogenic switch. Down-regulation of VE-cadherin expression in the aggressive melanoma cells abrogated their ability to form vasculogenic networks and directly tested the hypothesis that VE-cadherin is critical in melanoma vasculogenic mimicry. These results highlight the plasticity of aggressive melanoma cells and call into question their possible genetic reversion to an embryonic phenotype. This finding could pose a significant clinical challenge in targeting tumor cells that may masquerade as circulating endothelial cells or other embryonic-like stem cells.


Cancer Research | 2005

Lysyl Oxidase Regulates Breast Cancer Cell Migration and Adhesion through a Hydrogen Peroxide–Mediated Mechanism

Stacey L. Payne; Ben Fogelgren; Angela R. Hess; Elisabeth A. Seftor; Elizabeth L. Wiley; Sheri F. T. Fong; Katalin Csiszar; Mary J.C. Hendrix; Dawn A. Kirschmann

We have previously shown that lysyl oxidase (LOX) mRNA is up-regulated in invasive breast cancer cells and that catalytically active LOX facilitates in vitro cell invasion. Here we validate our in vitro studies by showing that LOX expression is up-regulated in distant metastatic breast cancer tissues compared with primary cancer tissues. To elucidate the mechanism by which LOX facilitates cell invasion, we show that catalytically active LOX regulates in vitro motility/migration and cell-matrix adhesion formation. Treatment of the invasive breast cancer cell lines, Hs578T and MDA-MB-231, with beta-aminopropionitrile (betaAPN), an irreversible inhibitor of LOX catalytic activity, leads to a significant decrease in cell motility/migration and adhesion formation. Conversely, poorly invasive MCF-7 cells expressing LOX (MCF-7/LOX32-His) showed an increase in migration and adhesion that was reversible with the addition of betaAPN. Moreover, a decrease in activated focal adhesion kinase (FAK) and Src kinase, key proteins involved in adhesion complex turnover, was observed when invasive breast cancer cells were treated with betaAPN. Additionally, FAK and Src activation was increased in MCF-7/LOX32-His cells, which was reversible on betaAPN treatment. Hydrogen peroxide was produced as a by-product of LOX activity and the removal of hydrogen peroxide by catalase treatment in invasive breast cancer cells led to a dose-dependent loss in Src activation. These results suggest that LOX facilitates migration and cell-matrix adhesion formation in invasive breast cancer cells through a hydrogen peroxide-mediated mechanism involving the FAK/Src signaling pathway. These data show the need to target LOX for treatment of aggressive breast cancer.


Oncogene | 2003

Molecular plasticity of human melanoma cells

Mary J.C. Hendrix; Elisabeth A. Seftor; Angela R. Hess; Richard E. B. Seftor

The molecular analysis of tumors, such as melanoma, has benefited significantly from microarray technology that can facilitate the classification of tumors based on the differential expression of genes. The data summarized in this review describe the molecular profile of aggressive cutaneous and uveal melanoma cells as that of multiple phenotypes similar to a pluripotent, embryonic-like stem cell. A noteworthy example of the plasticity of the aggressive melanoma cell phenotype is demonstrated by the ability of these tumor cells to engage in vasculogenic mimicry and neovascularization. A review of the current evidence demonstrating important cellular and molecular determinants of melanoma vasculogenic mimicry is presented. In addition, novel signaling pathways are discussed, involving VE-cadherin, EphA2, FAK, and PI 3-kinase, which promote cell migration, invasion, and matrix remodeling. The observations summarized in this review describe some of the key molecular events that regulate the process of melanoma vasculogenic mimicry and identify new signal transduction pathways that can serve as putative targets for therapeutic intervention.


Cancer Biology & Therapy | 2006

VE-cadherin regulates EphA2 in aggressive melanoma cells through a novel signaling pathway : Implications for vasculogenic mimicry

Angela R. Hess; Elisabeth A. Seftor; Lynn M. Gruman; Michael S. Kinch; Richard E.B. Seftor; Mary J.C. Hendrix

The formation of matrix-rich, vasculogenic-like networks, termed vasculogenic mimicry (VM), is a unique process characteristic of highly aggressive melanoma cells found to express genes previously thought to be exclusively associated with endothelial and epithelial cells. This study contributes new observations demonstrating that VE-cadherin can regulate the expression of EphA2 at the cell membrane by mediating its ability to become phosphorylated through interactions with its membrane bound ligand, ephrin-A1. VE-cadherin and EphA2 were also found to be co-localized in cell-cell adhesion junctions, both in vitro and in vivo. Immunoprecipitation studies revealed that EphA2 and VE-cadherin could interact, directly and/or indirectly, during VM. Furthermore, there was no change in the co-localization of EphA2 and VE-cadherin at cell-cell adhesion sites when EphA2 was phosphorylated on tyrosine residues. Although transient knockout of EphA2 expression did not alter VE-cadherin localization, transient knockout of VE-cadherin expression resulted in the reorganization of EphA2 on the cells’ surface, an accumulation of EphA2 in the cytoplasm, and subsequent dephosphorylation of EphA2. Collectively, these results suggest that VE-cadherin and EphA2 act in a coordinated manner as a key regulatory element in the process of melanoma VM and illuminate a novel signaling pathway that could be potentially exploited for therapeutic intervention.


Critical Reviews in Oncology Hematology | 2002

Expression of multiple molecular phenotypes by aggressive melanoma tumor cells : role in vasculogenic mimicry

Elisabeth A. Seftor; Paul S. Meltzer; Gina C. Schatteman; Lynn M. Gruman; Angela R. Hess; Dawn A. Kirschmann; Richard E. B. Seftor; Mary J.C. Hendrix

Cutaneous melanoma has been increasing at an alarming rate over the past two decades, however, there are no acceptable histopathological markers that classify various stages of melanoma progression. Recently, the molecular analysis of cancer has contributed significantly to our understanding of the cellular and molecular underpinnings of tumor progression. The data summarized in this review describe the molecular signature of aggressive cutaneous melanoma cells as that of multiple phenotypes which may be similar to a pluripotent, embryonic-like phenotype. An example of the plasticity of this phenotype is demonstrated by the ability of aggressive melanoma cells to engage in vasculogenic mimicry and neovascularization. A review of the current data demonstrating important cellular and molecular determinants of human melanoma vasculogenic mimicry is presented. These findings should stimulate additional studies to address the biological relevance of the multiple molecular phenotypes expressed by aggressive melanoma cells which may lead to the development of new diagnostic markers and therapeutic targets for clinical intervention.


American Journal of Pathology | 2003

Differential Regulation of EphA2 in Normal and Malignant Cells

Jennifer Walker-Daniels; Angela R. Hess; Mary J.C. Hendrix; Michael S. Kinch

Eph receptor tyrosine kinases (RTK) comprise the largest family of tyrosine kinases encoded in the human genome. 1,2 Fourteen Eph receptors and eight ephrin ligands have been identified to date and these molecules are increasingly understood to play important roles in disease and development. 3-5 Eph receptor family members share structural and functional similarities. Their extracellular regions include an N-terminal ligand binding domain, 6,7 a cysteine-rich motif, and two fibronectin-like repeats. Also, Eph receptors can be distinguished from other RTKs in that they all recognize ligands, known as ephrins, which are anchored to the membrane of apposing cells. 1,2,8,9 Ephrins do not necessarily share extensive homology and are so grouped based on their abilities to bind Eph receptors. The ephrins have been separated into two classes based on the means by which they are anchored to the cell membrane. 3,9 EphrinA ligands are linked to the cell membrane by a glycosylphophatidylinositol (GPI) linkage, whereas EphrinB ligands encode for a transmembrane domain. Based on the identity of their ligands, the Eph receptors themselves have been classified into either EphA or EphB subfamilies. 9 These families share a degree of specificity, which is determined by a four-amino-acid loop on the extracellular surface. 10 Moreover, Eph receptors and ephrin ligands each have overlapping specificity. 1,2,8 Several ligands can bind to one receptor and, in turn, several receptors can bind to one ligand. In general, however, EphA receptors bind EphrinA ligands and EphB receptors bind EphrinB ligands. 1,2,8,11-13 Ligand binding typically triggers tyrosine phosphorylation of Eph receptors. 2 In particular, two tyrosines near the transmembrane domain are highly conserved and phosphorylated in response to ligand binding. 14,15 These residues appear to be critical for function, as mutations of these tyrosines abolish the enzymatic activity of certain Eph kinases. 15 In addition, tyrosine phosphorylation creates binding sites for signaling or adapter proteins (Figure 1) ▶ . 16 Other sites of protein-protein interaction are also mediated by sterile α motifs (SAM) 17,18 and PDZ (postsynaptic density protein, disc large, zona occludens) binding motifs 19 located near the C-terminal end of some Eph receptors. Figure 1. Biological and biochemical pathways linked with EphA2. Shown is a synthesis of the reported protein interactions and cellular consequences of EphA2 signaling. The solid lines denote areas of known positive regulation, whereas dotted lines represent a ... Eph receptors have been studied extensively in the developing nervous system, where they regulate patterning during neural development. 2,11-13,20,21 At the cellular level, ephrin binding causes Eph receptors to initiate signals that promote cell-cell repulsion and these events appear to assist axon guidance and neural organization. 1


Cancer Research | 2005

Focal Adhesion Kinase Promotes the Aggressive Melanoma Phenotype

Angela R. Hess; Lynne-Marie Postovit; Naira V. Margaryan; Elisabeth A. Seftor; Galen B. Schneider; Richard E.B. Seftor; Brian J. Nickoloff; Mary J.C. Hendrix

Malignant melanoma continues to remain a significant health threat, with death often occurring as a result of metastasis. The metastatic phenotype typically is characterized by augmented tumor cell invasion and migration in addition to tumor cell plasticity as shown by vasculogenic mimicry. Therefore, understanding the molecular mechanisms that promote an aggressive phenotype is essential to predicting the likelihood of metastasis at a stage when intervention may be possible. This study focuses on the role of focal adhesion kinase (FAK), a cytoplasmic tyrosine kinase important for many cellular processes, including cell survival, invasion, and migration. We found FAK to be phosphorylated on its key tyrosine residues, Tyr397 and Tyr576, in only aggressive uveal and cutaneous melanoma cells, which correlates with their increased invasion, migration, and vasculogenic mimicry plasticity. Additionally, we confirmed the presence of FAK phosphorylated on Tyr397 and Tyr576 in both cutaneous and uveal melanoma tumors in situ. Examination of a functional role for FAK in aggressive melanoma revealed that disruption of FAK-mediated signal transduction pathways, through the expression of FAK-related nonkinase (FRNK), results in a decrease in melanoma cell invasion, migration, and inhibition of vasculogenic mimicry. Moreover, we found that FRNK expression resulted in a down-regulation of Erk1/2 phosphorylation resulting in a decrease in urokinase activity. Collectively, these data suggest a new mechanism involved in promoting the aggressive melanoma phenotype through FAK-mediated signal transduction pathways, thus providing new insights into possible therapeutic intervention strategies.


Developmental Dynamics | 2007

Deciphering the Signaling Events that Promote Melanoma Tumor Cell Vasculogenic Mimicry and Their Link to Embryonic Vasculogenesis: Role of the Eph Receptors

Angela R. Hess; Naira V. Margaryan; Elisabeth A. Seftor; Mary J.C. Hendrix

During embryogenesis, the primordial microcirculation is formed through a process known as vasculogenesis. The term “vasculogenic mimicry” has been used to describe the manner in which highly aggressive, but not poorly aggressive melanoma tumor cells express endothelial and epithelial markers and form vasculogenic‐like networks similar to embryonic vasculogenesis. Vasculogenic mimicry is one example of the remarkable plasticity demonstrated by aggressive melanoma cells and suggests that these cells have acquired an embryonic‐like phenotype. Since the initial discovery of tumor cell vasculogenic mimicry by our laboratory, we have been focusing on understanding the molecular mechanisms that regulate this process. This review will highlight recent findings identifying key signal transduction events that regulate melanoma vasculogenic mimicry and their similarity to the signal transduction events responsible for promoting embryonic vasculogenesis and angiogenesis. Specifically, this review will focus on the role of the Eph receptors and ligands in embryonic vasculogenesis, angiogenesis, and vasculogenic mimicry. Developmental Dynamics 236:3283–3296, 2007.

Collaboration


Dive into the Angela R. Hess's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul S. Meltzer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge