Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 49(2)2023 Feb.
Article in English | MEDLINE | ID: mdl-36633146

ABSTRACT

Structural alterations of collagen impact signaling that helps control tumor progression and the responses to therapeutic intervention. Integrins represent a class of receptors that include members that mediate collagen signaling. However, a strategy of directly targeting integrins to control tumor growth has demonstrated limited activity in the clinical setting. New molecular understanding of integrins have revealed that these receptors can regulate both pro­ and anti­tumorigenic functions in a cell type­dependent manner. Therefore, designing strategies that block pro­tumorigenic signaling, without impeding anti­tumorigenic functions, may lead to development of more effective therapies. In the present study, evidence was provided for a novel signaling cascade in which ß3­integrin­mediated binding to a secreted RGDKGE­containing collagen fragment stimulates an autocrine­like signaling pathway that differentially governs the activity of both YAP and (protein kinase­A) PKA, ultimately leading to alterations in the levels of immune checkpoint molecule PD­L1 by a proteasome dependent mechanism. Selectively targeting this collagen fragment, reduced nuclear YAP levels, and enhanced PKA and proteasome activity, while also exhibiting significant antitumor activity in vivo. The present findings not only provided new mechanistic insight into a previously unknown autocrine­like signaling pathway that may provide tumor cells with the ability to regulate PD­L1, but our findings may also help in the development of more effective strategies to control pro­tumorigenic ß3­integrin signaling without disrupting its tumor suppressive functions in other cellular compartments.


Subject(s)
B7-H1 Antigen , Collagen , Integrins , Neoplasms , Peptide Fragments , Proteasome Endopeptidase Complex , Humans , B7-H1 Antigen/metabolism , Collagen/chemistry , Collagen/metabolism , Integrins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Peptide Fragments/metabolism , YAP-Signaling Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism
2.
Am J Pathol ; 191(3): 527-544, 2021 03.
Article in English | MEDLINE | ID: mdl-33307038

ABSTRACT

The growth and spread of malignant tumors, such as ovarian carcinomas, are governed in part by complex interconnected signaling cascades occurring between stromal and tumor cells. These reciprocal cross-talk signaling networks operating within the local tissue microenvironment may enhance malignant tumor progression. Understanding how novel bioactive molecules generated within the tumor microenvironment regulate signaling pathways in distinct cellular compartments is critical for the development of more effective treatment paradigms. Herein, we provide evidence that blocking cellular interactions with an RGDKGE-containing collagen peptide that selectively binds integrin ß3 on ovarian tumor cells enhances the phosphorylation of the hippo effector kinase large tumor suppressor kinase-1 and reduces nuclear accumulation of yes-associated protein and its target gene c-Myc. Selectively targeting this RGDKGE-containing collagen fragment inhibited ovarian tumor growth and the development of ascites fluid in vivo. These findings suggest that this bioactive collagen fragment may represent a previously unknown regulator of the hippo effector kinase large tumor suppressor kinase-1 and regulate ovarian tumor growth by a yes-associated protein-dependent mechanism. Taken together, these data not only provide new mechanistic insight into how a unique collagen fragment may regulate ovarian cancer, but in addition may help provide a useful new alternative strategy to control ovarian tumor progression based on selectively disrupting a previously unappreciated signaling cascade.


Subject(s)
Biomarkers, Tumor/metabolism , Collagen/metabolism , Ovarian Neoplasms/pathology , Peptide Fragments/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred C57BL , Mice, Nude , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-yes/genetics , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays
3.
J Cell Physiol ; 235(12): 9005-9020, 2020 12.
Article in English | MEDLINE | ID: mdl-32400053

ABSTRACT

Structural remodeling of the extracellular matrix is a well-established process associated with tumor growth and metastasis. Tumor and stromal cells that compose the tumor mass function cooperatively to promote the malignant phenotype in part by physically interacting with intact and structurally altered matrix proteins. To this end, collagen represents the most abundant component of the extracellular matrix and is known to control the behavior of histologically distinct tumor types as well as a diversity of stromal cells. Although a significant molecular understanding has been established concerning how cellular interactions with intact collagen govern signaling pathways that control tumor progression, considerably less is known concerning how interactions with cryptic or hidden regions within remodeled collagen may selectively alter signaling cascades, or whether inhibition of these cryptic signaling pathways may represent clinically effective therapeutic strategies. Here, we review the emerging evidence concerning the possible mechanisms for the selective generation of cryptic or hidden elements within collagen and their potential cell surface receptors that may facilitate signal transduction. We discuss the concept that cellular communication links between cell surface receptors and these cryptic collagen elements may serve as functional signaling hubs that coordinate multiple signaling pathways operating within both tumor and stromal cells. Finally, we provide examples to help illustrate the possibility that direct targeting of these unique cryptic signaling hubs may lead to the development of more effective therapeutic strategies to control tumor growth and metastasis.


Subject(s)
Collagen/metabolism , Neoplasm Metastasis/pathology , Neoplasms/metabolism , Neovascularization, Pathologic/pathology , Animals , Cell Movement/physiology , Cell Proliferation/physiology , Humans
4.
Am J Pathol ; 188(10): 2356-2368, 2018 10.
Article in English | MEDLINE | ID: mdl-30118657

ABSTRACT

Stromal components not only help form the structure of neoplasms such as melanomas, but they also functionally contribute to their malignant phenotype. Thus, uncovering signaling pathways that integrate the behavior of both tumor and stromal cells may provide unique opportunities for the development of more effective strategies to control tumor progression. In this regard, extracellular matrix-mediated signaling plays a role in coordinating the behavior of both tumor and stromal cells. Here, evidence is provided that targeting a cryptic region of the extracellular matrix protein collagen (HU177 epitope) inhibits melanoma tumor growth and metastasis and reduces angiogenesis and the accumulation of α-SMA-expressing stromal cell in these tumors. The current study suggests that the ability of the HU177 epitope to control melanoma cell migration and metastasis depends on the transcriptional coactivator Yes-associated protein (YAP). Melanoma cell interactions with the HU177 epitope promoted nuclear accumulation of YAP by a cyclin-dependent kinase-5-associated mechanism. These findings provide new insights into the mechanism by which the anti-HU177 antibody inhibits metastasis, and uncovers an unknown signaling pathway by which the HU177 epitope selectively reprograms melanoma cells by regulating nuclear localization of YAP. This study helps to define a potential new therapeutic strategy to control melanoma tumor growth and metastasis that might be used alone or in combination with other therapeutics.


Subject(s)
Cell Movement/drug effects , Collagen/physiology , Epitopes/physiology , Melanoma/physiopathology , Skin Neoplasms/physiopathology , Adaptor Proteins, Signal Transducing/metabolism , Angiogenesis Inhibitors/pharmacology , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Anti-Idiotypic/physiology , Cell Proliferation/physiology , Collagen/immunology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/metabolism , Humans , Melanoma/pathology , Neoplasm Metastasis , Neovascularization, Pathologic/immunology , Phosphoproteins/metabolism , Phosphorylation/physiology , Skin Neoplasms/pathology , Stromal Cells/physiology , Talin/metabolism , Transcription Factors , Tumor Cells, Cultured , YAP-Signaling Proteins
5.
Am J Pathol ; 186(6): 1649-61, 2016 06.
Article in English | MEDLINE | ID: mdl-27216148

ABSTRACT

Evidence suggests that stromal cells play critical roles in tumor growth. Uncovering new mechanisms that control stromal cell behavior and their accumulation within tumors may lead to development of more effective treatments. We provide evidence that the HU177 cryptic collagen epitope is selectively generated within human ovarian carcinomas and this collagen epitope plays a role in SKOV-3 ovarian tumor growth in vivo. The ability of the HU177 epitope to regulate SKOV-3 tumor growth depends in part on its ability to modulate stromal cell behavior because targeting this epitope inhibited angiogenesis and, surprisingly, the accumulation of α-smooth muscle actin-expressing stromal cells. Integrin α10ß1 can serve as a receptor for the HU177 epitope in α-smooth muscle actin-expressing stromal cells and subsequently regulates Erk-dependent migration. These findings are consistent with a mechanism by which the generation of the HU177 collagen epitope provides a previously unrecognized α10ß1 ligand that selectively governs angiogenesis and the accumulation of stromal cells, which in turn secrete protumorigenic factors that contribute to ovarian tumor growth. Our findings provide a new mechanistic understanding into the roles by which the HU177 epitope regulates ovarian tumor growth and provide new insight into the clinical results from a phase 1 human clinical study of the monoclonal antibody D93/TRC093 in patients with advanced malignant tumors.


Subject(s)
Cell Proliferation , Collagen/metabolism , Epitopes , Ovarian Neoplasms/pathology , Tumor Microenvironment/physiology , Animals , Blotting, Western , Cell Adhesion/physiology , Cell Proliferation/physiology , Collagen/chemistry , Female , Heterografts , Humans , Mice , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism
6.
J Biol Chem ; 291(6): 2731-50, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26668310

ABSTRACT

Extracellular matrix (ECM) remodeling regulates angiogenesis. However, the precise mechanisms by which structural changes in ECM proteins contribute to angiogenesis are not fully understood. Integrins are molecules with the ability to detect compositional and structural changes within the ECM and integrate this information into a network of signaling circuits that coordinate context-dependent cell behavior. The role of integrin αvß3 in angiogenesis is complex, as evidence exists for both positive and negative functions. The precise downstream signaling events initiated by αvß3 may depend on the molecular characteristics of its ligands. Here, we identified an RGD-containing cryptic collagen epitope that is generated in vivo. Surprisingly, rather than inhibiting αvß3 signaling, this collagen epitope promoted αvß3 activation and stimulated angiogenesis and inflammation. An antibody directed to this RGDKGE epitope but not other RGD collagen epitopes inhibited angiogenesis and inflammation in vivo. The selective ability of this RGD epitope to promote angiogenesis and inflammation depends in part on its flanking KGE motif. Interestingly, a subset of macrophages may represent a physiologically relevant source of this collagen epitope. Here, we define an endothelial cell mechano-signaling pathway in which a cryptic collagen epitope activates αvß3 leading to an Src and p38 MAPK-dependent cascade that leads to nuclear accumulation of Yes-associated protein (YAP) and stimulation of endothelial cell growth. Collectively, our findings not only provide evidence for a novel mechano-signaling pathway, but also define a possible therapeutic strategy to control αvß3 signaling by targeting a pro-angiogenic and inflammatory ligand of αvß3 rather than the receptor itself.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Collagen/pharmacology , Endothelial Cells/metabolism , Epitopes/pharmacology , Mechanotransduction, Cellular/drug effects , Neovascularization, Physiologic/drug effects , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Line, Tumor , Collagen/chemistry , Endothelial Cells/cytology , Epitopes/chemistry , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mechanotransduction, Cellular/genetics , Mice , Phosphoproteins/genetics , Transcription Factors , YAP-Signaling Proteins , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases
7.
Angiogenesis ; 18(1): 31-46, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25249331

ABSTRACT

A more complete understanding of the mechanisms that regulate the angiogenic switch, which contributes to the conversion of small dormant tumors to actively growing malignancies, is important for the development of more effective anti-angiogenic strategies for cancer therapy. While significant progress has been made in understanding the complex mechanisms by which integrin αvß3 expressed in endothelial cells governs angiogenesis, less is known concerning the ability of αvß3 expressed within the tumor cell compartment to modulate the angiogenic output of a tumor. Here we provide evidence that αvß3 expressed in melanoma cells may contribute to the suppression of IGFBP-4, an important negative regulator of IGF-1 signaling. Given the multiple context-dependent roles for αvß3 in angiogenesis and tumor progression, our novel findings provide additional molecular insight into how αvß3 may govern the angiogenic switch by a mechanism associated with a p38 MAPK and matrix metalloproteinases-dependent regulation of the endogenous angiogenesis inhibitor IGFBP-4.


Subject(s)
Insulin-Like Growth Factor Binding Protein 4/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Melanoma/physiopathology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , DNA Primers/genetics , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Melanoma/complications , Neovascularization, Pathologic/etiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , X-Ray Microtomography
8.
J Biol Chem ; 287(3): 1779-89, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22134921

ABSTRACT

An in-depth understanding of the molecular and cellular complexity of angiogenesis continues to advance as new stimulators and inhibitors of blood vessel formation are uncovered. Gaining a more complete understanding of the response of blood vessels to both stimulatory and inhibitory molecules will likely contribute to more effective strategies to control pathological angiogenesis. Here, we provide evidence that endothelial cell interactions with structurally altered collagen type IV may suppress the expression of insulin-like growth factor binding protein-4 (IGFBP-4), a well documented inhibitor of the IGF-1/IGF-1R signaling axis. We report for the first time that IGFBP-4 differentially inhibits angiogenesis induced by distinct growth factor signaling pathways as IGFBP-4 inhibited FGF-2- and IGF-1-stimulated angiogenesis but failed to inhibit VEGF-induced angiogenesis. The resistance of VEGF-stimulated angiogenesis to IGFBP-4 inhibition appears to depend on sustained activation of p38 MAPK as blocking its activity restored the anti-angiogenic effects of IGFBP-4 on VEGF-induced blood vessel growth in vivo. These novel findings provide new insight into how blood vessels respond to endogenous inhibitors during angiogenesis stimulated by distinct growth factor signaling pathways.


Subject(s)
Angiogenesis Inhibitors/metabolism , Endothelial Cells/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/genetics , Animals , Chick Embryo , Endothelial Cells/cytology , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Vascular Endothelial Growth Factor A/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...