Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Biol Chem ; 298(6): 102029, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35569509

RESUMEN

Epidermal growth factor receptor (EGFR) is a causal factor in carcinoma, yet many carcinoma patients are resistant to EGFR inhibitors. Potential insight into this resistance stems from prior work that showed EGFR in normal epithelial cells docks to the extracellular domain of the plasma membrane proteoglycan syndecan-4 (Sdc4) engaged with α3ß1 and α6ß4 integrins. We now report that this receptor complex is modified by the recruitment of syndecan-2 (Sdc2), the Recepteur d'Origine Nantais (RON) tyrosine kinase, and the cellular signaling mediator Abelson murine leukemia viral oncogene homolog 1 (ABL1) in triple-negative breast carcinoma and head and neck squamous cell carcinoma, where it contributes to EGFR kinase-independent proliferation. Treatment with a peptide mimetic of the EGFR docking site in the extracellular domain of Sdc4 (called SSTNEGFR) disrupts the entire complex and causes a rapid, global arrest of the cell cycle. Normal epithelial cells do not recruit these additional receptors to the adhesion mechanism and are not arrested by SSTNEGFR. Although EGFR docking with Sdc4 in the tumor cells is required, cell cycle progression does not depend on EGFR kinase. Instead, progression depends on RON kinase, activated by its incorporation into the complex. RON activates ABL1, which suppresses p38 mitogen-activated protein kinase and prevents a p38-mediated signal that would otherwise arrest the cell cycle. These findings add to the growing list of receptor tyrosine kinases that support tumorigenesis when activated by their association with syndecans at sites of matrix adhesion and identify new potential targets for cancer therapy.


Asunto(s)
Carcinoma , Ciclo Celular , Receptores ErbB , Proteínas Tirosina Quinasas Receptoras , Sindecano-2 , Sindecano-4 , Carcinoma/patología , Membrana Celular/metabolismo , Receptores ErbB/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Sindecano-2/metabolismo , Sindecano-4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
J Cell Sci ; 132(20)2019 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-31562188

RESUMEN

When targeted by the tumor-promoting enzyme heparanase, cleaved and shed syndecan-1 (Sdc1) then couples VEGFR2 (also known as KDR) to VLA-4, activating VEGFR2 and the directed migration of myeloma cells. But how VEGFR2 activates VLA-4-mediated motility has remained unknown. We now report that VEGFR2 causes PKA-mediated phosphorylation of VLA-4 on S988, an event known to stimulate tumor metastasis while suppressing cytotoxic immune cells. A key partner in this mechanism is the chemokine receptor CXCR4, a well-known mediator of cell motility in response to gradients of the chemokine SDF-1 (also known as CXCL12). The entire machinery necessary to phosphorylate VLA-4, consisting of CXCR4, AC7 (also known as ADCY7) and PKA, is constitutively associated with VEGFR2 and is localized to the integrin by Sdc1. VEGFR2 carries out the novel phosphorylation of Y135 within the DRY microswitch of CXCR4, sequentially activating Gαißγ, AC7 and PKA, which phosphorylates S988 on the integrin. This mechanism is blocked by a syndecan-mimetic peptide (SSTNVEGFR2), which, by preventing VEGFR2 linkage to VLA-4, arrests tumor cell migration that depends on VLA-4 phosphorylation and stimulates the LFA-1-mediated migration of cytotoxic leukocytes.


Asunto(s)
Movimiento Celular/inmunología , Integrina alfa4beta1/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Receptores CXCR4/inmunología , Sindecano-1/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Línea Celular Tumoral , Movimiento Celular/genética , Humanos , Vigilancia Inmunológica , Integrina alfa4beta1/genética , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Fosforilación/genética , Fosforilación/inmunología , Receptores CXCR4/genética , Sindecano-1/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
3.
J Biol Chem ; 290(43): 26103-13, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26350464

RESUMEN

The α6ß4 integrin is known to associate with receptor tyrosine kinases when engaged in epithelial wound healing and in carcinoma invasion and survival. Prior work has shown that HER2 associates with α6ß4 integrin and syndecan-1 (Sdc1), in which Sdc1 engages the cytoplasmic domain of the ß4 integrin subunit allowing HER2-dependent motility and carcinoma cell survival. In contrast, EGFR associates with Sdc4 and the α6ß4 integrin, and EGFR-dependent motility depends on cytoplasmic engagement of ß4 integrin with Sdc4. However, how HER2 and EGFR assimilate into a complex with the syndecans and integrin, and why kinase capture is syndecan-specific has remained unknown. In the present study, we demonstrate that HER2 is captured via a site, comprised of amino acids 210-240, in the extracellular domain of human Sdc1, and EGFR is captured via an extracellular site comprised of amino acids 87-131 in human Sdc4. Binding assays using purified recombinant proteins demonstrate that the interaction between the EGFR family members and the syndecans is direct. The α3ß1 integrin, which is responsible for the motility of the cells, is captured at these sites as well. Peptides based on the interaction motifs in Sdc1 and Sdc4, called synstatins (SSTN210-240 and SSTN87-131) competitively displace the receptor tyrosine kinase and α3ß1 integrin from the syndecan with an IC50 of 100-300 nm. The syndecans remain anchored to the α6ß4 integrin via its cytoplasmic domain, but the activation of cell motility is disrupted. These novel SSTN peptides are potential therapeutics for carcinomas that depend on these HER2- and EGFR-coupled mechanisms for their invasion and survival.


Asunto(s)
Movimiento Celular , Receptores ErbB/metabolismo , Integrina alfa3beta1/metabolismo , Integrina alfa6beta4/metabolismo , Receptor ErbB-2/metabolismo , Sindecano-1/metabolismo , Sindecano-4/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Células Epiteliales/metabolismo , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Sindecano-1/química , Sindecano-4/química
4.
J Biol Chem ; 289(44): 30318-30332, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25202019

RESUMEN

Epithelial cells are highly dependent during wound healing and tumorigenesis on the α6ß4 integrin and its association with receptor tyrosine kinases. Previous work showed that phosphorylation of the ß4 subunit upon matrix engagement depends on the matrix receptor syndecan (Sdc)-1 engaging the cytoplasmic domain of the ß4 integrin and coupling of the integrin to human epidermal growth factor receptor-2 (HER2). In this study, HER2-dependent migration activated by matrix engagement is compared with migration stimulated by EGF. We find that whereas HER2-dependent migration depends on Sdc1, EGF-dependent migration depends on a complex consisting of human epidermal growth factor receptor-1 (HER1, commonly known as EGFR), α6ß4, and Sdc4. The two syndecans recognize distinct sites at the extreme C terminus of the ß4 integrin cytoplasmic domain. The binding motif in Sdc1 is QEEXYX, composed in part by its syndecan-specific variable (V) region and in part by the second conserved (C2) region that it shares with other syndecans. A cell-penetrating peptide containing this sequence competes for HER2-dependent epithelial migration and carcinoma survival, although it is without effect on the EGFR-stimulated mechanism. ß4 mutants bearing mutations specific for Sdc1 and Sdc4 recognition act as dominant negative mutants to block cell spreading or cell migration that depends on HER2 or EGFR, respectively. The interaction of the α6ß4 integrin with the syndecans appears critical for it to be utilized as a signaling platform; migration depends on α3ß1 integrin binding to laminin 332 (LN332; also known as laminin 5), whereas antibodies that block α6ß4 binding are without effect. These findings indicate that specific syndecan family members are likely to have key roles in α6ß4 integrin activation by receptor tyrosine kinases.


Asunto(s)
Movimiento Celular , Supervivencia Celular , Integrina alfa6beta4/metabolismo , Sindecano-1/metabolismo , Sindecano-4/metabolismo , Secuencia de Aminoácidos , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/fisiología , Humanos , Integrina alfa6beta4/química , Integrina alfa6beta4/genética , Datos de Secuencia Molecular , Mutación Missense , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptor ErbB-2/fisiología , Transducción de Señal , Sindecano-1/química , Sindecano-4/química , Kalinina
5.
J Biol Chem ; 287(42): 34927-34935, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22936802

RESUMEN

Syndecan-1 is a cell surface proteoglycan that can organize co-receptors into a multimeric complex to transduce intracellular signals. The syndecan-1 core protein has multiple domains that confer distinct cell- and tissue-specific functions. Indeed, the extracellular, transmembrane, and cytoplasmic domains have all been found to regulate specific cellular processes. Our previous work demonstrated that syndecan-1 controls lung epithelial migration and adhesion. Here, we identified the necessary domains of the syndecan-1 core protein that modulate its function in lung epithelial repair. We found that the syndecan-1 transmembrane domain has a regulatory function in controlling focal adhesion disassembly, which in turn controls cell migration speed. In contrast, the extracellular domain facilitates cell adhesion through affinity modulation of α(2)ß(1) integrin. These findings highlight the fact that syndecan-1 is a multidimensional cell surface receptor that has several regulatory domains to control various biological processes. In particular, the lung epithelium requires the syndecan-1 transmembrane domain to govern cell migration and is independent from its ability to control cell adhesion via the extracellular domain.


Asunto(s)
Movimiento Celular/fisiología , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Sindecano-1/metabolismo , Animales , Adhesión Celular/fisiología , Línea Celular Transformada , Células Epiteliales/citología , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Humanos , Integrina alfa2beta1/genética , Integrina alfa2beta1/metabolismo , Ratones , Estructura Terciaria de Proteína , Mucosa Respiratoria/citología , Sindecano-1/genética
6.
Cancer Res Commun ; 3(1): 97-108, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36968227

RESUMEN

Previous studies have shown that the type I IGFR (IGF1R) suppresses apoptosis when it is autoactivated by coupling its extracellular domain to a matrix adhesion receptor complex consisting of syndecan-1 (Sdc1) and αvß3 or αvß5 integrin. We now report that head and neck squamous cell carcinoma (HNSCC) relies on this receptor complex. Disruption of the complex in HNSCC cells in vitro with a peptide mimetic of the organizer site in Sdc1 (called SSTNIGF1R) inactivates IGF1R, even in the presence of IGF1, and relieves the suppression of apoptosis signal-regulating kinase-1 (ASK1), dramatically reducing tumor cell survival. Normal epithelial cells do not assemble this receptor complex, require IGF1 to activate the IGF1R, and are refractory to SSTNIGF1R. In vivo, SSTNIGF1R reduced the growth of patient-derived HNSCC tumors in immunodeficient mice by 85%-95%. IGF1R's assimilation into the matrix receptor complex, which is detected in these tumors using the proximity ligation assay (PLA), is quantitatively disrupted by SSTNIGF1R, coinciding with ASK1 activation. PLA also detects the IGF1R-containing receptor complex in the archival sections of tonsil carcinomas, whereas the adjacent benign epithelium is negative. Likewise, PLA screening of oropharyngeal and adenoid cystic tumor microarrays demonstrated that over 95% of the tumors contained this unique receptor complex with no detectable expression in benign tissue. These findings suggest that HNSCC upregulates and is highly dependent on IGF1R signaling via this adhesion receptor complex. Targeting this mechanism with novel therapeutics, including highly specific SSTNIGF1R, is likely to offer promising outcomes for patients with carcinoma. Significance: A newly developed biomarker reveals upregulation of an antiapoptotic IGF1R-integrin-syndecan receptor complex in head and neck cancer and documents disruption of the complex in patient-derived tumor xenografts (PDX) treated with the inhibitor SSTNIGF1R. A corresponding blockade in PDX growth in the presence of this inhibitor demonstrates that therapies designed to target this mechanism will likely offer promising outcomes for patients with head and neck cancer.


Asunto(s)
Neoplasias de Cabeza y Cuello , Peptidomiméticos , Humanos , Ratones , Animales , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Transducción de Señal , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Péptidos/farmacología , Receptor IGF Tipo 1
7.
J Cell Sci ; 123(Pt 21): 3796-807, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20971705

RESUMEN

Syndecan-1 (Sdc1) engages and activates the αvß3 (and/or αvß5) integrin when clustered in human carcinoma and endothelial cells. Although the engagement is extracellular, the activation mechanism is cytoplasmic. This talin-dependent, inside-out signaling pathway is activated downstream of the insulin-like growth factor-1 receptor (IGF1R), whose kinase activity is triggered by Sdc1 clustering. In vitro binding assays using purified receptors suggest that association of the Sdc1 ectodomain with the integrin provides a 'docking face' for IGF1R. IGF1R docking and activation of the associated integrin is blocked by synstatin (SSTN(92-119)), a peptide derived from the integrin engagement site in Sdc1. IGF1R colocalizes with αvß3 integrin and Sdc1 in focal contacts, but fails to associate with or activate the integrin in cells either lacking Sdc1 or expressing Sdc1(Δ67-121), a mutant that is unable to form the Sdc1-integrin-IGF1R ternary complex. Integrin activation is also blocked by IGF1R inhibitors or by silencing IGF1R or talin expression with small-interfering RNAs (siRNAs). In both cases, expression of the constitutively active talin F23 head domain rescues integrin activation. We recently reported that SSTN(92-119) blocks angiogenesis and impairs tumor growth in mice, therefore this Sdc1-mediated integrin regulatory mechanism might be a crucial regulator of disease processes known to rely on these integrins, including tumor cell metastasis and tumor-induced angiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Integrina alfaVbeta3/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores de Somatomedina/metabolismo , Sindecano-1/metabolismo , Animales , Sitios de Unión/genética , Línea Celular Tumoral , Células Endoteliales/patología , Adhesiones Focales/metabolismo , Humanos , Ratones , Mutación/genética , Fragmentos de Péptidos/genética , Unión Proteica/genética , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , Agregación de Receptores/genética , Receptores de Somatomedina/genética , Transducción de Señal/genética , Sindecano-1/genética , Talina/genética , Talina/metabolismo
8.
Blood ; 115(12): 2449-57, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20097882

RESUMEN

Heparanase enhances shedding of syndecan-1 (CD138), and high levels of heparanase and shed syndecan-1 in the tumor microenvironment are associated with elevated angiogenesis and poor prognosis in myeloma and other cancers. To explore how the heparanase/syndecan-1 axis regulates angiogenesis, we used myeloma cells expressing either high or low levels of heparanase and examined their impact on endothelial cell invasion and angiogenesis. Medium conditioned by heparanase-high cells significantly stimulated endothelial invasion in vitro compared with medium from heparanase-low cells. The stimulatory activity was traced to elevated levels of vascular endothelial growth factor (VEGF) and syndecan-1 in the medium. We discovered that the heparan sulfate chains of syndecan-1 captured VEGF and also attached the syndecan-1/VEGF complex to the extracellular matrix where it then stimulated endothelial invasion. In addition to its heparan sulfate chains, the core protein of syndecan-1 was also required because endothelial invasion was blocked by addition of synstatin, a peptide mimic of the integrin activating region present on the syndecan-1 core protein. These results reveal a novel mechanistic pathway driven by heparanase expression in myeloma cells whereby elevated levels of VEGF and shed syndecan-1 form matrix-anchored complexes that together activate integrin and VEGF receptors on adjacent endothelial cells thereby stimulating tumor angiogenesis.


Asunto(s)
Glucuronidasa/metabolismo , Mieloma Múltiple , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Sindecano-1/metabolismo , Aorta/citología , Línea Celular Tumoral , Medios de Cultivo Condicionados/farmacología , Endotelio/patología , Matriz Extracelular/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Mieloma Múltiple/fisiopatología , Invasividad Neoplásica , Técnicas de Cultivo de Órganos , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
J Biol Chem ; 285(18): 13569-79, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20181947

RESUMEN

The alpha6beta4 integrin is a laminin 332 (LN332) receptor central to the formation of hemidesmosomes in epithelial layers. However, the integrin becomes phosphorylated by keratinocytes responding to epidermal growth factor in skin wounds or by squamous cell carcinomas that overexpress/hyperactivate the tyrosine kinase ErbB2, epidermal growth factor receptor, or c-Met. We show here that the beta4-dependent signaling in A431 human squamous carcinoma cells is dependent on the syndecan family of matrix receptors. Yeast two-hybrid analysis identifies an interaction within the distal third (amino acids 1473-1752) of the beta4 cytoplasmic domain and the conserved C2 region of the syndecan cytoplasmic domain. Via its C2 region, Sdc1 forms a complex with the alpha6beta4 integrin along with the receptor tyrosine kinase ErbB2 and the cytoplasmic kinase Fyn in A431 cells. Engagement of LN332 or clustering of the alpha6beta4 integrin with integrin-specific antibodies causes phosphorylation of ErbB2, Fyn, and the beta4 subunit as well as activation of phosphatidylinositol 3-kinase and Akt and their assimilation into this complex. This leads to phosphatidylinositol 3-kinase-dependent cell spreading and Akt-dependent protection from apoptosis. This is disrupted by RNA interference silencing of Sdc1 but can be rescued by mouse Sdc1 or Sdc4 but not by syndecan mutants lacking their C-terminal C2 region. This disruption does not prevent the phosphorylation of ErbB2 or Fyn but blocks the Fyn-mediated phosphorylation of the beta4 tail. We propose that syndecans engage the distal region of the beta4 cytoplasmic domain and bring it to the plasma membrane, where it can be acted upon by Src family kinases.


Asunto(s)
Hemidesmosomas/metabolismo , Integrina alfa6beta4/metabolismo , Queratinocitos/metabolismo , Receptor ErbB-2/metabolismo , Sindecanos/metabolismo , Animales , Apoptosis/genética , Línea Celular , Activación Enzimática/genética , Hemidesmosomas/genética , Humanos , Integrina alfa6beta4/genética , Ratones , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptor ErbB-2/genética , Transducción de Señal/genética , Piel/lesiones , Sindecano-1/genética , Sindecano-1/metabolismo , Sindecano-4/genética , Sindecano-4/metabolismo , Sindecanos/genética , Heridas y Lesiones/genética , Heridas y Lesiones/metabolismo
10.
J Cell Biol ; 174(7): 1097-106, 2006 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-16982797

RESUMEN

Cell surface heparan sulfate (HS) proteoglycans are carbohydrate-rich regulators of cell migratory, mitogenic, secretory, and inflammatory activity that bind and present soluble heparin-binding growth factors (e.g., fibroblast growth factor, Wnt, Hh, transforming growth factor beta, amphiregulin, and hepatocyte growth factor) to their respective signaling receptors. We demonstrate that the deglycanated core protein of syndecan-1 (SDC1) and not HS chains nor SDC2 or -4, appears to target the epithelial selective prosecretory mitogen lacritin. An important and novel step in this mechanism is that binding necessitates prior partial or complete removal of HS chains by endogenous heparanase. This limits lacritin activity to sites where heparanase appears to predominate, such as sites of exocrine cell migration, secretion, renewal, and inflammation. Binding is mutually specified by lacritin's C-terminal mitogenic domain and SDC1's N terminus. Heparanase modification of the latter transforms a widely expressed HS proteoglycan into a highly selective surface-binding protein. This novel example of cell specification through extracellular modification of an HS proteoglycan has broad implications in development, homeostasis, and disease.


Asunto(s)
Células Epiteliales/metabolismo , Glucuronidasa/farmacología , Glicoproteínas/metabolismo , Sustancias de Crecimiento/metabolismo , Glicoproteínas de Membrana/metabolismo , Polisacáridos/metabolismo , Proteoglicanos/metabolismo , Línea Celular , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Glicoproteínas de Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Unión Proteica , Estructura Terciaria de Proteína , Proteoglicanos/efectos de los fármacos , Sindecano-1 , Sindecanos
11.
Front Oncol ; 11: 775349, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34778093

RESUMEN

Receptor tyrosine kinases (RTKs) and integrin matrix receptors have well-established roles in tumor cell proliferation, invasion and survival, often functioning in a coordinated fashion at sites of cell-matrix adhesion. Central to this coordination are syndecans, another class of matrix receptor, that organize RTKs and integrins into functional units, relying on docking motifs in the syndecan extracellular domains to capture and localize RTKs (e.g., EGFR, IGF-1R, VEGFR2, HER2) and integrins (e.g., αvß3, αvß5, α4ß1, α3ß1, α6ß4) to sites of adhesion. Peptide mimetics of the docking motifs in the syndecans, called "synstatins", prevent assembly of these receptor complexes, block their signaling activities and are highly effective against tumor cell invasion and survival and angiogenesis. This review describes our current understanding of these four syndecan-coupled mechanisms and their inhibitory synstatins (SSTNIGF1R, SSTNVEGFR2, SSTNVLA-4, SSTNEGFR and SSTNHER2).

12.
J Cell Biol ; 163(3): 637-48, 2003 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-14610064

RESUMEN

Heparan sulfate (HS) interacts with diverse growth factors, including Wnt, Hh, BMP, VEGF, EGF, and FGF family members, and is a necessary component for their signaling. These proteins regulate multiple cellular processes that are critical during development. However, a major question is whether developmental changes occur in HS that regulate the activity of these factors. Using a ligand and carbohydrate engagement assay, and focusing on FGF1 and FGF8b interactions with FGF receptor (FR)2c and FR3c, this paper reveals global changes in HS expression in mouse embryos during development that regulate FGF and FR complex assembly. Furthermore, distinct HS requirements are identified for both complex formation and signaling for each FGF and FR pair. Overall, these results suggest that changes in HS act as critical temporal regulators of growth factor and morphogen signaling during embryogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Embrión de Mamíferos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/metabolismo , Ratones/metabolismo , Proteínas Tirosina Quinasas , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Animales no Consanguíneos , Unión Competitiva/efectos de los fármacos , Unión Competitiva/fisiología , Células CHO , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , División Celular/fisiología , Cricetinae , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Femenino , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/farmacología , Factor 8 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/farmacología , Ratones/embriología , Embarazo , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Factores de Tiempo
13.
J Cell Biol ; 167(1): 171-81, 2004 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-15479743

RESUMEN

The alpha(v)beta(3) integrin participates in cell morphogenesis, growth factor signaling, and cell survival. Activation of the integrin is central to these processes and is influenced by specific ECM components, which engage both integrins and syndecans. This paper demonstrates that the alpha(v)beta(3) integrin and syndecan-1 (S1) are functionally coupled. The integrin is dependent on the syndecan to become activated and to mediate signals required for MDA-MB-231 and MDA-MB-435 human mammary carcinoma cell spreading on vitronectin or S1-specific antibody. Coupling of the syndecan to alpha(v)beta(3) requires the S1 ectodomain (ED), as ectopic expression of glycosylphosphatidylinositol-linked S1ED enhances alpha(v)beta(3) recognition of vitronectin; and treatments that target this domain, including competition with recombinant S1ED protein or anti-S1ED antibodies, mutation of the S1ED, or down-regulation of S1 expression by small-interfering RNAs, disrupt alpha(v)beta(3)-dependent cell spreading and migration. Thus, S1 is likely to be a critical regulator of many cellular behaviors that depend on activated alpha(v)beta(3) integrins.


Asunto(s)
Neoplasias de la Mama/metabolismo , Integrina alfaVbeta3/metabolismo , Glicoproteínas de Membrana/química , Proteoglicanos/química , Carcinoma/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Separación Celular , Supervivencia Celular , Citometría de Flujo , Eliminación de Gen , Glicosilfosfatidilinositoles/química , Humanos , Integrinas/metabolismo , Proteínas de Neoplasias/inmunología , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/química , Transducción de Señal , Sindecano-1 , Sindecanos , Vitronectina/metabolismo
14.
Sci Rep ; 9(1): 9126, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31235839

RESUMEN

Epidermal growth factor receptor (EGFR) and its downstream phosphoinositide 3-kinase (PI3K) pathway are commonly deregulated in cancer. Recently, we have shown that the IQ motif-containing GTPase-activating protein 1 (IQGAP1) provides a molecular platform to scaffold all the components of the PI3K-Akt pathway and results in the sequential generation of phosphatidylinositol-3,4,5-trisphosphate (PI3,4,5P3). In addition to the PI3K-Akt pathway, IQGAP1 also scaffolds the Ras-ERK pathway. To define the specificity of IQGAP1 for the control of PI3K signaling, we have focused on the IQ3 motif in IQGAP1 as PIPKIα and PI3K enzymes bind this region. An IQ3 deletion mutant loses interactions with the PI3K-Akt components but retains binding to ERK and EGFR. Consistently, blocking the IQ3 motif of IQGAP1 using an IQ3 motif-derived peptide mirrors the effect of IQ3 deletion mutant by reducing Akt activation but has no impact on ERK activation. Also, the peptide disrupts the binding of IQGAP1 with PI3K-Akt pathway components, while IQGAP1 interactions with ERK and EGFR are not affected. Functionally, deleting or blocking the IQ3 motif inhibits cell proliferation, invasion, and migration in a non-additive manner to a PIPKIα inhibitor, establishing the functional specificity of IQ3 motif towards the PI3K-Akt pathway. Taken together, the IQ3 motif is a specific target for suppressing activation of the PI3K-Akt but not the Ras-ERK pathway. Although EGFR stimulates the IQGAP1-PI3K and -ERK pathways, here we show that IQGAP1-PI3K controls migration, invasion, and proliferation independent of ERK. These data illustrate that the IQ3 region of IQGAP1 is a promising therapeutic target for PI3K-driven cancer.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Activadoras de ras GTPasa/química , Proteínas Activadoras de ras GTPasa/metabolismo , Secuencias de Aminoácidos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Humanos , Invasividad Neoplásica , Eliminación de Secuencia , Proteínas Activadoras de ras GTPasa/genética
15.
FEBS J ; 284(1): 42-55, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27758044

RESUMEN

Because of its impact on multiple biological pathways, heparanase has emerged as a major regulator of cancer, inflammation and other disease processes. Heparanase accomplishes this by degrading heparan sulfate which regulates the abundance and location of heparin-binding growth factors thereby influencing multiple signaling pathways that control gene expression, syndecan shedding and cell behavior. In addition, heparanase can act via nonenzymatic mechanisms that directly activate signaling at the cell surface. Clinical trials testing heparanase inhibitors as anticancer therapeutics are showing early signs of efficacy in patients further emphasizing the biological importance of this enzyme. This review focuses on recent developments in the field of heparanase regulation of cancer and inflammation, including the impact of heparanase on exosomes and autophagy, and novel mechanisms whereby heparanase regulates tumor metastasis, angiogenesis and chemoresistance. In addition, the ongoing development of heparanase inhibitors and their potential for treating cancer and inflammation are discussed.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glucuronidasa/genética , Neoplasias/genética , Neovascularización Patológica/genética , Sindecanos/genética , Antineoplásicos/uso terapéutico , Autofagia , Ensayos Clínicos como Asunto , Resistencia a Antineoplásicos/genética , Inhibidores Enzimáticos/uso terapéutico , Exosomas/metabolismo , Glucuronidasa/antagonistas & inhibidores , Glucuronidasa/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Inflamación , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Transducción de Señal , Sindecanos/metabolismo
17.
Clin Cancer Res ; 11(12): 4282-8, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15958608

RESUMEN

Fibroblast growth factor-2 (FGF-2) is a potent angiogenic cytokine that is dependent on heparan sulfate for its biological activity. We have investigated the relationship among heparan sulfate, FGF-2, and the signal-transducing receptors in human, advanced-stage, serous ovarian adenocarcinoma. Using a unique molecular probe, FR1c-Ap, which consisted of a soluble FGF receptor 1 isoform IIIc covalently linked to an alkaline phosphatase moiety, the distribution of heparan sulfate that had the ability to support the formation of a heparan sulfate/FGF-2/FGFR1 isoform IIIc alkaline phosphatase heparan sulfate construct complex was determined. This may be taken as a surrogate marker for the distribution of biologically active heparan sulfate and was distributed predominantly in endothelial cells and stroma but was absent from adenocarcinoma cells. In situ hybridization revealed the expression of FGFR1 mRNA in the endothelium and reverse transcription-PCR confirmed the presence of FGFR1 isoform IIIc but not isoform IIIb. The presence of FGF-2 around tumor endothelium was detected through immunohistochemistry. Double-staining techniques showed that heparan sulfate was found predominantly at the basal aspect of the endothelium and suggested that syndecan-3 might function as one of the proteoglycans involved in FGF-2 signaling in the endothelium. The data suggest that the entire extracellular signaling apparatus, consisting of FGF-2, biologically active heparan sulfate, and FGFRs capable of responding to FGF-2, is present in ovarian cancer endothelium, thereby highlighting the cytokine and its cognate receptor as potential targets for the antiangiogenic treatment of this disease.


Asunto(s)
Endotelio/patología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neoplasias Ováricas/patología , Fosfatasa Alcalina/metabolismo , Endotelio/química , Endotelio/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Heparitina Sulfato/metabolismo , Humanos , Inmunohistoquímica/métodos , Hibridación in Situ , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfatos/metabolismo
18.
Trends Cell Biol ; 26(5): 352-366, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26827089

RESUMEN

Epidermal growth factor receptor (EGFR) has fundamental roles in normal physiology and cancer, making it a rational target for cancer therapy. Surprisingly, however, inhibitors that target canonical, ligand-stimulated EGFR signaling have proven to be largely ineffective in treating many EGFR-dependent cancers. Recent evidence indicates that both intrinsic and therapy-induced cellular stress triggers robust, noncanonical pathways of ligand-independent EGFR trafficking and signaling, which provides cancer cells with a survival advantage and resistance to therapeutics. Here, we review the mechanistic regulation of noncanonical EGFR trafficking and signaling, and the pathological and therapeutic stresses that activate it. We also discuss the implications of this pathway in clinical treatment of EGFR-overexpressing cancers.


Asunto(s)
Receptores ErbB/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Estrés Fisiológico , Animales , Autofagia , Endosomas/metabolismo , Humanos , Transporte de Proteínas
19.
Trends Cancer ; 2(7): 378-390, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27819060

RESUMEN

Phosphoinositide 3-kinase (PI3K) generation of PI(3,4,5)P3 from PI(4,5)P2 and the subsequent activation of Akt and its downstream signaling cascades (e.g. mTORC1) dominates the landscape of phosphoinositide signaling axis in cancer research. However, PI(4,5)P2 is breaking its boundary as merely a substrate for PI3K and phospholipase C (PLC), and is now an established lipid messenger pivotal for different cellular events in cancer. Here, we review the phosphoinositide signaling axis in cancer, giving due weight to PI(4,5)P2 and its generating enzymes, the phosphatidylinositol phosphate (PIP) kinases (PIPKs). We highlighted how PI(4,5)P2 and PIP kinases serve as a proximal node in phosphoinositide signaling axis and how its interaction with cytoskeletal proteins regulates migratory and invasive nexus of metastasizing tumor cells.


Asunto(s)
Neoplasias/metabolismo , Fosfatidilinositoles/metabolismo , Animales , Movimiento Celular , Polaridad Celular , Citoesqueleto/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
20.
Cancer Res ; 76(17): 4981-93, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27364558

RESUMEN

Syndecan-1 (Sdc1/CD138) expression is linked to disease severity in multiple myeloma, although the causal basis for this link remains unclear. Here we report that capture of the IGF1 receptor (IGF1R) by Sdc1 suppresses ASK1-dependent apoptosis in multiple myeloma cells. Sdc1 binds two different fractions of IGF1R, one that is constitutively active and a second that is activated by IGF1 ligand. Notably, IGF1R kinase activity in both fractions is blocked by synstatinIGF1R (SSTNIGF1R), a peptide that inhibits IGF1R capture by Sdc1, as well as by a truncated peptide (SSTNIGF1R-T) that appears to be specific for multiple myeloma cells. Mechanistically, we show that ASK1 is bound to active IGF1R and inhibited by Tyr and Ser83/Ser966 phosphorylation. When IGF1R engagement with Sdc1 is blocked by SSTNIGF1R, ASK1 becomes activated, and initiates JNK- and caspase-3-mediated apoptosis. In pharmacologic tests, we find SSTNIGF1R is highly stable in human plasma and displays a half-life of 27 hours in mice, wherein it significantly reduces both the size and neovascularization of CAG myeloma tumor xenografts. Taken together, our results offer a preclinical proof of concept and mechanistic rationale for the exploration of SSTNIGF1R as an experimental therapeutic to dually attack multiple myeloma tumor cell survival and tumor angiogenesis. Cancer Res; 76(17); 4981-93. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Mieloma Múltiple/patología , Receptor IGF Tipo 1/farmacología , Receptores de Somatomedina/metabolismo , Sindecano-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Inmunoprecipitación , MAP Quinasa Quinasa Quinasa 5/metabolismo , Ratones , Ratones Desnudos , Mieloma Múltiple/metabolismo , Péptidos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA