Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mod Pathol ; 37(1): 100371, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38015043

RESUMO

B7-H4 (VTCN1), a member of the B7 family, is overexpressed in several types of cancer. Here we investigated the pattern of expression of B7-H4 in salivary gland carcinomas (SGC) and assessed its potential as a prognostic marker and therapeutic target. Immunohistochemistry (IHC) analyses were performed in a cohort of 340 patient tumors, composed of 124 adenoid cystic carcinomas (ACC), 107 salivary duct carcinomas (SDC), 64 acinic cell carcinomas, 36 mucoepidermoid carcinomas (MEC), 9 secretory carcinomas (SC), as well as 20 normal salivary glands (controls). B7-H4 expression was scored and categorized into negative (<5% expression of any intensity), low (5%-70% expression of any intensity or >70% with weak intensity), or high (>70% moderate or strong diffuse intensity). The associations between B7-H4 expression and clinicopathologic characteristics, as well as overall survival, were assessed. Among all tumors, B7-H4 expression was more prevalent in ACC (94%) compared with those of SC (67%), MEC (44%), SDC (32%), and acinic cell carcinomas (0%). Normal salivary gland tissue did not express B7-H4. High expression of B7-H4 was found exclusively in ACC (27%), SDC (11%), and MEC (8%). In SDC, B7-H4 expression was associated with female gender (P = .002) and lack of androgen receptor expression (P = .012). In ACC, B7-H4 expression was significantly associated with solid histology (P < .0001) and minor salivary gland primary (P = .02). High B7-H4 expression was associated with a poorer prognosis in ACC, regardless of clinical stage and histologic subtype. B7-H4 expression was not prognostic in the non-ACC SGC evaluated. Our comparative study revealed distinct patterns of B7-H4 expression according to SGC histology, which has potential therapeutic implications. B7-H4 expression was particularly high in solid ACC and was an independent prognostic marker in this disease but not in the other SGC assessed.


Assuntos
Neoplasias da Mama , Carcinoma de Células Acinares , Carcinoma Adenoide Cístico , Carcinoma Mucoepidermoide , Carcinoma , Neoplasias das Glândulas Salivares , Humanos , Feminino , Carcinoma Adenoide Cístico/patologia , Prognóstico , Carcinoma de Células Acinares/patologia , Neoplasias das Glândulas Salivares/patologia , Carcinoma Mucoepidermoide/patologia , Carcinoma/patologia , Glândulas Salivares/química , Glândulas Salivares/metabolismo , Glândulas Salivares/patologia , Biomarcadores Tumorais/análise
2.
Int J Mol Sci ; 24(19)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37834100

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) cells display extensive crosstalk with their surrounding environment to regulate tumor growth, immune evasion, and metastasis. Recent advances have attributed many of these interactions to intercellular communication mediated by small extracellular vesicles (sEVs), involving cancer-associated fibroblasts (CAF). To explore the impact of sEVs on monocyte lineage transition as well as the expression of checkpoint receptors and activation markers, peripheral blood monocytes from healthy subjects were exposed to PDAC-derived sEVs. Additionally, to analyze the role of sEV-associated HA in immune regulation and tissue-resident fibroblasts, monocytes and pancreatic stellate cells were cultured in the presence of PDAC sEVs with or depleted of HA. Exposure of monocytes to sEVs resulted in unique phenotypic changes in HLA-DR, PD-L1, CD86 and CD64 expression, and cytokine secretion that was HA-independent except for IL-1ß and MIP1ß. In contrast, monocyte suppression of autologous T cell proliferation was reduced following exposure to HA-low sEVs. In addition, exposure of stellate cells to sEVs upregulated the secretion of various cytokines, including MMP-9, while removal of HA from PDAC-derived sEVs attenuated the secretion of MMP-9, demonstrating the role of sEV-associated HA in regulating expression of this pro-tumorigenic cytokine from stellate cells. This observation lends credence to the findings from the TCGA database that PDAC patients with high levels of enzymes in the HA synthesis pathway had worse survival rates compared with patients having low expression of these enzymes. PDAC-derived sEVs have an immune modulatory role affecting the activation state of monocyte subtypes. However, sEV-associated HA does not affect monocyte phenotype but alters cytokine secretion and suppression of autologous T cell proliferation and induces secretion of pro-tumorigenic factors by pancreatic stellate cells (PSC), as has been seen following the conversion of PSCs to cancer-associated fibroblasts (CAFs). Interruption of the hexosamine biosynthetic pathway, activated in PDAC producing the key substrate (UDP-GlcNAc) for HA synthesis, thus, represents a potential clinical interception strategy for PDAC patients. Findings warrant further investigations of underlying mechanisms involving larger sample cohorts.


Assuntos
Carcinoma Ductal Pancreático , Vesículas Extracelulares , Neoplasias Pancreáticas , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Citocinas/metabolismo , Vesículas Extracelulares/metabolismo , Microambiente Tumoral , Células Estreladas do Pâncreas
3.
J Biol Chem ; 299(3): 102935, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36693448

RESUMO

Although cancer is a genetic disease, physical changes such as stiffening of the extracellular matrix also commonly occur in cancer. Cancer cells sense and respond to extracellular matrix stiffening through the process of mechanotransduction. Cancer cell mechanotransduction can enhance cancer-promoting cell behaviors such as survival signaling, proliferation, and migration. Glycans, carbohydrate-based polymers, have recently emerged as important mediators and/or modulators of cancer cell mechanotransduction. Stiffer tumors are characterized by increased glycan content on cancer cells and their associated extracellular matrix. Here we review the role of cancer-associated glycans in coupled mechanical and biochemical alterations during cancer progression. We discuss the recent evidence on how increased expression of different glycans, in the form of glycoproteins and proteoglycans, contributes to both mechanical changes in tumors and corresponding cancer cell responses. We conclude with a summary of emerging tools that can be used to modify glycans for future studies in cancer mechanobiology.


Assuntos
Mecanotransdução Celular , Neoplasias , Polissacarídeos , Humanos , Biofísica , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Mecanotransdução Celular/fisiologia , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Polissacarídeos/metabolismo
4.
Adv Exp Med Biol ; 1221: 331-349, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32274716

RESUMO

It has been speculated for many years that heparanase plays an important role in the progression of cancer due largely to the finding that its expression is weak or absent in normal tissues but generally as tumors become more aggressive heparanase expression increases. However, it is only in the last decade or so that we have begun to understand the molecular mechanism behind the sinister role that heparanase plays in cancer. In this review, we describe the many functions of heparanase in promoting the growth, angiogenesis and metastasis of multiple myeloma, a devastating cancer that localizes predominantly within the bone marrow and spreads throughout the skeletal system devouring bone and ultimately leading to death of almost all patients diagnosed with this disease. We also explore recent discoveries related to how heparanase primes exosome biogenesis and how heparanase enhances myeloma tumor chemoresistance. Discovery of these multiple tumor-promoting pathways that are driven by heparanase identified the enzyme as an ideal target for therapy, an approach recently tested in a Phase I trial in myeloma patients.


Assuntos
Glucuronidase/metabolismo , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Exossomos , Glucuronidase/antagonistas & inibidores , Humanos , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/tratamento farmacológico
5.
Methods Mol Biol ; 1952: 233-244, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30825179

RESUMO

Exosomes are small vesicles of endosomal origin secreted by most cell types. Recent studies have identified exosomes as important mediators of intercellular communication and as important source materials for many clinical applications, including minimal invasive disease diagnosis. Exosomes have been purified from in vitro cell culture supernatants by many different methods; however the most simple and reliable method involves purification by ultracentrifugation. This chapter describes a detailed protocol for isolating exosomes from cell culture-conditioned medium using ultracentrifugation and their characterization based upon size, number, and protein expression by several complementary methods such as transmission electron microscopy, nanoparticle tracking analysis, western blotting, and flow cytometry.


Assuntos
Exossomos/química , Exossomos/ultraestrutura , Ultracentrifugação/métodos , Animais , Western Blotting/métodos , Técnicas de Cultura de Células/métodos , Meios de Cultivo Condicionados/química , Citometria de Fluxo/métodos , Humanos , Microscopia Eletrônica de Transmissão/métodos , Proteínas/análise
6.
Matrix Biol ; 65: 104-118, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28888912

RESUMO

The heparan sulfate-degrading enzyme heparanase promotes the progression of many cancers by driving tumor cell proliferation, metastasis and angiogenesis. Heparanase accomplishes this via multiple mechanisms including its recently described effect on enhancing biogenesis of tumor exosomes. Because we recently discovered that heparanase expression is upregulated in myeloma cells that survive chemotherapy, we were prompted to investigate the impact of anti-myeloma drugs on exosome biogenesis. When myeloma cells were exposed to the commonly utilized anti-myeloma drugs bortezomib, carfilzomib or melphalan, exosome secretion by the cells was dramatically enhanced. These chemotherapy-induced exosomes (chemoexosomes) have a proteome profile distinct from cells not exposed to drug including a dramatic elevation in the level of heparanase present as exosome cargo. The chemoexosome heparanase was not found inside the chemoexosome, but was present on the exosome surface where it was capable of degrading heparan sulfate embedded within an extracellular matrix. When exposed to myeloma cells, chemoexosomes transferred their heparanase cargo to those cells, enhancing their heparan sulfate degrading activity and leading to activation of ERK signaling and an increase in shedding of the syndecan-1 proteoglycan. Exposure of chemoexosomes to macrophages enhanced their secretion of TNF-α, an important myeloma growth factor. Moreover, chemoexosomes stimulated macrophage migration and this effect was blocked by H1023, a monoclonal antibody that inhibits heparanase enzymatic activity. These data suggest that anti-myeloma therapy ignites a burst of exosomes having a high level of heparanase that remodels extracellular matrix and alters tumor and host cell behaviors that likely contribute to chemoresistance and eventual patient relapse. SUMMARY: We find that anti-myeloma chemotherapy dramatically stimulates secretion of exosomes and alters exosome composition. Exosomes secreted during therapy contain high levels of heparanase on their surface that can degrade ECM and also can be transferred to both tumor and host cells, altering their behavior in ways that may enhance tumor survival and progression.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Exossomos/metabolismo , Matriz Extracelular/metabolismo , Glucuronidase/metabolismo , Mieloma Múltiplo/metabolismo , Animais , Bortezomib/farmacologia , Linhagem Celular Tumoral , Tratamento Farmacológico , Exossomos/efeitos dos fármacos , Exossomos/enzimologia , Regulação Neoplásica da Expressão Gênica , Humanos , Melfalan/farmacologia , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Oligopeptídeos/farmacologia , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
7.
Oncotarget ; 8(43): 73723-73732, 2017 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-29088739

RESUMO

Tumor cells produce and utilize exosomes to promote tumor growth and metastasis. Tumor-cell-derived exosomes deliver cargos that partially mimic the contents of the parent cell to nearby or distant normal or abnormal cells, thereby reprogramming the recipient cells to support tumor progression. Mechanisms by which tumor-derived exosomes subserve the tumor are under intense investigation. Here we demonstrate a critical role of the chondroitin sulfate proteoglycan serglycin in regulating the protein cargo and functions of myeloma cell-derived exosomes. Previous studies have shown that serglycin, the only known intracellular proteoglycan, functions mainly in the storage of basically charged components within the intracellular granules/vesicles via serglycin's densely clustered, negatively charged glycosaminoglycan chains. Here we demonstrate that serglycin plays a critical role in the protein cargo loading of tumor-derived exosomes. Serglycin was detected in exosomes derived from cell culture supernatants of human myeloma cell lines and serum of myeloma patients. Mass spectrometry analysis of exosomal proteins identified significantly fewer protein components within exosomes derived from serglycin-knockdown myeloma cells than within exosomes from control cells. On gene ontology analysis, exosomes derived from serglycin-knockdown cells, but not from control cells, lacked many proteins that are required for mediating different cellular processes. In functional assays, exosomes from serglycin-knockdown cells failed to induce an invasive phenotype in myeloma cells and failed to promote migration of macrophages. These findings reveal that serglycin plays an important role in maintaining the protein cargo in tumor-derived exosomes and suggest that targeting serglycin may temper the influence of these exosomes on cancer progression.

8.
J Biol Chem ; 291(4): 1652-1663, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26601950

RESUMO

Exosomes regulate cell behavior by binding to and delivering their cargo to target cells; however, the mechanisms mediating exosome-cell interactions are poorly understood. Heparan sulfates on target cell surfaces can act as receptors for exosome uptake, but the ligand for heparan sulfate on exosomes has not been identified. Using exosomes isolated from myeloma cell lines and from myeloma patients, we identify exosomal fibronectin as a key heparan sulfate-binding ligand and mediator of exosome-cell interactions. We discovered that heparan sulfate plays a dual role in exosome-cell interaction; heparan sulfate on exosomes captures fibronectin, and on target cells it acts as a receptor for fibronectin. Removal of heparan sulfate from the exosome surface releases fibronectin and dramatically inhibits exosome-target cell interaction. Antibody specific for the Hep-II heparin-binding domain of fibronectin blocks exosome interaction with tumor cells or with marrow stromal cells. Regarding exosome function, fibronectin-mediated binding of exosomes to myeloma cells activated p38 and pERK signaling and expression of downstream target genes DKK1 and MMP-9, two molecules that promote myeloma progression. Antibody against fibronectin inhibited the ability of myeloma-derived exosomes to stimulate endothelial cell invasion. Heparin or heparin mimetics including Roneparstat, a modified heparin in phase I trials in myeloma patients, significantly inhibited exosome-cell interactions. These studies provide the first evidence that fibronectin binding to heparan sulfate mediates exosome-cell interactions, revealing a fundamental mechanism important for exosome-mediated cross-talk within tumor microenvironments. Moreover, these results imply that therapeutic disruption of fibronectin-heparan sulfate interactions will negatively impact myeloma tumor growth and progression.


Assuntos
Comunicação Celular , Exossomos/metabolismo , Fibronectinas/metabolismo , Mieloma Múltiplo/metabolismo , Linhagem Celular Tumoral , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Exossomos/genética , Fibronectinas/genética , Heparina/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/fisiopatologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
J Biol Chem ; 289(38): 26584-26596, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25107907

RESUMO

Galnt3, UDP-N-acetyl-α-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 3, transfers N-acetyl-D-galactosamine to serine and threonine residues, initiating mucin type O-glycosylation of proteins. We searched the target genes of Runx2, which is an essential transcription factor for chondrocyte maturation, in chondrocytes and found that Galnt3 expression was up-regulated by Runx2 and severely reduced in Runx2(-/-) cartilaginous skeletons. To investigate the function of Galnt3 in chondrocytes, we generated Galnt3(-/-) mice and chondrocyte-specific Galnt3 transgenic mice under the control of the Col2a1 promoter-enhancer. Galnt3(-/-) mice showed a delay in endochondral ossification and shortened limbs at embryonic day 16.5, suggesting that Galnt3 is involved in chondrocyte maturation. Galnt3 transgenic mice presented dwarfism, the chondrocyte maturation was retarded, the cell cycle in chondrocytes was accelerated, premature chondrocyte apoptosis occurred, and the growth plates were disorganized. The binding of Vicia villosa agglutinin, which recognizes the Tn antigen (GalNAc-O-Ser/Thr), was drastically increased in chondrocytes, and aggrecan (Acan) was highly enriched with Tn antigen. However, safranin O staining, which recognizes glycosaminoglycans (GAGs), and Acan were severely reduced. Chondroitin sulfate was reduced in amount, but the elongation of chondroitin sulfate chains had not been severely disturbed in the isolated GAGs. These findings indicate that overexpression of Galnt3 in chondrocytes caused dwarfism due to the increase of mucin-type O-glycans and the reduction of GAGs, probably through competition with xylosyltransferases, which initiate GAG chains by attaching O-linked xylose to serine residues, suggesting a negative effect of Galnt family proteins on Acan deposition in addition to the positive effect of Galnt3 on chondrocyte maturation.


Assuntos
Condrócitos/metabolismo , Sulfatos de Condroitina/metabolismo , Nanismo/metabolismo , N-Acetilgalactosaminiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Agrecanas/genética , Agrecanas/metabolismo , Animais , Apoptose , Cartilagem/metabolismo , Cartilagem/patologia , Proliferação de Células , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Expressão Gênica , Glicosilação , Lâmina de Crescimento/metabolismo , Lâmina de Crescimento/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Acetilgalactosaminiltransferases/genética , Osteogênese , Polipeptídeo N-Acetilgalactosaminiltransferase
10.
J Biol Chem ; 289(9): 5499-509, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24403068

RESUMO

Recently, it was discovered that serglycin, a hematopoietic cell proteoglycan, is the major proteoglycan expressed and constitutively secreted by multiple myeloma (MM) cells. High levels of serglycin are present in the bone marrow aspirates of at least 30% of newly diagnosed MM patients. However, its contribution to the pathophysiology of MM is unknown. Here, we show that serglycin knockdown (by ∼85% compared with normal levels), using lentiviral shRNA, dramatically attenuated MM tumor growth in mice with severe combined immunodeficiency. Tumors formed from cells deficient in serglycin exhibited diminished levels of hepatocyte growth factor expression and impaired development of blood vessels, indicating that serglycin may affect tumor angiogenesis. Furthermore, knockdown of serglycin significantly decreased MM cell adhesion to bone marrow stromal cells and collagen I. Even though serglycin proteoglycan does not have a transmembrane domain, flow cytometry showed that serglycin is present on the MM cell surface, and attachment to the cell surface is, at least in part, dependent on its chondroitin sulfate side chains. Co-precipitation of serglycin from conditioned medium of MM cells using a CD44-Fc chimera suggests that CD44 is the cell surface-binding partner for serglycin, which therefore may serve as a major ligand for CD44 at various stages during myeloma progression. Finally, we demonstrate that serglycin mRNA expression in MM cells is up-regulated by activin, a predominant cytokine among those increased in MM patients with osteolytic lesions. These studies provide direct evidence for a critical role for serglycin in MM pathogenesis and show that targeting serglycin may provide a novel therapeutic approach for MM.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/biossíntese , Neovascularização Patológica/metabolismo , Proteoglicanas/biossíntese , Proteínas de Transporte Vesicular/biossíntese , Ativinas/genética , Ativinas/metabolismo , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Adesão Celular/genética , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/genética , Camundongos , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Proteoglicanas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Células Estromais/metabolismo , Células Estromais/patologia , Proteínas de Transporte Vesicular/genética
11.
FEBS J ; 280(10): 2294-306, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23374281

RESUMO

Heparanase is an endoglucuronidase that cleaves heparan sulfate chains of proteoglycans. In many malignancies, high heparanase expression and activity correlate with an aggressive tumour phenotype. A major consequence of heparanase action in cancer is a robust up-regulation of growth factor expression and increased shedding of syndecan-1 (a transmembrane heparan sulfate proteoglycan). Substantial evidence indicates that heparanase and syndecan-1 work together to drive growth factor signalling and regulate cell behaviours that enhance tumour growth, dissemination, angiogenesis and osteolysis. Preclinical and clinical studies have demonstrated that therapies targeting the heparanase/syndecan-1 axis hold promise for blocking the aggressive behaviour of cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glucuronidase/metabolismo , Neoplasias/tratamento farmacológico , Sindecana-1/metabolismo , Animais , Antineoplásicos/farmacologia , Materiais Biomiméticos/farmacologia , Núcleo Celular/enzimologia , Núcleo Celular/patologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Exossomos/metabolismo , Exossomos/patologia , Glucuronidase/antagonistas & inibidores , Heparitina Sulfato/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Neoplasias/enzimologia , Neoplasias/patologia , Transdução de Sinais , Sindecana-1/genética
12.
J Biol Chem ; 288(14): 10093-10099, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23430739

RESUMO

Emerging evidence indicates that exosomes play a key role in tumor-host cross-talk and that exosome secretion, composition, and functional capacity are altered as tumors progress to an aggressive phenotype. However, little is known regarding the mechanisms that regulate these changes. Heparanase is an enzyme whose expression is up-regulated as tumors become more aggressive and is associated with enhanced tumor growth, angiogenesis, and metastasis. We have discovered that in human cancer cells (myeloma, lymphoblastoid, and breast cancer), when expression of heparanase is enhanced or when tumor cells are exposed to exogenous heparanase, exosome secretion is dramatically increased. Heparanase enzyme activity is required for robust enhancement of exosome secretion because enzymatically inactive forms of heparanase, even when present in high amounts, do not dramatically increase exosome secretion. Heparanase also impacts exosome protein cargo as reflected by higher levels of syndecan-1, VEGF, and hepatocyte growth factor in exosomes secreted by heparanase-high expressing cells as compared with heparanase-low expressing cells. In functional assays, exosomes from heparanase-high cells stimulated spreading of tumor cells on fibronectin and invasion of endothelial cells through extracellular matrix better than did exosomes secreted by heparanase-low cells. These studies reveal that heparanase helps drive exosome secretion, alters exosome composition, and facilitates production of exosomes that impact both tumor and host cell behavior, thereby promoting tumor progression.


Assuntos
Exossomos/metabolismo , Regulação Enzimológica da Expressão Gênica , Glucuronidase/química , Linhagem Celular Tumoral , DNA Complementar/metabolismo , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Heparitina Sulfato/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Mieloma Múltiplo/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/enzimologia , Sindecana-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
J Biol Chem ; 287(49): 41288-96, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23048032

RESUMO

ERK signaling regulates proliferation, survival, drug resistance, and angiogenesis in cancer. Although the mechanisms regulating ERK activation are not fully understood, we previously demonstrated that ERK phosphorylation is elevated by heparanase, an enzyme associated with aggressive behavior of many cancers. In the present study, myeloma cell lines expressing either high or low levels of heparanase were utilized to determine how heparanase stimulates ERK signaling. We discovered that the insulin receptor was abundant on cells expressing either high or low levels of heparanase, but the receptor was highly phosphorylated in heparanase-high cells compared with heparanase-low cells. In addition, protein kinase C activity was elevated in heparanase-high cells, and this enhanced expression of insulin receptor substrate-1 (IRS-1), the principle intracellular substrate for phosphorylation by the insulin receptor. Blocking insulin receptor function with antibody or a small molecule inhibitor or knockdown of IRS-1 expression using shRNA diminished heparanase-mediated ERK activation in the tumor cells. In addition, up-regulation of the insulin signaling pathway by heparanase and the resulting ERK activation were dependent on heparanase retaining its enzyme activity. These results reveal a novel mechanism whereby heparanase enhances activation of the insulin receptor signaling pathway leading to ERK activation and modulation of myeloma behavior.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glucuronidase/biossíntese , Mieloma Múltiplo/enzimologia , Receptor de Insulina/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Citometria de Fluxo/métodos , Hormônio do Crescimento/metabolismo , Humanos , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Modelos Biológicos , Fosforilação , Proteína Quinase C/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
14.
J Biol Chem ; 287(5): 2935-42, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22094467

RESUMO

Chondroitin sulfate/dermatan sulfate (CS/DS) proteoglycans, major components of the central nervous system, have the potential to interact with a wide range of growth factors and neurotrophic factors that influence neuronal migration, axon guidance pathways, and neurite outgrowth. Recent studies have also revealed the role of CS/DS chains in the orchestration of the neural stem/progenitor cell micromilieu. Individual functional proteins recognize a set of multiple overlapping oligosaccharide sequences decorated to give different sulfation patterns, which are termed here "wobble CS/DS oligosaccharide motifs," and induce signaling pathways essential for the proliferation, self-renewal, and cell lineage commitment of neural stem/progenitor cells.


Assuntos
Diferenciação Celular/fisiologia , Sulfatos de Condroitina/metabolismo , Dermatan Sulfato/metabolismo , Células-Tronco Neurais/fisiologia , Proteoglicanas/metabolismo , Animais , Configuração de Carboidratos , Movimento Celular/fisiologia , Humanos , Células-Tronco Neurais/citologia , Transdução de Sinais/fisiologia
15.
J Biol Chem ; 286(35): 30377-30383, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21757697

RESUMO

Heparanase acts as a master regulator of the aggressive tumor phenotype in part by enhancing expression of proteins known to drive tumor progression (e.g. VEGF, MMP-9, hepatocyte growth factor (HGF), and RANKL). However, the mechanism whereby this enzyme regulates gene expression remains unknown. We previously reported that elevation of heparanase levels in myeloma cells causes a dramatic reduction in the amount of syndecan-1 in the nucleus. Because syndecan-1 has heparan sulfate chains and because exogenous heparan sulfate has been shown to inhibit the activity of histone acetyltransferase (HAT) enzymes in vitro, we hypothesized that the reduction in nuclear syndecan-1 in cells expressing high levels of heparanase would result in increased HAT activity leading to stimulation of protein transcription. We found that myeloma cells or tumors expressing high levels of heparanase and low levels of nuclear syndecan-1 had significantly higher levels of HAT activity when compared with cells or tumors expressing low levels of heparanase. High levels of HAT activity in heparanase-high cells were blocked by SST0001, an inhibitor of heparanase. Restoration of high syndecan-1 levels in heparanase-high cells diminished nuclear HAT activity, establishing syndecan-1 as a potent inhibitor of HAT. Exposure of heparanase-high cells to anacardic acid, an inhibitor of HAT activity, significantly suppressed their expression of VEGF and MMP-9, two genes known to be up-regulated following elevation of heparanase. These results reveal a novel mechanistic pathway driven by heparanase expression, which leads to decreased nuclear syndecan-1, increased HAT activity, and up-regulation of transcription of multiple genes that drive an aggressive tumor phenotype.


Assuntos
Núcleo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica , Glucuronidase/metabolismo , Histona Acetiltransferases/metabolismo , Sindecana-1/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Fenótipo , Proteoglicanas/metabolismo , Transcrição Gênica , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Cancer Lett ; 297(2): 231-43, 2010 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-20831981

RESUMO

A novel pyranoside mimetic compound, DMBO (2-(2,6-difluorophenyl)-5-(4-methoxyphenyl)-1-oxa-3-azaspiro[5.5]undecane), was designed and synthesized. The sugar mimicking behavior of DMBO was addressed by its ability to bind several growth factors/cytokines such as vascular endothelial growth factor (VEGF), heparin-binding epidermal growth factor-like growth factor (HB-EGF), and tumor necrosis factor (TNF)-α as demonstrated by the recently developed surface plasmon resonance assay. DMBO exhibited strong anti-proliferation activity in vitro against tumor cells including a highly metastatic murine osteosarcoma cell line LM8G7 that secretes VEGF as well as two human ovarian cell lines, OVSAHO and SKOV-3, which secrete TNF-α and HB-EGF respectively. Furthermore, DMBO inhibited the metastatic activity to the mouse liver of LM8G7 cells injected from a lateral tail vein, and affected the heparan-degrading activity of LM8G7 cells. Here, we report that DMBO acts as a human heparanase inhibitor in vitro possibly as a substrate mimetic. DMBO also inhibited the migration and invasion of LM8G7 cells and angiogenic events such as endothelial cell proliferation, migration and capillary tube-like formation in vitro. More prominently, the administration of DMBO with heparin resulted in synergistic anti-tumor effects in mouse modelofosteosarcoma. These preclinical data shows the potential anti-cancer effects of DMBO.


Assuntos
Benzoxazinas/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Glucuronidase/antagonistas & inibidores , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/secundário , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ligação Proteica
17.
Blood ; 115(12): 2449-57, 2010 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-20097882

RESUMO

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.


Assuntos
Glucuronidase/metabolismo , Mieloma Múltiplo , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Sindecana-1/metabolismo , Aorta/citologia , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Endotélio/patologia , Matriz Extracelular/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Mieloma Múltiplo/fisiopatologia , Invasividade Neoplásica , Técnicas de Cultura de Órgãos , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Histochem Cell Biol ; 132(1): 117-27, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19360434

RESUMO

Dermatan sulfate (DS) expression in normal tissue and ovarian cancer was investigated using the novel, phage display-derived antibody GD3A12 that was selected against embryonic glycosaminoglycans (GAGs). Antibody GD3A12 was especially reactive with DS rich in IdoA-GalNAc4S disaccharide units. IdoA residues are important for antibody recognition as DS polymers with low numbers of IdoA residues were less reactive, and expression of the DS epimerase in ovarian carcinoma cells was associated with expression of the GD3A12 epitope. Moreover, staining of antibody GD3A12 was abolished by chondroitinase-B lyase digestion. Expression of DS domains defined by antibody GD3A12 was confined to connective tissue of most organs examined and presented as a typical fibrillar-type of staining. Differential expression of the DS epitopes recognized by antibodies GD3A12 and LKN1 (4/2,4 di-O-sulfated DS) was best seen in thymus and spleen, indicating differential expression of various DS domains in these organs. In ovarian carcinomas strong DS expression was found in the stromal parts, and occasionally on tumor cells. Partial co-localization in ovarian carcinomas was observed with decorin, versican and type I collagen suggesting a uniform distribution of this specific DS epitope. This unique anti-DS antibody may be instrumental to investigate the function, expression, and localization of specific DS domains in health and disease.


Assuntos
Adenocarcinoma/metabolismo , Anticorpos/imunologia , Dermatan Sulfato/metabolismo , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , Animais , Linhagem Celular Tumoral , Dermatan Sulfato/imunologia , Embrião de Mamíferos/metabolismo , Epitopos , Feminino , Humanos , Masculino , Camundongos , Especificidade de Órgãos , Ratos , Ratos Wistar
19.
J Biol Chem ; 283(47): 32628-36, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18812315

RESUMO

High levels of heparanase are an indicator of poor prognosis in myeloma patients, and up-regulation of the enzyme enhances tumor growth, angiogenesis, and metastasis in animal models. At least part of the impact of heparanase in driving the aggressive tumor phenotype is due to its effect on increasing the expression and shedding of the heparan sulfate proteoglycan syndecan-1, a molecule known to promote myeloma progression. The present work demonstrated that elevation in heparanase expression in myeloma cells stimulates sustained ERK phosphorylation that in turn drives MMP-9 expression. In addition, urokinase-type plasminogen activator (uPA) and uPA receptor expression levels increased, and blocking the proteolytic activation of either MMP-9 or uPA inhibited the heparanase-induced increase in syndecan-1 shedding. Together these data provide a mechanism for heparanase-induced syndecan-1 shedding and, more importantly, demonstrate that heparanase activity in myeloma cells can lead to increased levels of proteases that are known to play important roles in the aggressive behavior of myeloma tumors. This in addition to its other known biological roles, indicates that heparanase acts as a master regulator of the aggressive tumor phenotype by up-regulating protease expression and activity within the tumor microenvironment.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glucuronidase/metabolismo , Mieloma Múltiplo/patologia , Progressão da Doença , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Modelos Biológicos , Invasividade Neoplásica , Fenótipo , Fosforilação , Transdução de Sinais , Sindecana-1/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
20.
Am J Pathol ; 171(4): 1324-33, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17717144

RESUMO

Chondroitin sulfate (CS) is abundantly present in the tumor stroma, and tumor-specific CS modifications might be potential targets to influence tumor development. We applied the phage display technology to select antibodies that identify these tumor-specific CS modifications. Antibody GD3G7 was selected against embryonic glycosaminoglycans, and it reacted strongly with CS-E (rich in GlcA-GalNAc4S6S units). In ovarian adenocarcinomas, strong expression of this CS-E epitope was found in the extracellular matrix, and occasionally on tumor cells. No expression was found in normal ovary and cystadenomas. Differential expression was found in ovarian carcinoma cell lines, which correlated with the gene expression of the GalNAc4S-6st enzyme, involved in biosynthesis of CS-E. Vascular endothelial growth factor (VEGF)-sensitive fenestrated (in normal tissues) and tumor blood vessels were both identified by antibody GD3G7, which might implicate a role for CS-E in VEGF biology. VEGF bound to CS-E and antibody GD3G7 could compete for binding of VEGF to CS-E. In conclusion, antibody GD3G7 identified rare CS-E-like structures that were strongly expressed in ovarian adenocarcinomas. This antibody might therefore be instrumental for identifying tumor-related CS alterations.


Assuntos
Adenocarcinoma/metabolismo , Anticorpos Antineoplásicos/imunologia , Sulfatos de Condroitina/análise , Sulfatos de Condroitina/metabolismo , Neoplasias Ovarianas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenocarcinoma/química , Adenocarcinoma/genética , Animais , Linhagem Celular Tumoral , Sulfatos de Condroitina/imunologia , Epitopos/análise , Epitopos/imunologia , Epitopos/metabolismo , Feminino , Expressão Gênica , Glicosaminoglicanos/imunologia , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Neoplasias Ovarianas/química , Neoplasias Ovarianas/genética , Biblioteca de Peptídeos , Ratos , Sulfotransferases/química , Sulfotransferases/genética , Sulfotransferases/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...