Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34099563

RESUMEN

Tetraspanins are an evolutionary conserved family of proteins involved in multiple aspects of cell physiology, including proliferation, migration and invasion, protein trafficking, and signal transduction; yet their detailed mechanism of action is unknown. Tetraspanins have no known natural ligands, but their engagement by antibodies has begun to reveal their role in cell biology. Studies of tetraspanin knockout mice and of germline mutations in humans have highlighted their role under normal and pathological conditions. Previously, we have shown that mice deficient in the tetraspanin CD81 developed fewer breast cancer metastases compared to their wild-type (WT) counterparts. Here, we show that a unique anti-human CD81 antibody (5A6) effectively halts invasion of triple-negative breast cancer (TNBC) cell lines. We demonstrate that 5A6 induces CD81 clustering at the cell membrane and we implicate JAM-A protein in the ability of this antibody to inhibit tumor cell invasion and migration. Furthermore, in a series of in vivo studies we demonstrate that this antibody inhibits metastases in xenograft models, as well as in syngeneic mice bearing a mouse tumor into which we knocked in the human CD81 epitope recognized by the 5A6 antibody.


Asunto(s)
Neoplasias de la Mama/patología , Tetraspanina 28/metabolismo , Animales , Anticuerpos/farmacología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Epítopos/metabolismo , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Receptores de Superficie Celular/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nat Cancer ; 2(1): 18-33, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-35121890

RESUMEN

Innate pattern recognition receptor agonists, including Toll-like receptors (TLRs), alter the tumor microenvironment and prime adaptive antitumor immunity. However, TLR agonists present toxicities associated with widespread immune activation after systemic administration. To design a TLR-based therapeutic suitable for systemic delivery and capable of safely eliciting tumor-targeted responses, we developed immune-stimulating antibody conjugates (ISACs) comprising a TLR7/8 dual agonist conjugated to tumor-targeting antibodies. Systemically administered human epidermal growth factor receptor 2 (HER2)-targeted ISACs were well tolerated and triggered a localized immune response in the tumor microenvironment that resulted in tumor clearance and immunological memory. Mechanistically, ISACs required tumor antigen recognition, Fcγ-receptor-dependent phagocytosis and TLR-mediated activation to drive tumor killing by myeloid cells and subsequent T-cell-mediated antitumor immunity. ISAC-mediated immunological memory was not limited to the HER2 ISAC target antigen since ISAC-treated mice were protected from rechallenge with the HER2- parental tumor. These results provide a strong rationale for the clinical development of ISACs.


Asunto(s)
Inmunoterapia , Neoplasias , Inmunidad Adaptativa , Animales , Antígenos de Neoplasias , Inmunoterapia/métodos , Ratones , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
3.
Clin Exp Metastasis ; 37(3): 413-424, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32335861

RESUMEN

Patient-derived orthotopic xenograft (PDOX) models have been verified as a useful method for studying human cancers in mice. Previous studies on the extent of metastases in these models have been limited by the necessity of welfare euthanasia (primary tumors reaching threshold size), at which point metastases may only be micrometers in diameter, few in number, and solely identified by step-sectioning of formalin-fixed paraffin-embedded tissue. These small micro-metastases are less suitable for many downstream molecular analyses than macro-metastases. Resection of the primary tumor by survival surgery has been proven to allow further time for metastases to grow. Although PDOX models of triple-negative breast cancer (TNBC) shed circulating tumor cells (CTCs) into the bloodstream and metastasize, similar to human TNBC, little data has been collected in these TNBC PDOX models regarding the association between CTC characteristics and distant metastasis following excision of the primary tumor xenograft. This study assembles a timeline of PDOX tumor shedding and metastatic tumor progression before and after tumor excision surgery. We report the ability to use tumorectomies to increase the lifespan of TNBC PDOX models with the potential to obtain larger metastases. CTC clusters and CTCs expressing a mesenchymal marker (vimentin) were associated with metastatic burden in lung and liver. The data collected through these experiments will guide the further use of PDOX models in studying metastatic TNBC.


Asunto(s)
Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Micrometástasis de Neoplasia/patología , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Recuento de Células , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Células Neoplásicas Circulantes/patología , Neoplasias de la Mama Triple Negativas/cirugía , Vimentina/metabolismo
4.
Matrix Biol ; 88: 53-68, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31812535

RESUMEN

Heparanase is known to enhance the progression of many cancer types and is associated with poor patient prognosis. We recently reported that after patients with multiple myeloma were treated with high dose chemotherapy, the tumor cells that emerged upon relapse expressed a much higher level of heparanase than was present prior to therapy. Because tumor cells having stemness properties are thought to seed tumor relapse, we investigated whether heparanase had a role in promoting myeloma stemness. When plated at low density and grown in serum-free conditions that support survival and expansion of stem-like cells, myeloma cells expressing a low level of heparanase formed tumor spheroids poorly. In contrast, cells expressing a high level of heparanase formed significantly more and larger spheroids than did the heparanase low cells. Importantly, heparanase-low expressing cells exhibited plasticity and were induced to exhibit stemness properties when exposed to recombinant heparanase or to exosomes that contained a high level of heparanase cargo. The spheroid-forming heparanase-high cells had elevated expression of GLI1, SOX2 and ALDH1A1, three genes known to be associated with myeloma stemness. Inhibitors that block the heparan sulfate degrading activity of heparanase significantly diminished spheroid formation and expression of stemness genes implying a direct role of the enzyme in regulating stemness. Blocking the NF-κB pathway inhibited spheroid formation and expression of stemness genes demonstrating a role for NF-κB in heparanase-mediated stemness. Myeloma cells made deficient in heparanase exhibited decreased stemness properties in vitro and when injected into mice they formed tumors poorly compared to the robust tumorigenic capacity of cells expressing higher levels of heparanase. These studies reveal for the first time a role for heparanase in promoting cancer stemness and provide new insight into its function in driving tumor progression and its association with poor prognosis in cancer patients.


Asunto(s)
Regulación hacia Abajo , Glucuronidasa/genética , Mieloma Múltiple/patología , Células Madre Neoplásicas/patología , Familia de Aldehído Deshidrogenasa 1/genética , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Exosomas/enzimología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Mieloma Múltiple/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Retinal-Deshidrogenasa/genética , Factores de Transcripción SOXB1/genética , Esferoides Celulares/citología , Proteína con Dedos de Zinc GLI1/genética
5.
Breast Cancer Res ; 21(1): 98, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31462307

RESUMEN

BACKGROUND: Circulating tumor cells (CTCs) represent a temporal "snapshot" of a patient's cancer and changes that occur during disease evolution. There is an extensive literature studying CTCs in breast cancer patients, and particularly in those with metastatic disease. In parallel, there is an increasing use of patient-derived models in preclinical investigations of human cancers. Yet studies are still limited demonstrating CTC shedding and metastasis formation in patient-derived models of breast cancer. METHODS: We used seven patient-derived orthotopic xenograft (PDOX) models generated from triple-negative breast cancer (TNBC) patients to study CTCs and distant metastases. Tumor fragments from PDOX tissue from each of the seven models were implanted into 57 NOD scid gamma (NSG) mice, and tumor growth and volume were monitored. Human CTC capture from mouse blood was first optimized on the marker-agnostic Vortex CTC isolation platform, and whole blood was processed from 37 PDOX tumor-bearing mice. RESULTS: Staining and imaging revealed the presence of CTCs in 32/37 (86%). The total number of CTCs varied between different PDOX tumor models and between individual mice bearing the same PDOX tumors. CTCs were heterogeneous and showed cytokeratin (CK) positive, vimentin (VIM) positive, and mixed CK/VIM phenotypes. Metastases were detected in the lung (20/57, 35%), liver (7/57, 12%), and brain (1/57, less than 2%). The seven different PDOX tumor models displayed varying degrees of metastatic potential, including one TNBC PDOX tumor model that failed to generate any detectable metastases (0/8 mice) despite having CTCs present in the blood of 5/5 tested, suggesting that CTCs from this particular PDOX tumor model may typify metastatic inefficiency. CONCLUSION: PDOX tumor models that shed CTCs and develop distant metastases represent an important tool for investigating TNBC.


Asunto(s)
Neoplasias Mamarias Experimentales/patología , Células Neoplásicas Circulantes/patología , Neoplasias de la Mama Triple Negativas/patología , Animales , Biomarcadores de Tumor/metabolismo , Encéfalo/patología , Recuento de Células , Línea Celular Tumoral , Femenino , Humanos , Queratinas/metabolismo , Hígado/patología , Pulmón/patología , Ratones , Ratones Mutantes , Metástasis de la Neoplasia , Trasplante de Neoplasias , Células Neoplásicas Circulantes/metabolismo , Vimentina/metabolismo
6.
Nano Lett ; 19(4): 2334-2342, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30895796

RESUMEN

Optical coherence tomography (OCT) can be utilized with significant speckle reduction techniques and highly scattering contrast agents for non-invasive, contrast-enhanced imaging of living tissues at the cellular scale. The advantages of reduced speckle noise and improved targeted contrast can be harnessed to track objects as small as 2 µm in vivo, which enables applications for cell tracking and quantification in living subjects. Here we demonstrate the use of large gold nanorods as contrast agents for detecting individual micron-sized polystyrene beads and single myeloma cells in blood circulation using speckle-modulating OCT. This report marks the first time that OCT has been used to detect individual cells within blood in vivo. This technical capability unlocks exciting opportunities for dynamic detection and quantification of tumor cells circulating in living subjects.


Asunto(s)
Medios de Contraste/farmacología , Mieloma Múltiple/sangre , Nanotubos/química , Células Neoplásicas Circulantes/patología , Animales , Medios de Contraste/química , Oro/química , Humanos , Ratones , Mieloma Múltiple/patología , Poliestirenos/química , Análisis de la Célula Individual/métodos , Tomografía de Coherencia Óptica/métodos
7.
SLAS Technol ; 23(1): 16-29, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29355087

RESUMEN

Tumor tissue biopsies are invasive, costly, and collect a limited cell population not completely reflective of patient cancer cell diversity. Circulating tumor cells (CTCs) can be isolated from a simple blood draw and may be representative of the diverse biology from multiple tumor sites. The VTX-1 Liquid Biopsy System was designed to automate the isolation of clinically relevant CTC populations, making the CTCs available for easy analysis. We present here the transition from a cutting-edge microfluidic innovation in the lab to a commercial, automated system for isolating CTCs directly from whole blood. As the technology evolved into a commercial system, flexible polydimethylsiloxane microfluidic chips were replaced by rigid poly(methyl methacrylate) chips for a 2.2-fold increase in cell recovery. Automating the fluidic processing with the VTX-1 further improved cancer cell recovery by nearly 1.4-fold, with a 2.8-fold decrease in contaminating white blood cells and overall improved reproducibility. Two isolation protocols were optimized that favor either the cancer cell recovery (up to 71.6% recovery) or sample purity (≤100 white blood cells/mL). The VTX-1's performance was further tested with three different spiked breast or lung cancer cell lines, with 69.0% to 79.5% cell recovery. Finally, several cancer research applications are presented using the commercial VTX-1 system.


Asunto(s)
Automatización de Laboratorios/métodos , Células Sanguíneas , Separación Celular/métodos , Biopsia Líquida/métodos , Microfluídica/métodos , Células Neoplásicas Circulantes , Automatización de Laboratorios/instrumentación , Separación Celular/instrumentación , Humanos , Biopsia Líquida/instrumentación , Microfluídica/instrumentación , Reproducibilidad de los Resultados
8.
Matrix Biol ; 65: 104-118, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28888912

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Exosomas/metabolismo , Matriz Extracelular/metabolismo , Glucuronidasa/metabolismo , Mieloma Múltiple/metabolismo , Animales , Bortezomib/farmacología , Línea Celular Tumoral , Quimioterapia , Exosomas/efectos de los fármacos , Exosomas/enzimología , Regulación Neoplásica de la Expresión Génica , Humanos , Melfalán/farmacología , Ratones , Mieloma Múltiple/tratamiento farmacológico , Oligopéptidos/farmacología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
9.
Blood Adv ; 1(25): 2375-2385, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-29296887

RESUMEN

The current treatment options for multiple myeloma (MM) osteolytic lesions are mainly combinations of chemotherapy and other small-molecule inhibitors, but toxic side effects still remain a major concern. Studies have shown that osteoclast activity is enhanced in MM patients through increased expression of receptor activator of nuclear factor κB ligand (RANKL), triggering RANK signaling on osteoclast precursors, which results in aggressive bone resorption. Furthermore, osteoprotegerin (OPG), a decoy receptor for RANKL, and the osteogenic potential of mesenchymal stem cells (MSCs) are significantly decreased in myeloma patients with multiple bone lesions. Thus, the use of OPG as a therapeutic molecule would greatly decrease osteolytic damage and reduce morbidity. However, in addition to inhibiting osteoclast activation, OPG binds to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), thereby rendering the tumor cells resistant to TRAIL-induced apoptosis and limiting the use of OPG for therapy. The present study developed a bone-disseminated myeloma disease model in mouse and successfully tested a cell therapy approach using MSCs, genetically engineered to express OPG variants that retain the capacity to bind RANKL, but do not bind TRAIL. Our results of skeletal remodeling following this regenerative stem cell therapy with OPG variants indicated a significant protection against myeloma-induced osteolytic bone damage in areas of major myeloma skeletal dissemination, suggesting the potential of this therapy for treating osteolytic damage in myeloma patients.

10.
Oncotarget ; 7(51): 85349-85364, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27863403

RESUMEN

Treatment of advanced colorectal cancer (CRC) requires multimodal therapeutic approaches and need for monitoring tumor plasticity. Liquid biopsy biomarkers, including CTCs and ctDNA, hold promise for evaluating treatment response in real-time and guiding therapeutic modifications. From 15 patients with advanced CRC undergoing liver metastasectomy with curative intent, we collected 41 blood samples at different time points before and after surgery for CTC isolation and quantification using label-free Vortex technology. For mutational profiling, KRAS, BRAF, and PIK3CA hotspot mutations were analyzed in CTCs and ctDNA from 23 samples, nine matched liver metastases and three primary tumor samples. Mutational patterns were compared. 80% of patient blood samples were positive for CTCs, using a healthy baseline value as threshold (0.4 CTCs/mL), and 81.4% of captured cells were EpCAM+ CTCs. At least one mutation was detected in 78% of our blood samples. Among 23 matched CTC and ctDNA samples, we found a concordance of 78.2% for KRAS, 73.9% for BRAF and 91.3% for PIK3CA mutations. In several cases, CTCs exhibited a mutation that was not detected in ctDNA, and vice versa. Complementary assessment of both CTCs and ctDNA appears advantageous to assess dynamic tumor profiles.


Asunto(s)
Biomarcadores de Tumor/genética , ADN Tumoral Circulante/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN/métodos , Mutación , Células Neoplásicas Circulantes/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Biomarcadores de Tumor/sangre , ADN Tumoral Circulante/sangre , Fosfatidilinositol 3-Quinasa Clase I/sangre , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Análisis Mutacional de ADN/instrumentación , Predisposición Genética a la Enfermedad , Células HCT116 , Humanos , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas , Células Neoplásicas Circulantes/patología , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Proteínas Proto-Oncogénicas B-raf/sangre , Proteínas Proto-Oncogénicas p21(ras)/sangre , Reproducibilidad de los Resultados , Factores de Tiempo
11.
Matrix Biol ; 55: 22-34, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27016342

RESUMEN

High heparanase expression is associated with enhanced tumor growth, angiogenesis, and metastasis in many types of cancer. However, the mechanisms driving high heparanase expression are not fully understood. In the present study, we discovered that drugs used in the treatment of myeloma upregulate heparanase expression. Frontline anti-myeloma drugs, bortezomib and carfilzomib activate the nuclear factor-kappa B (NF-κB) pathway to trigger heparanase expression in tumor cells. Blocking the NF-κB pathway diminished this chemotherapy-induced upregulation of heparanase expression. Activated NF-κB signaling was also found to drive high heparanase expression in drug resistant myeloma cell lines. In addition to enhancing heparanase expression, chemotherapy also caused release of heparanase by tumor cells into the conditioned medium. This soluble heparanase was taken up by macrophages and triggered an increase in TNF-α production. Heparanase is also taken up by tumor cells where it induced expression of HGF, VEGF and MMP-9 and activated ERK and Akt signaling pathways. These changes induced by heparanase are known to be associated with the promotion of an aggressive tumor phenotype. Importantly, the heparanase inhibitor Roneparstat diminished the uptake and the downstream effects of soluble heparanase. Together, these discoveries reveal a novel mechanism whereby chemotherapy upregulates heparanase, a known promoter of myeloma growth, and suggest that therapeutic targeting of heparanase during anti-cancer therapy may improve patient outcome.


Asunto(s)
Antineoplásicos/farmacología , Glucuronidasa/genética , Melanoma/enzimología , Bortezomib/farmacología , Línea Celular Tumoral , Doxorrubicina/farmacología , Inducción Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucuronidasa/metabolismo , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Melfalán/farmacología , Oligopéptidos/farmacología , Fenotipo
12.
Mol Oncol ; 10(3): 374-94, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26897752

RESUMEN

Circulating tumor cells, a component of the "liquid biopsy", hold great potential to transform the current landscape of cancer therapy. A key challenge to unlocking the clinical utility of CTCs lies in the ability to detect and isolate these rare cells using methods amenable to downstream characterization and other applications. In this review, we will provide an overview of current technologies used to detect and capture CTCs with brief insights into the workings of individual technologies. We focus on the strategies employed by different platforms and discuss the advantages of each. As our understanding of CTC biology matures, CTC technologies will need to evolve, and we discuss some of the present challenges facing the field in light of recent data encompassing epithelial-to-mesenchymal transition, tumor-initiating cells, and CTC clusters.


Asunto(s)
Separación Celular/métodos , Neoplasias/diagnóstico , Células Neoplásicas Circulantes/patología , Animales , Separación Celular/instrumentación , Centrifugación/instrumentación , Centrifugación/métodos , Electroforesis/instrumentación , Electroforesis/métodos , Filtración/instrumentación , Filtración/métodos , Humanos , Dispositivos Laboratorio en un Chip , Neoplasias/sangre , Neoplasias/patología
13.
Oncotarget ; 7(2): 1598-607, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26624982

RESUMEN

In most myeloma patients, even after several rounds of intensive therapy, drug resistant tumor cells survive and proliferate aggressively leading to relapse. In the present study, gene expression profiling of tumor cells isolated from myeloma patients after sequential rounds of chemotherapy, revealed for the first time that heparanase, a potent promoter of myeloma growth and progression, was elevated in myeloma cells that survived therapy. Based on this clinical data, we hypothesized that heparanase was involved in myeloma resistance to drug therapy. In several survival and viability assays, elevated heparanase expression promoted resistance of myeloma tumor cells to chemotherapy. Mechanistically, this enhanced survival was due to heparanase-mediated ERK signaling. Importantly, use of the heparanase inhibitor Roneparstat in combination with chemotherapy clearly diminished the growth of disseminated myeloma tumors in vivo. Moreover, use of Roneparstat either during or after chemotherapy diminished regrowth of myeloma tumors in vivo following therapy. These results provide compelling evidence that heparanase is a promising, novel target for overcoming myeloma resistance to therapy and that targeting heparanase has the potential to prevent relapse in myeloma and possibly other cancers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Glucuronidasa/antagonistas & inhibidores , Inhibidores de Glicósido Hidrolasas/farmacología , Mieloma Múltiple/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glucuronidasa/genética , Glucuronidasa/metabolismo , Humanos , Masculino , Ratones Endogámicos ICR , Ratones SCID , Terapia Molecular Dirigida , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Recurrencia , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nat Commun ; 6: 6985, 2015 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-25912399

RESUMEN

Herpesviruses exemplified by herpes simplex virus-1 (HSV-1) attach to cell surface heparan sulfate (HS) for entry into host cells. However, during a productive infection, the HS moieties on parent cells can trap newly exiting viral progenies and inhibit their release. Here we demonstrate that a HS-degrading enzyme of the host, heparanase (HPSE), is upregulated through NF-kB and translocated to the cell surface upon HSV-1 infection for the removal of HS to facilitate viral release. We also find a significant increase in HPSE release in vivo during infection of murine corneas and that knockdown of HPSE in vivo inhibits virus shedding. Overall, we propose that HPSE acts as a molecular switch for turning a virus-permissive 'attachment mode' of host cells to a virus-deterring 'detachment mode'. Since many human viruses use HS as an attachment receptor, the HPSE-HS interplay may delineate a common mechanism for virus release.


Asunto(s)
Glucuronidasa/metabolismo , Heparitina Sulfato/metabolismo , Herpes Simple/enzimología , Herpesvirus Humano 1/fisiología , Virión/fisiología , Animales , Chlorocebus aethiops , Femenino , Células HEK293 , Células HeLa , Herpes Simple/virología , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos BALB C , Regulación hacia Arriba , Células Vero , Liberación del Virus
15.
J Biol Chem ; 290(2): 941-9, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25404732

RESUMEN

The heparan sulfate proteoglycan syndecan-1 is proteolytically shed from the surface of multiple myeloma cells and is abundant in the bone marrow microenvironment where it promotes tumor growth, angiogenesis, and metastasis. In this study, we demonstrate for the first time that shed syndecan-1 present in the medium conditioned by tumor cells is taken up by bone marrow-derived stromal cells and transported to the nucleus. Translocation of shed syndecan-1 (sSDC1) to the nucleus was blocked by addition of exogenous heparin or heparan sulfate, pretreatment of conditioned medium with heparinase III, or growth of cells in sodium chlorate, indicating that sulfated heparan sulfate chains are required for nuclear translocation. Interestingly, cargo bound to sSDC1 heparan sulfate chains (i.e. hepatocyte growth factor) was transported to the nucleus along with sSDC1, and removal of heparan sulfate-bound cargo from sSDC1 abolished its translocation to the nucleus. Once in the nucleus, sSDC1 binds to the histone acetyltransferase enzyme p300, and histone acetyltransferase activity and histone acetylation are diminished. These findings reveal a novel function for shed syndecan-1 in mediating tumor-host cross-talk by shuttling growth factors to the nucleus and by altering histone acetylation in host cells. In addition, this work has broad implications beyond myeloma because shed syndecan-1 is present in high levels in many tumor types as well as in other disease states.


Asunto(s)
Carcinogénesis/genética , Histonas/metabolismo , Mieloma Múltiple/genética , Sindecano-1/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células del Estroma/metabolismo , Sindecano-1/biosíntesis , Microambiente Tumoral/genética , Factores de Transcripción p300-CBP/genética
16.
Matrix Biol ; 35: 215-22, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24145151

RESUMEN

In patients with multiple myeloma, the heparan sulfate proteoglycan syndecan-1 (CD138) is shed from the surface of tumor cells and accumulates in the serum and within the extracellular matrix of the bone marrow where it promotes tumor growth and metastasis. In the present study we discovered that commonly used anti-myeloma drugs stimulate syndecan-1 shedding both in vitro and in animals bearing myeloma tumors. Enhanced shedding is accompanied by increased syndecan-1 synthesis prior to drug induced tumor cell death. Addition of a caspase inhibitor blocks the drug-induced shedding of syndecan-1 in vitro indicating that shedding is linked to the onset of apoptosis. ADAM inhibitors or siRNA targeting ADAMs blocked drug-induced shedding suggesting that upregulation or activation of ADAMs is responsible for cleaving syndecan-1 from the tumor cell surface. These results reveal that myeloma chemotherapy stimulates synthesis and shedding of syndecan-1, a potentially negative side effect that may lead to the accumulation of high levels of syndecan-1 to establish a microenvironment that nurtures relapse and promotes tumor progression. Interestingly, we also found that chemotherapeutic drugs stimulated syndecan-1 shedding from pancreatic cancer cells as well, indicating that drug-induced shedding of syndecan-1 may occur in many cancer types. Overall, our results indicate that the use of metalloproteinase inhibitors (to inhibit syndecan-1 shedding) in combination with chemotherapy may represent a novel therapeutic strategy to prevent re-establishment of a microenvironment conducive for tumor relapse.


Asunto(s)
Antineoplásicos/efectos adversos , Médula Ósea/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Mieloma Múltiple/tratamiento farmacológico , Sindecano-1/metabolismo , Microambiente Tumoral/fisiología , Proteínas ADAM/metabolismo , Animales , Antineoplásicos/metabolismo , Western Blotting , Inhibidores de Caspasas/metabolismo , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/metabolismo , Ratones , Mieloma Múltiple/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Recurrencia , Sindecano-1/biosíntesis , Sindecano-1/sangre , Sales de Tetrazolio , Tiazoles
17.
FEBS J ; 280(10): 2294-306, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23374281

RESUMEN

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.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glucuronidasa/metabolismo , Neoplasias/tratamiento farmacológico , Sindecano-1/metabolismo , Animales , Antineoplásicos/farmacología , Materiales Biomiméticos/farmacología , Núcleo Celular/enzimología , Núcleo Celular/patología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Exosomas/metabolismo , Exosomas/patología , Glucuronidasa/antagonistas & inhibidores , Heparitina Sulfato/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Neoplasias/enzimología , Neoplasias/patología , Transducción de Señal , Sindecano-1/genética
18.
J Biol Chem ; 288(14): 10093-10099, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23430739

RESUMEN

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.


Asunto(s)
Exosomas/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucuronidasa/química , Línea Celular Tumoral , ADN Complementario/metabolismo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Heparitina Sulfato/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Mieloma Múltiple/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/enzimología , Sindecano-1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
J Biol Chem ; 287(13): 9952-9961, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22298773

RESUMEN

Matrix metalloproteinases release intact syndecan-1 ectodomains from the cell surface giving rise to a soluble, shed form of the proteoglycan. Although it is known that shed syndecan-1 controls diverse pathophysiological responses in cancer, wound healing, inflammation, infection, and immunity, the mechanisms regulating shedding remain unclear. We have discovered that the heparan sulfate chains present on syndecan core proteins suppress shedding of the proteoglycan. Syndecan shedding is dramatically enhanced when the heparan sulfate chains are enzymatically degraded or absent from the core protein. Exogenous heparan sulfate or heparin does not inhibit shedding, indicating that heparan sulfate must be attached to the core protein to suppress shedding. Regulation of shedding by heparan sulfate occurs in multiple cell types, for both syndecan-1 and syndecan-4 and in murine and human syndecans. Mechanistically, the loss of heparan sulfate enhances the susceptibility of the core protein to proteolytic cleavage by matrix metalloproteinases. Enhanced shedding of syndecan-1 following loss of heparan sulfate is accompanied by a dramatic increase in core protein synthesis. This suggests that in response to an increase in the rate of shedding, cells attempt to maintain a significant level of syndecan-1 on the cell surface. Together these data indicate that the amount of heparan sulfate present on syndecan core proteins regulates both the rate of syndecan shedding and core protein synthesis. These findings assign new functions to heparan sulfate chains, thereby broadening our understanding of their physiological importance and implying that therapeutic inhibition of heparan sulfate degradation could impact the progression of some diseases.


Asunto(s)
Heparitina Sulfato/metabolismo , Biosíntesis de Proteínas/fisiología , Sindecano-1/biosíntesis , Animales , Línea Celular , Femenino , Humanos , Ratones , Estructura Terciaria de Proteína , Sindecano-1/genética , Sindecano-4/biosíntesis , Sindecano-4/genética
20.
Biochim Biophys Acta ; 1813(8): 1525-31, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21616098

RESUMEN

The gelatinases, matrix metalloproteinase (MMP)-9 and -2, are produced as latent, inactive enzymes that can be proteolytically activated by a number of proteases. In many normal and pathological conditions, where the expression of MMPs is deregulated, changes in the expression of other proteases have also been reported. Human kallikrein-related peptidase 7 (KLK7), a chymotryptic-like serine protease, is overexpressed in many different types of neoplastic conditions, which have also been shown to express high levels of both MMP-9 and -2. Since the activation of MMPs by KLK7 has never been examined, we sought to determine whether KLK7 can activate these MMPs. To test this hypothesis KLK7 was incubated with the recombinant MMPs and the products of the reaction were analyzed for their activity. Incubation of proMMP-9 with KLK7 resulted in the production of a novel truncated, active MMP-9 lacking the C-terminal hemopexin domains. In contrast, KLK7 degraded, but did not activate, proMMP-2. The novel activation of proMMP-9 by KLK7 was further confirmed using conditioned medium prepared from an MMP-9-expressing cell line, MDA-MMP-9. Our results clearly establish that KLK7 activates proMMP-9 to produce a novel truncated, active MMP-9 product not generated by other proteases. These findings suggest that KLK7 may play an important role in the activation of MMP-9 in tumors that express high levels of both these proteases and the resulting truncated MMP may possess altered substrate specificities compared with full-length MMP-9 activated by other proteases.


Asunto(s)
Calicreínas/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Activación Enzimática , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Femenino , Gelatinasas/química , Gelatinasas/metabolismo , Hemopexina/química , Hemopexina/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Estructura Terciaria de Proteína , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA