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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 87(1): 67-79, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23055571

RESUMEN

Macrophages (MΦ) are first targets during human cytomegalovirus (HCMV) infection and are thought to be crucial for viral persistence and dissemination. However, since MΦ are also a first line of defense and key modulators of the immune response, these cells are at the crossroad between protection and viral pathogenesis. To date, the MΦ-specific contribution to the immune response against HCMV is still poorly understood. In view of the opposite roles of M1 and M2 MΦ during initiation and resolution of the immune response, we characterized the effects of HCMV infection on classically activated M1 MΦ and alternatively activated M2 MΦ. Although HCMV susceptibility was higher in M2 MΦ, HCMV established a productive and persistent infection in both types of MΦ. Upon HCMV encounter, both types of MΦ acquired similar features of classical activation and secreted high levels of proinflammatory cytokines and chemokines. As a functional consequence, conditioned media obtained from HCMV-infected M1 and M2 MΦ potently activated freshly isolated monocytes. Finally, compared to HCMV-infected monocyte-derived dendritic cells, infected M1 and M2 MΦ were more efficient in stimulating proliferation of autologous T cells from HCMV-seropositive donors at early times (24 h) postinfection, while the MΦ immunostimulatory properties were reduced, but not abrogated, at later times (72 h postinfection). In summary, our findings indicate that MΦ preserve proper antigen presentation capacity upon HCMV infection while enhancing inflammation, thus suggesting that MΦ play a role in the maintenance of the large HCMV-specific T-cell repertoire in seropositive individuals.


Asunto(s)
Proliferación Celular , Infecciones por Citomegalovirus/inmunología , Macrófagos/inmunología , Macrófagos/virología , Linfocitos T/inmunología , Células Cultivadas , Citocinas/metabolismo , Humanos , Inflamación , Linfocitos T/citología
2.
Carcinogenesis ; 33(1): 84-93, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22095073

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) and chronic pancreatitis, representing one risk factor for PDAC, are characterized by a marked desmoplasia enriched of pancreatic myofibroblasts (PMFs). Thus, PMFs are thought to essentially promote pancreatic tumorigenesis. We recently demonstrated that the adhesion molecule L1CAM is involved in epithelial-mesenchymal transition of PMF-cocultured H6c7 human ductal epithelial cells and that L1CAM is expressed already in ductal structures of chronic pancreatitis with even higher elevation in primary tumors and metastases of PDAC patients. This study aimed at investigating whether PMFs and L1CAM drive malignant transformation of pancreatic ductal epithelial cells by enhancing their tumorigenic potential. Cell culture experiments demonstrated that in the presence of PMFs, H6c7 cells exhibit a profound resistance against death ligand-induced apoptosis. This apoptosis protection was similarly observed in H6c7 cells stably overexpressing L1CAM. Intrapancreatic inoculation of H6c7 cells together with PMFs (H6c7co) resulted in tumor formation in 7/8 and liver metastases in 6/8 severe combined immunodeficiency (SCID) mice, whereas no tumors and metastases were detectable after inoculation of H6c7 cells alone. Likewise, tumor outgrowth and metastases resulted from inoculation of L1CAM-overexpressing H6c7 cells in 5/7 and 3/7 SCID mice, respectively, but not from inoculation of mock-transfected H6c7 cells. Treatment of H6c7co tumor-bearing mice with the L1CAM antibody L1-9.3/2a inhibited tumor formation and liver metastasis in 100 and 50%, respectively, of the treated animals. Overall, these data provide new insights into the mechanisms of how PMFs and L1CAM contribute to malignant transformation of pancreatic ductal epithelial cells in early stages of pancreatic tumorigenesis.


Asunto(s)
Carcinoma Ductal Pancreático/etiología , Miofibroblastos/fisiología , Molécula L1 de Adhesión de Célula Nerviosa/fisiología , Neoplasias Pancreáticas/etiología , Animales , Apoptosis , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Neoplasias Hepáticas/secundario , Ratones , Ratones SCID , Neoplasias Pancreáticas/patología
3.
Lung ; 190(3): 303-12, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22310878

RESUMEN

BACKGROUND: Pulmonary sarcoidosis has a variable course ranging from self-limiting disease to progressive fibrosis. Activation of fibroblasts, myofibroblast transformation, and matrix production may contribute to pulmonary damage in sarcoidosis. These processes are influenced by pulmonary cytokines which can be measured in bronchoalveolar lavage fluid (BALF). In order to clarify the incompletely understood fibrotic process in sarcoidosis, we classified activity of sarcoidosis according to WASOG criteria, measured TNF-α, IL-6, and HGF in BALF, and assessed the effect of HGF and BALF on proliferation and matrix production of human lung fibroblasts. RESULTS: BALF was obtained from 34 consecutive patients with sarcoidosis. BALF of active sarcoidosis contained elevated levels of TNF-α, HGF, and IL-6 and stimulated fibroblast proliferation. BALF of inactive sarcoidosis, but not of active sarcoidosis, stimulated the production of matrix proteins. HGF levels in inactive sarcoidosis were below those of control patients. HGF suppressed TGF-ß-induced matrix expression and transformation of fibroblasts into myofibroblasts. CONCLUSION: Prevention of TGF-ß-induced myofibroblast transformation may account for the inhibitory effect of HGF on matrix production. The strong fibrogenic effect of BALF of inactive sarcoidosis corresponds to the worse clinical course of inactive sarcoidosis compared with active disease and may be related to a lack of protective HGF.


Asunto(s)
Líquido del Lavado Bronquioalveolar/inmunología , Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Interleucina-6/metabolismo , Sarcoidosis Pulmonar/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Actinas/metabolismo , Adulto , Albúminas/análisis , Líquido del Lavado Bronquioalveolar/química , Proliferación Celular , Transdiferenciación Celular , Células Cultivadas , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Femenino , Fibronectinas/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis , Humanos , Pulmón , Masculino , Persona de Mediana Edad , Sarcoidosis Pulmonar/metabolismo , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas , Factor de Crecimiento Transformador beta
4.
Hepatogastroenterology ; 59(117): 1604-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22683979

RESUMEN

BACKGROUND/AIMS: Human pancreatic cancer is characterised by an extensive desmoplastic reaction. Activation of pancreatic stellate cells (PSCs) and their interactions with pancreatic cancer cells seem to be of essential importance in this process. Expression of fibroblast growth factor (FGF) receptor (FGFR) 1 splice variants may be of special interest in this communication as they are known to modulate the malignant phenotype of pancreatic cancer. The aim of the present study was to characterize interactions between PSCs and pancreatic cancer cells focusing on the Ig-domain III variants of fibroblast growth factor (FGF) receptor (FGFR) 1. METHODOLOGY: Expression of FGF ligands and FGFR1-III isoforms was determined by immunoblotting and specific RT-PCR analysis, respectively. RESULTS: PSCs and COLO-357, MIA PaCa-2, and PANC-1 pancreatic cancer cells expressed and secreted FGF2 and FGF5. Both FGFR1-III isoforms were coexpressed in PSCs and cancer cells. Conditioned medium of COLO-357 cells induced expression of both FGFR1- III isoforms in PSCs. In contrast, conditioned medium of PSCs induced FGFR1-IIIc, but reduced FGFR1- IIIb expression in the cancer cells. Neutralizing the effects of FGFs by heparin-sepharose precipitation abolished these effects completely. FGF2 and other growth factors secreted by PSCs resulted in upregulation of FGFR1-IIIc and downregulation of FGFR1-IIIb in pancreatic cancer cells. CONCLUSIONS: We identified in this study a mechanism based on tumor-stroma interactions involving PSCs that can contribute to enhance the malignant phenotype of human pancreatic cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Comunicación Celular , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 5 de Crecimiento de Fibroblastos/metabolismo , Humanos , Isoformas de Proteínas , ARN Mensajero/metabolismo , Regulación hacia Arriba
5.
Gut ; 60(2): 225-37, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20876774

RESUMEN

BACKGROUND: Evasion of apoptosis is a hallmark of pancreatic cancer. However, the underlying mechanisms are still only partly understood and may involve antiapoptotic proteins such as c-FLIP. Here, the role of c-FLIP in the regulation of death receptor-mediated apoptosis in pancreatic cancer was investigated. METHODS: Expression of c-FLIP(L) and c-FLIP(S) was analysed in primary pancreatic carcinoma samples, pancreatic carcinoma cell lines and primary tumour cells together with its function as a regulator of death receptor-induced apoptosis by knockdown and overexpression studies and through modulation by chemotherapeutics. RESULTS: c-FLIP is expressed in pancreatic intraepithelial neoplasm (PanIN) lesions and in pancreatic ductal adenocarcinomas, whereas normal pancreatic ducts were consistently negative for c-FLIP. Simultaneous downregulation of c-FLIP(L) and c-FLIP(S) as well as individual knockdown of either isoform by RNA interference significantly enhances TRAIL (tumour necrosis factor-related apoptosis-inducing ligand)- and CD95-induced caspase activation and caspase-dependent apoptosis. Also, pretreatment with chemotherapeutic drugs--that is, 5-fluorouracil (5-FU), cisplatin or gemcitabine--downregulates c-FLIP and renders cells sensitive to death receptor-triggered apoptosis. Similarly, primary cultured pancreatic cancer cells are primed for TRAIL-induced apoptosis by pre-exposure to 5-FU or cisplatin. Mechanistic studies revealed that 5-FU-mediated suppression of c-FLIP results in increased TRAIL-induced recruitment and activation of caspase-8 at the death-inducing signalling complex (DISC), leading to caspase-3 activation and caspase-dependent cell death. Overexpression of c-FLIP(L) rescues cells from 5-FU- or cisplatin-mediated sensitisation for TRAIL-induced apoptosis, indicating that c-FLIP suppression is a key event in this chemotherapy-mediated sensitisation to TRAIL. Further, concomitant neutralisation of c-FLIP and XIAP acts in concert to potentiate TRAIL-induced apoptosis. CONCLUSIONS: Both the long and the short isoform of the antiapoptotic protein c-FLIP are critical regulators of death receptor-induced apoptosis in pancreatic carcinoma cells and are suppressed by chemotherapeutics. Targeting either c-FLIP(L) or c-FLIP(S) is sufficient to promote death receptor-induced apoptosis in pancreatic carcinoma cells. These findings have important implications for the design of TRAIL-based combination protocols in pancreatic cancer.


Asunto(s)
Apoptosis/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/fisiología , Proteínas de Neoplasias/fisiología , Neoplasias Pancreáticas/patología , Receptores de Muerte Celular/fisiología , Antimetabolitos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Cisplatino/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Fluorouracilo/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Células Tumorales Cultivadas , Receptor fas/fisiología
6.
Eur J Clin Invest ; 41(2): 151-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20860757

RESUMEN

BACKGROUND: Pancreatic stellate cells (PSC) play a central role in fibrogenesis associated with acute and chronic pancreatitis. Pancreatic stone protein/regenerating protein (PSP/reg) belongs to a family of secretory stress proteins (SSP) that are constitutively synthesized by pancreatic acinar cells and upregulated dramatically during acute and chronic pancreatitis. Assuming a protective role of this stress protein, we investigated its effects on human PSC. MATERIAL AND METHODS: Pancreatic stellate cells were obtained by outgrowth from fibrotic human pancreas tissue. PSP/reg was expressed in the yeast Pichia pastoris and purified from medium supernatants. PSP/reg was added at concentrations of 100 ng/mL to cultured PSC. Cell proliferation was determined by bromodeoxyuridine incorporation. PSC migration was assessed by a wound healing assay. Extracellular matrix (collagen type I and fibronectin), matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) were demonstrated on protein level. RESULTS: Pancreatic stone protein/regenerating protein inhibited PSC proliferation and migration. Soluble collagen I and fibronectin were reduced after the addition of PSP/reg. PSP/reg slightly decreased the synthesis of MMP-1 and MMP-2 and strongly decreased TIMP-1 and TIMP-2 concentrations in PSC supernatants. CONCLUSIONS: Our work describes a novel aspect that in vitro PSP/reg reduces PSC activity (proliferation and migration) and stimulates fibrolysis by increasing MMP/TIMP ratio. The findings suggest that PSP/reg might have a protective function in the repair phase of acute and chronic pancreatitis by promoting resolution of fibrosis. We highlight PSP/reg as an antifibrogenic protein in pancreatic injury.


Asunto(s)
Litostatina/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Pancreatitis/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Anciano , Análisis de Varianza , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1163-71, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19779015

RESUMEN

Mechanisms leading to acute pancreatitis after a fat-enriched meal combined with excess alcohol are incompletely understood. We have studied the effects of alcohol and fat (VLDL) on pancreatic acinar cell (PAC) function, oxidative stress, and repair mechanisms by pancreatic stellate cells (PSC) leading to fibrogenesis. To do so, PAC (rat) were isolated and cultured up to 24 h. Ethanol and/or VLDL were added to PAC. We measured PAC function (amylase, lipase), injury (lactic dehydrogenase), apoptosis (TUNEL, Apo2.7, annexin V binding), oxidative stress, and lipid peroxidation (conjugated dienes, malondialdehyde, chemoluminescence); we also measured PSC proliferation (bromodeoxyuridine incorporation), matrix synthesis (immunofluorescence of collagens and fibronectin, fibronectin immunoassay), and fatty acids in PAC supernatants (gas chromatography). Within 6 h, cultured PAC degraded and hydrolyzed VLDL completely. VLDL alone (50 microg/ml) and in combination with alcohol (0.2, 0.5, and 1% vol/vol) induced PAC injury (LDL, amylase, and lipase release) within 2 h through generation of oxidative stress. Depending on the dose of VLDL and alcohol, apoptosis and/or necrosis were induced. Antioxidants (Trolox, Probucol) reduced the cytotoxic effect of alcohol and VLDL. Supernatants of alcohol/VLDL-treated PAC stimulated stellate cell proliferation and extracellular matrix synthesis. We concluded that, in the presence of lipoproteins, alcohol induces acinar cell injury. Our results provide a biochemical pathway for the clinical observation that a fat-enriched meal combined with excess alcohol consumption can induce acinar cell injury (acute pancreatitis) followed by repair mechanisms (proliferation and increased matrix synthesis in PSC).


Asunto(s)
Proliferación Celular/efectos de los fármacos , Etanol/toxicidad , Matriz Extracelular/metabolismo , Lipoproteínas VLDL/toxicidad , Páncreas Exocrino/efectos de los fármacos , Pancreatitis/etiología , Enfermedad Aguda , Amilasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Fibrosis , L-Lactato Deshidrogenasa/metabolismo , Lipasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Lipoproteínas VLDL/metabolismo , Estrés Oxidativo/efectos de los fármacos , Páncreas Exocrino/metabolismo , Páncreas Exocrino/patología , Pancreatitis/metabolismo , Pancreatitis/patología , Ratas , Ratas Wistar , Factores de Tiempo
8.
Thromb Haemost ; 101(4): 682-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19350111

RESUMEN

The Extracellular Matrix Metalloproteinase Inducer (EMMPRIN, CD147, basigin) is an immunoglobulin-like receptor expressed in various cell types. During cellular interactions homotypic EMMPRIN-EMMPRIN interactions are known to induce the synthesis of matrix metalloproteinases. Recently, we have identified EMMPRIN as a novel receptor on platelets. To our knowledge EMMPRIN has not been shown to serve as adhesion receptor, yet. Here we characterise platelet glycoprotein VI (GPVI) as a novel adhesion receptor for EMMPRIN. Human platelets were prestimulated with ADP and perfused over immobilised recombinant EMMPRIN-Fc or Fc-fragments under arterial shear conditions. ADP-stimulated platelets showed significantly enhanced rolling (but not enhanced firm adhesion) on immobilised EMMPRIN-Fc compared to Fc. Pretreatment of platelets with blocking mAbs anti-EMMPRIN or anti-GPVI leads to a significant reduction of rolling platelets on immobilised EMMPRIN-Fc, whereas pretreatment with blocking mAbs anti-p-selectin, anti-alpha4-integrin or anti-GPIIb/IIIa complex (20 microg/ml each) had no effect. Consistently, chinese hamster ovary (CHO) cells stably transfected with GPVI showed enhanced rolling (but not adhesion) on immobilised EMMPRIN-Fc in comparison to non-transfected CHO cells. Similarly, CHO cells stably transfected with EMMPRIN showed enhanced rolling on immobilised GPVI-Fc (or EMMPRIN-Fc) compared to non transfected CHO-cells. Finally, specific binding of EMMPRIN to GPVI was demonstrated by a modified ELISA and surface plasmon resonance technology with a dissociation constant of 88 nM. Platelet GPVI is a novel receptor for EMMPRIN and can mediate platelet rolling via GPVI-EMMPRIN interaction.


Asunto(s)
Basigina/metabolismo , Plaquetas/metabolismo , Antígenos CD36/metabolismo , Movimiento Celular , Activación Plaquetaria , Adenosina Difosfato/metabolismo , Animales , Anticuerpos Monoclonales , Basigina/genética , Basigina/inmunología , Antígenos CD36/genética , Antígenos CD36/inmunología , Células CHO , Adhesión Celular , Cricetinae , Cricetulus , Humanos , Ratones , Selectina-P/metabolismo , Adhesividad Plaquetaria , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Transfección
9.
Pancreatology ; 9(1-2): 99-110, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19077460

RESUMEN

BACKGROUND/AIMS: Pancreatic stellate cells (PSCs) play a key role in fibrogenesis associated with acute and chronic pancreatitis. Pancreatitis-associated protein (PAP), an acute-phase protein, is dramatically upregulated during acute and chronic pancreatitis. Assuming a protective role of PAP, we investigated its effects on human PSCs. METHODS: PSCs were obtained by outgrowth from fibrotic human pancreas tissue. PAP was expressed in the yeast Pichia pastoris. PAP was added at 10 ng/ml to cultured PSCs. Cell proliferation was determined by bromodeoxyuridine incorporation. PSC migration was assessed by a wound healing assay. Collagen types I and III, fibronectin, matrix metalloproteinases (MMPs), tissue inhibitors of MMPs (TIMPs) and reversion-inducing cysteine-rich protein with Kazal motifs (RECK) were demonstrated on protein and mRNA level. RESULTS: PAP had no significant effect on PSC proliferation and migration. Cell-associated fibrillar collagen types I and III and fibronectin increased after addition of PAP to PSCs. PAP diminished the expression of MMP-1 and -2 and TIMP-1 and -2 and their concentrations in PSC supernatants. RECK was detected on the surface of PSCs and its expression was reduced after PAP application. CONCLUSIONS: Our data offer new insights into the biological functions of PAP, which may play an important role in wound healing response and cell-matrix interactions.


Asunto(s)
Antígenos de Neoplasias/fisiología , Biomarcadores de Tumor/fisiología , Lectinas Tipo C/fisiología , Inhibidores de la Metaloproteinasa de la Matriz , Glicoproteínas de Membrana/biosíntesis , Páncreas/efectos de los fármacos , Inhibidor Tisular de Metaloproteinasa-1/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-2/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas Ligadas a GPI , Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Páncreas/citología , Proteínas Asociadas a Pancreatitis , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
10.
Cell Signal ; 20(4): 695-704, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18207700

RESUMEN

Phagocytosis is an essential event in the complex process of tissue repair. Here we examined the effect of low intensity pulsed ultrasound (US), which promotes fracture and wound healing, on phagocytosis by mouse macrophage cell line J774A.1 and human monocyte-derived macrophages. First, 10 to 40 min low intensity pulsed US increased uptake of serum opsonized E. coli by J774A.1 cells during a 50 min phagocytosis period. In addition, when the E. coli exposure time was varied between 35 to 80 min, the maximum increase in phagocytosis was observed in the first 35 min upon US exposure. In parallel, US induced robust actin polymerization in a time dependent manner in J774A.1 cells, showing the peak effect 30 min after stimulation. Interestingly, a low concentration of cytochalasin D (0.25-0.5 microM) prevented US-induced phagocytosis of E. coli. Furthermore, we demonstrated US enhanced activation of RhoA. Blocking its downstream effector Rho associated kinase (ROCK) with Y27632 abrogated US-induced phagocytosis. We also show that US induced activation of ERK and p38 MAPK. Pretreatment of the cells with the corresponding inhibitors PD98059 and SB203580 reduced US-induced phagocytosis. In addition, activity of tyrosine kinase Src was required for US-induced phagocytosis. Here Src represents an upstream activator of ERK and p38 MAPK. Depolymerization of actin by cytochalasin D prevented US-induced Src, ERK, and p38 activation. Our data provide a new insight into the cellular and molecular mechanisms by which low intensity pulsed US promotes tissue repair.


Asunto(s)
Actinas/metabolismo , Macrófagos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fagocitosis , Transducción de Señal , Ultrasonido , Proteína de Unión al GTP rhoA/metabolismo , Familia-src Quinasas/metabolismo , Amidas/farmacología , Animales , Línea Celular , Células Cultivadas , Activación de Complemento , Citocalasina D/farmacología , Escherichia coli/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Imidazoles/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/inmunología , Ratones , Fagocitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
11.
Cancer Res ; 67(6): 2712-9, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363592

RESUMEN

Fibroblast growth factor receptors (FGFR) play important roles in many biological processes. Nothing is presently known about possible roles of the human FGFR1-IIIb mRNA splice variant. In this study, we characterized for the first time the effects of FGFR1-IIIb expression on the transformed phenotype of human pancreatic cancer cells. The full-length FGFR1-IIIb cDNA was generated and stably expressed in PANC-1 and MIA PaCa-2 pancreatic cancer and TAKA-1 pancreatic ductal cells. FGFR1-IIIb-expressing cells synthesized a glycosylated 110-kDa protein enhancing tyrosine phosphorylation of FGFR substrate-2 on FGF-1 stimulation. The basal anchorage-dependent and anchorage-independent cell growth was significantly inhibited. These effects were associated with a marked reduction of p44/42 mitogen-activated protein kinase (MAPK) phosphorylation in combination with enhanced activity of p38 MAPK and c-Jun NH(2)-terminal kinase. FGFR1-IIIb expression inhibited single-cell movement and in vitro invasion as determined by time-lapse microscopy and Boyden chamber assay as well as in vivo tumor formation and growth in nude mice. Microscopic analysis of the xenograft tumors revealed a reduced Ki-67 labeling and a lower amount of tumor necrosis in FGFR1-IIIb-expressing tumors. Our results show that FGFR1-IIIb is a functional FGFR that inhibits the transformed phenotype of human pancreatic cancer cells.


Asunto(s)
Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Pancreáticas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis
12.
Langenbecks Arch Surg ; 393(6): 891-900, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18204855

RESUMEN

BACKGROUND: Adenocarcinomas of the pancreas are characterized by a rapid progression, an early metastasis, a limited response to chemo- and radiotherapy, and an intense fibrotic reaction known as tumor desmoplasia. Carcinoma cells are surrounded by a dense stroma consisting of myofibroblast-like cells, collagens, and fibronectin. MATERIALS AND METHODS: This review describes the interaction of activated pancreatic stellate cells (myofibroblast-like cells) with tumor cells in pancreas adenocarcinomas. Our data were obtained in cell culture experiments and in in vivo investigations. RESULTS: Carcinoma cells produce soluble mediators and stimulate motility, proliferation, matrix-, and MMP synthesis of stellate cells. Vice versa-activated stellate cells release mitogens, stimulating proliferation of cancer cells. Cancer cell proliferation and resistance to apoptosis might further be induced by the microenvironment (extracellular matrix), which is primarily provided by stellate cells. A very important aspect in the interaction of stellate cells with cancer cells is the expression of EMMPRIN (extracellular matrix metalloproteinase inducer) by cancer cells, the shedding of the extracellular part of EMMPRIN by matrix metalloproteinases (MMPs), and the induction of MMPs in stellate cells by soluble EMMPRIN. In particular, the stellate cells in close proximity to tumor cells therefore express MMPs and degrade connective tissue. CONCLUSION: Through complex interactions between stellate cells and carcinoma cells, tumor progression and cancer cell invasion are accelerated. As we gain better understanding of these mechanisms, adequate therapies to reduce tumor cell invasion and cancer progression might be developed.


Asunto(s)
Adenocarcinoma/fisiopatología , Transformación Celular Neoplásica/patología , Matriz Extracelular/fisiología , Páncreas/fisiopatología , Neoplasias Pancreáticas/fisiopatología , Adenocarcinoma/patología , Animales , Basigina/metabolismo , Comunicación Celular/fisiología , División Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Colágeno/metabolismo , Matriz Extracelular/patología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Páncreas/patología , Neoplasias Pancreáticas/patología , Lesiones Precancerosas/patología , Lesiones Precancerosas/fisiopatología
14.
Pancreatology ; 7(4): 380-5, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17703087

RESUMEN

We have previously reported that inducible overexpression of the serine protease inhibitor nexin 2 (SERPINE2) significantly increases local invasiveness of subclones of the pancreatic cancer cell-line SUIT-2 in nude mouse xenografts. This was associated with a striking increase of extracellular matrix deposition in the invasive tumors. Pancreatic stellate cells (PSCs) have recently been identified as the major source of fibrosis in pancreatic adenocarcinomas. Here we report that co-injection of PSCs and tumor cells dramatically enhances the invasive potential of serine protease inhibitor Nexin 2 (SERPINE2)-expressing SUIT-2 cells. 100% (24 of 24) of the SERPINE2-expressing tumors with PSCs grew aggressively invasive, as compared to 39% of SERPINE2-negative tumors with PSCs and 27% of SERPINE2-expressing tumors without PSCs. In contrast to pure cancer cell preparations, SERPINE2 overexpression in the presence of PSCs also resulted in increased tumor growth. Histological evaluation demonstrated the presence of large amounts of ECM deposits co-localizing with cells staining positive for the PSC marker alpha-SMA. We conclude that PSCs actively proliferate in pancreatic cancer xenograft tumors and significantly contribute to the local invasive potential of the tumors. Presence of PSCs enhances the pro-invasive effects of SERPINE2 expression, and SERPINE2 influences tumor growth (as opposed to invasiveness) only in the presence of PSCs. Our data thus suggest that SERPINE2 is an important modulator of tumor cell/host interactions in pancreatic cancer.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Páncreas/citología , Receptores de Superficie Celular/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neoplasias Experimentales , Neoplasias Pancreáticas , Nexinas de Proteasas , Trasplante Heterólogo
15.
Arterioscler Thromb Vasc Biol ; 26(4): 845-50, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16410460

RESUMEN

BACKGROUND: Therapeutic strategies to stabilize advanced arteriosclerotic lesions may prevent plaque rupture and reduce the incidence of acute coronary syndromes. Thiazolidinediones (TZDs), like rosiglitazone, are oral antidiabetic drugs with additional antiinflammatory and potential antiatherogenic properties. In a randomized, placebo-controlled, single-blind trial, we examined the effect of 4 weeks of rosiglitazone therapy on histomorphological characteristics of plaque stability in artery specimen of nondiabetic patients scheduled for elective carotid endarterectomy. METHODS AND RESULTS: A total of 24 nondiabetic patients with symptomatic carotid artery stenosis were randomly assigned to rosiglitazone (4 mg BID) or placebo in addition to standard therapy. In this population of nondiabetic patients, rosiglitazone treatment did not significantly change fasting blood glucose, fasting insulin, or lipid parameters. In contrast, rosiglitazone significantly reduced CD4-lymphocyte content as well as macrophage HLA-DR expression in the shoulder region, reflecting less inflammatory activation of these cells by lymphocyte interferon-gamma. Moreover, rosiglitazone significantly increased plaque collagen content (7.7+/-1.6% versus 3.7+/-0.7% of plaque area; P=0.036) compared with placebo, suggesting that TZD treatment may stabilize arteriosclerotic lesions. In addition, rosiglitazone reduced serum levels of 2 inflammatory arteriosclerosis markers: C-reactive protein and serum amyloid A. CONCLUSIONS: Four weeks of treatment with rosiglitazone significantly reduces vascular inflammation in nondiabetic patients, leading to a more stable type of arteriosclerotic lesion.


Asunto(s)
Estenosis Carotídea/tratamiento farmacológico , Colágeno/metabolismo , Hipoglucemiantes/administración & dosificación , Tiazolidinedionas/administración & dosificación , Anciano , Glucemia/efectos de los fármacos , Índice de Masa Corporal , Proteína C-Reactiva/metabolismo , Estenosis Carotídea/metabolismo , Estenosis Carotídea/fisiopatología , Colesterol/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Diabetes Mellitus , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Inflamación/tratamiento farmacológico , Insulina/sangre , Masculino , Persona de Mediana Edad , Rosiglitazona , Proteína Amiloide A Sérica/metabolismo , Tiazolidinedionas/uso terapéutico
16.
Anticancer Res ; 27(3B): 1415-21, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17595756

RESUMEN

BACKGROUND: Human blood outgrowth endothelial cells (BOECs), used as cellular vehicles for experimental cancer therapy, infiltrate tumors. Infiltration may determine their therapeutic efficacy. The role of matrix metalloproteinases (MMPs) in BOEC infiltration and its potential enhancement by irradiation were investigated. MATERIALS AND METHODS: Infiltration of BOECs into spheroids made of tumor cells and fibroblasts was determined in the presence of low-dose EDTA, a potent inhibitor of MMPs. Expression and secretion of MMP-2 and -9 by these tumor cells and tumor-stromal cells were investigated with zymography and immunohistochemistry. RESULTS: Infiltration of BOECs into spheroids was blocked by low-dose EDTA. Irradiation enhanced secretion of MMP-2 and, less so, of MMP-9 by tumor-stromal cells and tumor cells, and increased the amount of MMP-2 and -9 in subcutaneous LLC tumors. However, irradiation did not increase infiltration by BOECs. CONCLUSION: MMPs are involved in the infiltration of BOECs into spheroids. Infiltration of BOECs is not increased by irradiation at the doses employed.


Asunto(s)
Células Sanguíneas/fisiología , Movimiento Celular , Células Endoteliales/fisiología , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/fisiología , Esferoides Celulares/fisiología , Animales , Células Sanguíneas/enzimología , Células Sanguíneas/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Ácido Edético/farmacología , Células Endoteliales/enzimología , Células Endoteliales/efectos de la radiación , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Comunicación Paracrina , Radiación , Células Tumorales Cultivadas , Regulación hacia Arriba
17.
J Mol Med (Berl) ; 83(10): 795-805, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15976918

RESUMEN

Pancreatic stellate cells (PSCs) are thought to be the primary source of the extensive fibrotic reaction characteristic of pancreatic cancer and chronic pancreatitis in humans. PSCs share many morphological and functional characteristics with hepatic stellate cells (HSCs), whose central role in liver fibrosis is well established. However, it has remained unclear if hepatic and pancreatic stellate cells are derived from a common cell lineage and if they are completely similar or if they possess organ-specific features. We have analysed the transcriptomes of HSCs, PSCs and skin fibroblasts to assess how the transcriptional phenotype of stellate cells differs from that of a typical fibroblast lineage cell and if there is evidence for a common stellate cell precursor. To this end, we have performed expression profiling of primary cultures of human HSCs, PSCs and skin fibroblasts using 23,000-feature 'whole genome' oligonucleotide micro-arrays. Expression data were verified using real-time PCR. The expression profiles of HSCs and PSCs displayed a great extent of similarity, clearly separating them from the fibroblasts. Predominantly extracellular and cell surface genes, but also signalling molecules, transcription factors and novel neural markers, were concordantly expressed in both stellate cell types. Despite this high degree of similarity, distinct differences in expression patterns were observed between HSCs and PSCs, reflecting organ-specific variations of the common stellate cell-specific phenotype.


Asunto(s)
Hígado/citología , Páncreas/citología , Transcripción Genética , Linaje de la Célula , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Hígado/metabolismo , Especificidad de Órganos , Páncreas/metabolismo , Fenotipo , Piel/citología , Piel/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 24(10): 1823-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15319264

RESUMEN

OBJECTIVE: Matrix metalloproteinases (MMPs) seem to play a prominent role in atherogenesis. Extracellular MMP inducer (EMMPRIN), a cell surface glycoprotein which stimulates MMP synthesis, has recently been detected in human atheroma. We have investigated the influence of oxidized low-density lipoproteins (oxLDLs) on EMMPRIN expression in human coronary artery smooth muscle cells (HCA-SMCs). METHODS AND RESULTS: OxLDL induced a significant increase of EMMPRIN release into HCA-SMC supernatants and a concomitant decrease of cell-associated EMMPRIN. These effects were antagonized by antioxidants as well as by EDTA and the MMP inhibitor GM6001. Western blot analysis demonstrated that MMP-1 and MMP-2 induce the cleavage of the extracellular domain from cell-associated EMMPRIN. MMP-1 and MMP-2 synthesis was upregulated by oxLDL, and, in addition, we have shown that soluble EMMPRIN, isolated from macrophage supernatants, increased MMP-1 and MMP-2 synthesis in HCA-SMC. CONCLUSIONS: Our data suggest that oxLDLs stimulate the release of soluble EMMPRIN, at least in part, by MMP-dependent shedding from the cell surface. Additionally, oxLDLs might induce a circular upregulation of matrix degradation because, in turn, soluble EMMPRIN stimulates MMP synthesis in HCA-SMC.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Lipoproteínas LDL/farmacología , Músculo Liso Vascular/efectos de los fármacos , Antígenos CD , Basigina , Vasos Coronarios/citología , Vasos Coronarios/enzimología , Humanos , Macrófagos/química , Metaloproteinasas de la Matriz/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Proteínas Recombinantes/farmacología
19.
Clin Cancer Res ; 10(14): 4776-83, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15269152

RESUMEN

PURPOSE: The aim of this study was to investigate the expression and significance of decorin in pancreatic cancer. EXPERIMENTAL DESIGN: Decorin expression in normal pancreas and excised tumors was examined by real-time quantitative PCR, Western blot analysis, and immunohistochemistry. Reverse transcription-PCR was used to analyze cultures of pancreatic cancer and stellate cells. Growth-inhibitory effects of decorin in vitro were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test, Western blot, and fluorescence-activated cell-sorting analysis. RESULTS: Pancreatic cancer was characterized by striking overexpression of decorin mRNA in tumor tissues (9-fold by real-time quantitative PCR; 44 patients versus 18 healthy donors; P < 0.01). Strong decorin immunostaining was observed in the extracellular matrix of pancreatic cancer tissue, whereas tumor cells were devoid of decorin. Double staining for anti-smooth muscle actin and decorin and reverse transcription-PCR analysis of primary cultures revealed pancreatic stellate cells as the putative source of decorin. Human recombinant decorin was able to suppress growth of pancreatic cancer cells in vitro through p21 mediated G(1)-S block of the cell cycle. However, in contrast to the previously described chemotherapy-potentiating capacity of decorin, this proteoglycan attenuated the cytostatic action of carboplatin and gemcitabine toward pancreatic cancer cells. CONCLUSIONS: Decorin might exert an antiproliferative effect toward pancreatic cancer cells, thus playing a role in a host stromal reaction aimed at sequestering and inhibiting growing malignant cells. However, in clinical settings, the importance of collagen-associated decorin as a moderate antitumor modality would be undermined by its ability to attenuate the efficiency of chemotherapeutics. Considering the general failure of adjuvant therapies in pancreatic cancer, the role of decorin in this process warrants further investigation.


Asunto(s)
Desoxicitidina/análogos & derivados , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/patología , Proteoglicanos/genética , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/farmacología , Carboplatino/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Decorina , Desoxicitidina/farmacología , Interacciones Farmacológicas , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Páncreas/química , Páncreas/citología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/genética , Gemcitabina
20.
Int J Cardiol ; 103(3): 298-306, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16098393

RESUMEN

Hepatocyte growth factor (HGF) is a potent regeneration factor for endothelial and epithelial cells, and has also been shown to modulate extracellular matrix synthesis and matrix metalloproteinase activity in renal epithelial cells and tumor cells. Controversial results have been published concerning the possible role of HGF in the pathogenesis of coronary atherosclerosis. In this study, we have investigated the effect of oxidized low density lipoproteins (LDL) and elevated glucose concentrations on HGF synthesis in cultured human coronary artery smooth muscle cells. In addition, we have studied whether HGF modulates the release of extracellular matrix, extracellular matrix metalloproteinase inducer (EMMPRIN) and matrix metalloproteinases (MMP) by coronary artery smooth muscle cells. Oxidized LDL (1-10 microg/ml) induced a significant dose-dependent decrease of HGF release and a concomitant decrease of HGF mRNA expression, whereas native LDL and elevated glucose concentrations induced no significant changes of HGF synthesis. Incubation of cultured human coronary smooth muscle cells with human HGF (1-100 ng/ml) did not significantly alter cell migration and collagen I, fibronectin, EMMPRIN, MMP-1, MMP-2 and MMP-9 release. In summary, our results provide evidence that HGF does not promote coronary plaque growth or plaque destabilization. Regarding the fact that HGF is a potent endothelial cell regeneration factor, the observed downregulation of HGF synthesis by oxidized LDL supports the concept that HGF might be a protective factor in coronary atherosclerosis and that a decrease rather than an increase of HGF synthesis might promote coronary atherosclerosis.


Asunto(s)
LDL-Colesterol/farmacología , Vasos Coronarios/citología , Factor de Crecimiento de Hepatocito/biosíntesis , Músculo Liso Vascular/metabolismo , Estrés Oxidativo/fisiología , Basigina/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/fisiología , Humanos , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA