Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Matrix Biol ; 81: 50-69, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30412725

RESUMO

It is predicted that pancreatic ductal adenocarcinoma (PDAC) will become the second most lethal cancer in the US by 2030. PDAC includes a fibrous-like stroma, desmoplasia, encompassing most of the tumor mass, which is produced by cancer-associated fibroblasts (CAFs) and includes their cell-derived extracellular matrices (CDMs). Since elimination of desmoplasia has proven detrimental to patients, CDM reprogramming, as opposed to stromal ablation, is therapeutically desirable. Hence, efforts are being made to harness desmoplasia's anti-tumor functions. We conducted biomechanical manipulations, using variations of pathological and physiological substrates in vitro, to culture patient-harvested CAFs and generate CDMs that restrict PDAC growth and spread. We posited that extrinsic modulation of the environment, via substrate rigidity, influences CAF's cell-intrinsic forces affecting CDM production. Substrates used were polyacrylamide gels of physiological (~1.5 kPa) or pathological (~7 kPa) stiffnesses. Results showed that physiological substrates influenced CAFs to generate CDMs similar to normal/control fibroblasts. We found CDMs to be softer than the corresponding underlying substrates, and CDM fiber anisotropy (i.e., alignment) to be biphasic and informed via substrate-imparted morphological CAF aspect ratios. The biphasic nature of CDM fiber anisotropy was mathematically modeled and proposed a correlation between CAF aspect ratios and CDM alignment; regulated by extrinsic and intrinsic forces to conserve minimal free energy. Biomechanical manipulation of CDMs, generated on physiologically soft substrates, leads to reduction in nuclear translocation of pERK1/2 in KRAS mutated pancreatic cells. ERK2 was found essential for CDM-regulated tumor cell spread. In vitro findings correlated with in vivo observations; nuclear pERK1/2 is significantly high in human PDAC samples. The study suggests that altering underlying substrates enable CAFs to remodel CDMs and restrict pancreatic cancer cell spread in an ERK2 dependent manner.


Assuntos
Fibroblastos Associados a Câncer/citologia , Carcinoma Ductal Pancreático/metabolismo , Matriz Extracelular/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Anisotropia , Fenômenos Biomecânicos , Fibroblastos Associados a Câncer/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Proliferação de Células , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Células NIH 3T3 , Microambiente Tumoral
2.
Oncogene ; 33(5): 589-98, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23318431

RESUMO

ErbB2 is frequently highly expressed in premalignant breast cancers, including ductal carcinoma in situ (DCIS); however, little is known about the signals or pathways it contributes to progression into the invasive/malignant state. Radiotherapy is often used to treat early premalignant lesions regardless of ErbB2 status. Here, we show that clinically relevant doses of ionizing radiation (IR)-induce cellular invasion of ErbB2-expressing breast cancer cells, as well as MCF10A cells overexpressing ErbB2. ErbB2-negative breast cancer cells, such as MCF7 and T47D, do not invade following treatment with IR nor do MCF10A cells overexpressing epidermal growth factor receptor. ErbB2 becomes phosphorylated at tyrosine 877 in a dose- and time- dependent manner following exposure to X-rays, and activates downstream signaling cascades including PI3K/Akt. Inhibition of these pathways, as well as inhibition of reactive oxygen species (ROS) with antioxidants, prevents IR-induced invasion. Activation of ErbB2-dependent signaling results in upregulation of the forkhead family transcription factor, FoxM1, and its transcriptional targets, including matrix metalloproteinase 2 (MMP2). Inhibition of FoxM1 by RNA interference prevented induction of invasion by IR, and overexpression of FoxM1 in MCF10A cells was sufficient to promote IR-induced invasion. Moreover, we found that 14-3-3ζ is also upregulated by IR in cancer cells in a ROS-dependent manner, is required for IR-induced invasion in ErbB2-positive breast cancer cells and together with FoxM1 is sufficient for invasion in ErbB2-negative breast cancer cells. Thus, our data show that IR-mediated activation of ErbB2 and induction of 14-3-3ζ collaborate to regulate FoxM1 and promote invasion of breast cancer cells and furthermore, may serve as therapeutic targets to enhance radiosensitivity of breast cancers.


Assuntos
Proteínas 14-3-3/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/radioterapia , Fatores de Transcrição Forkhead/metabolismo , Receptor ErbB-2/metabolismo , Proteínas 14-3-3/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Humanos , Células MCF-7 , Metaloproteinase 2 da Matriz/biossíntese , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Radiação Ionizante , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos da radiação
3.
Artigo em Inglês | MEDLINE | ID: mdl-21096749

RESUMO

Cardiovascular diseases, specifically myocardial infarction and end-stage heart failure represent some of the major pathologies that threaten human life. Here we present a novel approach for a bioactive cardiac patch based on a combination of biomedical and textile manufacturing techniques in concert with nano-biotechnology based tissue-engineering stratagems. The technological goal is to create BioNanoTextiles™ (BNT) by using "conventional" fabrics as templates for creating three-dimensional nanofibrous scaffolds. Electrospinning nanofibrous scaffolds templated after "ordinary" textiles is a novel way to create complex-patterned, 3-D scaffolds intrinsically mimicking some of the anisotropic structural features of the ventricular wall's extracellular matrix. In preliminary studies, we established that this approach will yield anisotropic 3-D scaffolds with mechanical properties dependent upon the yarn type of the textile-templates. These scaffolds are biocompatible, as inferred from their support of H9C2 cardiac myoblast adhesion which promotes their proliferation as well as cardiac-like anisotropic organization. The use of textile manufacturing strategies will enhance the complexity of the 3-D scaffold structures and enable their commercialization, while providing an opportunity for the textile industry to advance established "low-tech" manufacturing technologies into the realm of "high-tech" BioNanoTextiles.


Assuntos
Nanocompostos/química , Medicina Regenerativa/métodos , Têxteis , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Adesão Celular/fisiologia , Linhagem Celular , Forma Celular/fisiologia , Técnicas Eletroquímicas , Teste de Materiais , Microscopia Eletrônica de Varredura , Mioblastos/citologia , Nanocompostos/ultraestrutura , Ratos
4.
J Cell Biochem ; 109(6): 1201-9, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20127722

RESUMO

T lymphocytes fail to proliferate or secrete cytokines in response to T cell receptor (TCR) agonists during culture in spaceflight or ground-based microgravity analogs such as rotating wall-vessel (RWV) bioreactors. In RWVs, these responses can be rescued by co-stimulation with sub-mitogenic doses of the diacyl glycerol (DAG) mimetic phorbol myristate acetate. Based on this result we hypothesized that TCR activation is abrogated in the RWV due to impaired DAG signaling downstream of the TCR. To test this hypothesis we compared TCR-induced signal transduction by primary, human, CD4(+) T cells in RWV, and static culture. Surprisingly, we found little evidence of impaired DAG signaling in the RWV. Upstream of DAG, the tyrosine phosphorylation of several key components of the TCR-proximal signal was not affected by culture in the RWV. Similarly, the phosphorylation and compartmentalization of ERK and the degradation of IkappaB were unchanged by culture in the RWV indicating that RAS- and PKC-mediated signaling downstream of DAG are also unaffected by simulated microgravity. We conclude from these data that TCR signaling through DAG remains intact during culture in the RWV, and that the loss of functional T cell activation in this venue derives from the affect of simulated microgravity on cellular processes that are independent of the canonical TCR pathway.


Assuntos
Reatores Biológicos , Linfócitos T CD4-Positivos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Células Cultivadas , Diglicerídeos/metabolismo , Eletroforese em Gel de Poliacrilamida , Citometria de Fluxo , Humanos , Fosforilação , Ausência de Peso
5.
Endothelium ; 15(5-6): 288-98, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19065320

RESUMO

Very few studies describe endothelial cell (EC) properties under three-dimensional (3D) conditions using (31)P magnetic resonance spectroscopy (MRS). The authors developed a model in which living ECs growing in Matrigel threads (3D conditions) for 5 days are monitored by (31)P MRS, providing the fingerprint of the major EC phosphometabolites. Organic extracts of membranal phospholipids were also analyzed by (31)P MRS. For comparison and as a model for two-dimensional (2D) tissue culture conditions, (31)P MRS spectra of aqueous extracts of EC phosphometabolites grown under 2D conditions were also evaluated. The phosphometabolites fingerprint of the cells cultured under 3D was significantly different from that of ECs maintained under 2D. Moreover, the pattern of phosphometabolites was affected by coculture with C6-glioma cells and upon treatment with valproic acid, which is under clinical investigation as an antioangiogenic anticancer drug. The major effects were modulation of (i) energy metabolism intermediates such as phosphocreatine, (ii) precursors of phospholipids such as phosphomonoesters, and (iii) degradation products of phospholipids such as glycerophosphocholine. This endothelial model will be usefull as an enabling platform technology for tissue engineering.


Assuntos
Reatores Biológicos , Células Endoteliais/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Neuroglia/metabolismo , Fosfolipídeos/metabolismo , Ácido Valproico/farmacologia , Animais , Anticonvulsivantes/farmacologia , Bioquímica/métodos , Bioensaio/instrumentação , Bioensaio/métodos , Comunicação Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Colágeno/farmacologia , Combinação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Laminina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Técnicas de Cultura de Órgãos/métodos , Fosfocreatina/metabolismo , Isótopos de Fósforo/química , Proteoglicanas/farmacologia , Ratos
6.
Endothelium ; 15(5-6): 299-307, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19065321

RESUMO

In the accompanying study, the authors presented phosphometabolite patterns of endothelial cells grown under three-dimensional (3D) conditions using (31)P magnetic resonance spectroscopy (MRS). Here the authors describe the effect of nonsteroidal anti-inflammatory drugs (NSAIDs), using this enabling platform technology, which is relevant for evaluating drug effects in tissue-engineered endothelial constructs. Treatment with indomethacin significantly changed the phosphometabolite fingerprint in this endothelial model, by, respectively, increasing (81%) and decreasing (42%) glycerophosphocholine (GPC) and phosphomonoesters (PM). Furthermore, a safer approach using a NSAID prodrug was also demonstrated in this study with a indomethacin phospholipid-derived prodrug (DP-155). Like the parental drug, DP-155 increased and decreased the levels of GPC and PM by 100% and 20%, respectively. These changes represent useful biomarkers to monitor NSAID effects on endothelized tissue-engineered constructs for the purpose of controlling endothelial cell survival and inflammation upon implantation.


Assuntos
Anti-Inflamatórios/farmacologia , Reatores Biológicos , Células Endoteliais/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Neuroglia/metabolismo , Fosfolipídeos/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Bioquímica/métodos , Bioensaio/instrumentação , Bioensaio/métodos , Comunicação Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Colágeno/farmacologia , Combinação de Medicamentos , Células Endoteliais/efeitos dos fármacos , Indometacina/análogos & derivados , Indometacina/farmacologia , Inflamação/tratamento farmacológico , Laminina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Técnicas de Cultura de Órgãos/métodos , Fosfatidilcolinas/farmacologia , Isótopos de Fósforo/química , Proteoglicanas/farmacologia , Ratos
7.
Am J Physiol Heart Circ Physiol ; 294(2): H1027-35, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18156201

RESUMO

The migration of vascular endothelial cells under flow can be modulated by the addition of chemical or mechanical stimuli. The aim of this study was to investigate how topographic cues derived from a substrate containing three-dimensional microtopography interact with fluid shear stress in directing endothelial cell migration. Subconfluent bovine aortic endothelial cells were seeded on fibronectin-coated poly(dimethylsiloxane) substrates patterned with a combinatorial array of parallel and orthogonal microgrooves ranging from 2 to 5 microm in width at a constant depth of 1 microm. During a 4-h time-lapse observation in the absence of flow, the majority of the prealigned cells migrated parallel to the grooves with the distribution of their focal adhesions (FAs) depending on the groove width. No change in this migratory pattern was observed after the cells were exposed to moderate shear stress (13.5 dyn/cm(2)), irrespective of groove direction with respect to flow. After 4-h exposure to high shear stress (58 dyn/cm(2)) parallel to the grooves, the cells continued to migrate in the direction of both grooves and flow. By contrast, when microgrooves were oriented perpendicular to flow, most cells migrated orthogonal to the grooves and downstream with flow. Despite the change in the migration direction of the cells under high shear stress, most FAs and actin microfilaments maintained their original alignment parallel to the grooves, suggesting that topographic cues were more effective than those derived from shear stress in guiding the orientation of cytoskeletal and adhesion proteins during the initial exposure to flow.


Assuntos
Movimento Celular/fisiologia , Células Endoteliais/fisiologia , Células Endoteliais/ultraestrutura , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Aorta Torácica/citologia , Prótese Vascular , Bovinos , Células Cultivadas , Interpretação Estatística de Dados , Microscopia Confocal , Microscopia Eletrônica de Varredura , Desenho de Prótese , Reologia , Estresse Mecânico , Propriedades de Superfície , Ferimentos e Lesões/patologia
8.
J Biomater Sci Polym Ed ; 18(10): 1263-81, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17939885

RESUMO

Many different types of microelectrodes have been developed for use as a direct brain-machine interface (BMI) to chronically recording single-neuron action potentials from ensembles of neurons. Unfortunately, the recordings from these microelectrode devices are not consistent and often last for only on the order of months. For most microelectrode types, the loss of these recordings is not due to failure of the electrodes, but most likely due to damage to surrounding tissue that results in the formation of non-conductive glial scar. Since the extracellular matrix consists of nanostructured fibrous protein assemblies, we have postulated that neurons may prefer a more complex surface structure than the smooth surface typical of thin-film microelectrodes. This porous structure could then act as a drug-delivery reservoir to deliver bioactive agents to aid in the repair or survival of cells around the microelectrode, further reducing the glial scar. We, therefore, investigated the suitability of a nanoporous silicon surface layer to increase the biocompatibility of our thin film ceramic-insulated multisite electrodes. In vitro testing demonstrated increased extension of neurites from PC12 pheochromocytoma cells on porous silicon surfaces compared to smooth silicon surfaces. Moreover, the size of the pores and the pore coverage did not interfere with this bioactive surface property, suggesting that large highly porous nanostructured surfaces can be used for drug delivery. The most porous nanoporous surfaces were then tested in vivo and found to be more biocompatible than smooth surface, producing less glial activation and allowing more neurons to remain close to the device. Collectively, these results support our hypothesis that nanoporous silicon may be an ideal material to improve biocompatibility of chronically implanted microelectrodes. The next step in this work will be to apply these surfaces to active microelectrodes, use them to deliver bioactive agents, and test that they do improve neural recordings.


Assuntos
Materiais Biocompatíveis/química , Nanoestruturas/química , Neurônios/metabolismo , Silício/química , Potenciais de Ação , Animais , Encéfalo/metabolismo , Proliferação de Células , Sistemas de Liberação de Medicamentos , Eletrodos , Eletrofisiologia , Imuno-Histoquímica/métodos , Microscopia Eletrônica de Varredura , Neuritos/metabolismo , Neuroglia/metabolismo , Células PC12 , Ratos , Propriedades de Superfície
9.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L639-50, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17526596

RESUMO

In extending our previous studies toward development of an engineered distal lung tissue construct (M. J. Mondrinos, S. Koutzaki, E. Jiwanmall, M. Li, J. P. Dechadarevian, P. I. Lelkes, and C. M. Finck. Tissue Eng 12: 717-728, 2006), we studied the effects of exogenous fibroblast growth factors FGF10, FGF7, and FGF2 on mixed populations of embryonic day 17.5 murine fetal pulmonary cells cultured in three-dimensional collagen gels. The morphogenic effects of the FGFs alone and in various combinations were assessed by whole mount immunohistochemistry and confocal microscopy. FGF10/7 significantly increased epithelial budding and proliferation; however, only FGF10 alone induced widespread budding. FGF7 alone induced dilation of epithelial structures but not widespread budding. FGF2 alone had a similar dilation, but not budding, effect in epithelial structures, and, in addition, significantly enhanced endothelial tubular morphogenesis and network formation, as well as mesenchymal proliferation. The combination of FGF10/7/2 induced robust budding of epithelial structures and the formation of uniform endothelial networks in parallel. These data suggest that appropriate combinations of exogenous FGFs chosen to target specific FGF receptor isoforms will allow for control of lung epithelial and mesenchymal cell behavior in the context of an engineered system. We propose that tissue-engineered fetal distal lung constructs could provide a potential source of tissue or cells for lung augmentation in pediatric pulmonary pathologies, such as pulmonary hypoplasia and bronchopulmonary dysplasia. In addition, engineered systems will provide alternative in vitro venues for the study of lung developmental biology and pathobiology.


Assuntos
Feto/citologia , Pulmão/embriologia , Modelos Biológicos , Engenharia Tecidual/métodos , Animais , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Feto/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Pulmão/irrigação sanguínea , Camundongos , Microscopia Confocal , Morfogênese/efeitos dos fármacos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo
10.
J Appl Physiol (1985) ; 100(4): 1287-92, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16384837

RESUMO

Depressed immune function is a well-documented effect of spaceflight. Both in-flight studies and ground-based studies using microgravity analogs, such as rotating wall vessel (RWV) bioreactors, have demonstrated that mitogen-stimulated T lymphocytes exhibit decreased proliferation, IL-2 secretion, and activation marker expression in true microgravity and the dynamic RWV-culture environment. This study investigates the kinetics of RWV-induced T lymphocyte inhibition by monitoring the ability of Balb/c mouse splenocytes to become activated under static culture conditions after concanavalin A (Con A) stimulation in an RWV. Splenocytes were stimulated with Con A and cultured for up to 24 h in the RWV before being allowed to "recover" under static culture conditions in the continued presence of Con A. The T-lymphocyte fraction of splenocytes was assayed during the recovery period for IL-2 secretion, expansion of the T-lymphocyte population, and expression of the activation marker CD25. Our results indicate that CD25 expression was not affected by any duration of RWV exposure. In contrast, proliferation and IL-2 secretion were inhibited by >8 and 12 h of exposure, respectively. Culture in the RWV for 24 h resulted in a near-complete loss of cellular viability during the recovery period, which was not seen in cells maintained in the RWV for 16 h or less. Taken together, these results indicate that for up to 8 h of RWV culture activation is not significantly impaired upon return to static conditions; longer duration RWV culture results in a gradual loss of activation during the recovery period most likely because of decreased T-cell viability and/or IL-2 production.


Assuntos
Reatores Biológicos , Ativação Linfocitária , Mitógenos/farmacologia , Linfócitos T/efeitos dos fármacos , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Concanavalina A/farmacologia , Interleucina-2/metabolismo , Cinética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Rotação , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Linfócitos T/metabolismo , Simulação de Ausência de Peso/instrumentação
11.
Endothelium ; 8(1): 49-63, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11409851

RESUMO

The vasopressin-activated calcium-mobilizing (VACM-1) protein is a unique arginine vasopressin (AVP) receptor which shares sequence homology with the cullins, genes involved in the regulation of cell cycle transitions. Unlike either cullins or AVP receptors, however, VACM-1 is expressed exclusively in the vascular endothelial cells and in the renal collecting tubule cells. In order to test the hypothesis that the expression of VACM-1 might be correlated with the cell cycle, and to establish an endothelial cell model for the VACM-1 receptor, we examined VACM-1 expression in rat adrenal medulla endothelial cells (RAMEC). Northern and Western blot analyses of mRNA and protein from RAMEC identified presence of 6.4 kb mRNA and a Mr 81 kDa protein, respectively. Immunostaining of RAMEC with anti-VACM-1 antibodies and Western blot analyses indicated that in RAMEC, VACM-1 protein expression is dependent on the cell cycle. VACM-1 protein virtually disappears during the S phase and localizes to the cytosol during cell division and to the cell membrane at the completion of cytokinesis. Furthermore, pretreatment of RAMEC with anti-VACM-1 specific antibodies increased basal levels of Ca2+and attenuated the AVP-dependent increase in cytosolic Ca2+. In summary, these results indicate that VACM-1 protein expression in RAMEC membrane is linked to the cell cycle, and consequently, VACM-1 may be involved in the regulation of cell division.


Assuntos
Medula Suprarrenal/fisiologia , Ciclo Celular/fisiologia , Proteínas Culina , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/genética , Receptores de Vasopressinas/genética , Transcrição Gênica , Medula Suprarrenal/citologia , Animais , Afidicolina/farmacologia , Sinalização do Cálcio/fisiologia , Ciclo Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Núcleo Celular/fisiologia , Células Cultivadas , Endotélio/citologia , Endotélio/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Ratos
12.
Neurotox Res ; 3(2): 189-203, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14715473

RESUMO

Diabetic neuropathies, affecting the autonomic, sensory, and motor peripheral nervous system, are among the most frequent complications of diabetes. The symptoms of diabetic polyneuropathies are multi-faceted; the etiology and the underlying mechanisms are as yet unclear. Clinical studies established a significant correlation between the control of the patients' blood glucose level and the severity of the damage to the peripheral nervous system. Recent in vitro studies suggest that elevated glucose levels induced dysfunction and apoptosis in cultured cells of neuronal origin, possibly through the formation of reactive oxygen species (ROS). Based on these results, we hypothesized that elevated glucose levels impair neuronal survival and function via ROS dependent intracellular signaling pathways. In order to test this hypothesis, we cultured neural crest-derived PC12 pheochromocytoma cells under euglycemic (5 mM) and hyperglycemic (25 mM) conditions. Continuous exposure of undifferentiated PC12 cells for up to 72 h to elevated glucose induced the enhanced generation of ROS, as assessed from the increase in the cell-associated fluorescence of the ROS-sensitive fluorogenic indicator, 2,7-dichlorodihydrofluorescein diacetate. In cells cultured in high glucose, both basal and secretagogue-stimulated catecholamine release were enhanced. Furthermore, high glucose, reduced (by ca. 30%) the rate of cell proliferation and enhanced the occurrence of apoptosis, as assessed by DNA fragmentation, TUNEL assay and the activation of an apoptosis-specific protease, caspase CCP32. Elevated glucose levels significantly attenuated nerve growth factor (NGF)-induced neurite extension, as quantitated by computer-aided image analysis. Culturing PC12 cells in high glucose resulted in alterations in basal and NGF-stimulated mitogen-activated protein kinase (MAPK) signaling pathways, specifically in a switch from the neuronal survival/differentiation-associated MAPK ERK to that of apoptosis/stress-associated MAPK p38 and JNK. Based on our results we present a model in which the prolonged, excess formation of ROS represents a common mechanism for hyperglycemia-induced damage to neuronal cells. We propose that this simple in vitro system might serve as an appropriate model for evaluating some of the effects of elevated glucose on cultured cells of neuronal origin.

13.
Endothelium ; 8(4): 243-53, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11824476

RESUMO

We previously reported that short-term exposure of cultured rat adrenal medullary endothelial cells (RAMEC) to thrombin enhances the subendothelial deposition of extracellular matrix (ECM) proteins fibronectin, laminin, and collagen types I (C-I) and IV (C-IV) (Papadimitriou et al. 1997). In this work, we extended our previous studies on factors that effect ECM protein deposition to include agents that activate or inhibit some of the most common intracellular signals such as cAMP, protein kinase C (PKC), and calcium. Furthermore, we investigated the possible link between the observed alterations in ECM protein deposition and the secretion of matrix metalloproteinase-2 (MMP-2). Forskolin (adenylyl cyclase activator) caused a dose-dependent increase in the deposition of all four ECM proteins studied. Isoproterenol beta-adrenergic receptor agonist) and the membrane permeant cAMP analogue dibutyryl-cAMP significantly increased the deposited amounts of ECM proteins at low concentrations, and this increase was reversed at higher concentrations of both agents. All these agents had the opposite effect on MMP-2 secretion, increasing it at doses where they decreased ECM protein deposition and vice versa. However, elevation of cAMP by the phosphodiesterase inhibitor IBMX had no effect either on the deposited amounts of any of the ECM proteins studied or on MMP-2 secretion. Activation of PKC by phorbol ester (PMA) resulted in a decrease in ECM protein deposition and an increase in MMP-2 secretion. Finally, chelation of intercellular calcium with BAPTA-AM resulted in an increased ECM deposition and a decrease in MMP-2 secretion. Our results show a complex pattern of regulation of ECM protein deposition by cAMP-mobilizing agents and also indicate an inverse correlation between ECM protein deposition and secretion of MMP-2. The concerted regulation of both of these processes is essential in the formation of new blood vessels, and for the integrity of the vascular wall.


Assuntos
Medula Suprarrenal/citologia , Ácido Egtázico/análogos & derivados , Endotélio/citologia , Matriz Extracelular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Animais , Técnicas de Cultura de Células , Colforsina/farmacologia , Ácido Egtázico/farmacologia , Endotélio/metabolismo , Endotélio/ultraestrutura , Ativação Enzimática , Matriz Extracelular/efeitos dos fármacos , Proteínas da Matriz Extracelular/efeitos dos fármacos , Ratos , Sistemas do Segundo Mensageiro , Acetato de Tetradecanoilforbol/farmacologia
14.
Gen Pharmacol ; 34(2): 107-16, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10974418

RESUMO

Osmotic cell swelling activates an outwardly rectifying Cl(-) current in endothelial cells that is mediated by volume-regulated anion channels (VRACs). In the past, we have shown that serum-induced proliferation of endothelial cells is arrested in the presence of compounds that potently block the endothelial VRACs. Here we report on the effects of four chemically distinct VRAC blockers [5-nitro-2-(3-phenylpropylamino)benzoic acid] (NPPB), mibefradil, tamoxifen, and clomiphene-on several models of experimental angiogenesis. Mibefradil (20 microM), NPPB (100 microM), tamoxifen (20 microM), and clomiphene (20 microM) inhibited tube formation by rat microvascular endothelial cells plated on matrigel by 42.9 +/- 8.8%, 25.3 +/- 10.4%, 32.2 +/- 4.5%, and 20 +/- 5.8%, respectively (p < 0.05). Additionally, NPPB (50-100 microM) and mibefradil (10-30 microM) significantly inhibited bFGF (10 ng/ml) + TNFalpha (2.5 ng/ml)-stimulated microvessel formation by human microvascular endothelial cells plated on fibrin by 30-70%. Furthermore, NPPB, mibefradil, and clomiphene concentration dependently inhibited spontaneous microvessel formation in the rat aorta-ring assay and vessel development in the chick chorioallantoic membrane assay. These results suggest that VRAC blockers are potent inhibitors of angiogenesis and thus might serve as therapeutic tools in tumor growth and other angiogenesis-dependent diseases.


Assuntos
Inibidores da Angiogênese/farmacologia , Canais Iônicos/antagonistas & inibidores , Neovascularização Fisiológica/efeitos dos fármacos , Alantoide/irrigação sanguínea , Alantoide/efeitos dos fármacos , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/crescimento & desenvolvimento , Bovinos , Células Cultivadas , Embrião de Galinha , Córion/irrigação sanguínea , Córion/efeitos dos fármacos , Clomifeno/farmacologia , Colágeno , Combinação de Medicamentos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fibrina , Humanos , Técnicas In Vitro , Laminina , Masculino , Mibefradil/farmacologia , Nitrobenzoatos/farmacologia , Proteoglicanas , Ratos , Ratos Wistar , Tamoxifeno/farmacologia
15.
Artif Organs ; 24(2): 137-47, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10718767

RESUMO

In using autologous muscles for cardiac assistance, it is crucial to reduce ischemia-reperfusion injury in the surgically traumatized skeletal muscle. In adult sheep, we developed a simple model of surgically designed 2 latissimus dorsi muscle leaflets by modifying the vascular supply to these leaflets. Three pockets with graded injury were established, and muscle morphology and vascular remodeling were monitored in 3 experimental groups: muscle leaflets without any treatment (Group 1, n = 6) that served as controls; muscle leaflets integrated with a fibrin interlayer (Group 2, n = 6); and leaflets integrated with fibrin and entrapped pyrrolostatin (Group 3, n = 6). We applied the fibrinogen and thrombin solutions, which polymerize to form a three-dimensional meshwork joining the tissues, creating a provisional matrix for angiogenesis, and acting as a delivery depot for agents aimed at minimizing ischemia-reperfusion lesion formation. After 2 months, the muscle leaflets biointegrated with the fibrin interface showed none of the signs of necrosis or ischemia-reperfusion lesions seen in the controls. Although no angiogenic factors were incorporated, the fibrin interlayer rapidly (<2 weeks) became a densely vascularized tissue replete with a voluminous capillary network. In contrast, controls showed poor bonding between the tissues, muscle fiber deterioration, and a compromised vascular network. Muscle structure was best preserved and angiogenesis was greatest when pyrrolostatin, a free radical scavenger, was added to the fibrin meshwork to reduce damage caused by overproduction of free radicals. This newly designed model will be useful to study many current approaches in cardiovascular biology, from pharmaceuticals to gene therapy, which might prove advantageous in muscle-designed cardiac assistance.


Assuntos
Cardiomioplastia , Músculo Esquelético/transplante , Retalhos Cirúrgicos/patologia , Animais , Capilares/ultraestrutura , Preparações de Ação Retardada , Fibrina/uso terapêutico , Fibrinogênio/uso terapêutico , Sequestradores de Radicais Livres/uso terapêutico , Radicais Livres/antagonistas & inibidores , Sobrevivência de Enxerto , Microscopia Eletrônica , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/patologia , Neovascularização Fisiológica , Prolina/análogos & derivados , Prolina/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Ovinos , Retalhos Cirúrgicos/irrigação sanguínea , Trombina/uso terapêutico , Adesivos Teciduais/uso terapêutico , Transplante Autólogo
16.
Am J Physiol ; 277(2): C233-42, 1999 08.
Artigo em Inglês | MEDLINE | ID: mdl-10444399

RESUMO

We tested the hypothesis that elevated blood pressure, a known stimulus for vascular remodeling and an independent risk factor for the development of atherosclerotic disease, can modulate basal and cytokine-induced tissue factor (TF; CD 142) expression in cultured human endothelial cells (EC). Using a chromogenic enzymatic assay, we measured basal and tumor necrosis factor-alpha (TNF-alpha; 10 ng/ml, 5 h)-induced TF activities in human aortic EC (HAEC) and vena cava EC (HVCEC) cultured at atmospheric pressure and at 170 mmHg imposed pressure for up to 48 h. Basal TF activities were 22 +/- 10 U/mg protein for HAEC and 14 +/- 9 U/mg protein for HVCEC and were upregulated in both cell types >10-fold by TNF-alpha. Exposure to pressure for 5 h induced additional elevation of basal TF activity by 47 +/- 16% (P < 0.05, n = 6) for HAEC and 17 +/- 5% (P < 0.05, n = 3) for HVCEC. Pressurization also enhanced TF activity in TNF-alpha-treated cells from 240 +/- 28 to 319 +/- 32 U/mg protein in HAEC (P < 0.05, n = 4) and from 148 +/- 25 to 179 +/- 0.8 U/mg protein (P < 0.05, n = 3) in HVCEC. Cytokine stimulation caused an approximately 100-fold increase in steady-state TF mRNA levels in HAEC, whereas pressurization did not alter either TF mRNA or cell surface antigen expression, as determined by quantitative RT-PCR methodology and ELISA. Elevated pressure, however, modulated the EC plasma membrane organization and/or permeability as inferred from the increased cellular uptake of the fluorescent amphipathic dye merocyanine 540 (33 +/- 7%, P < 0.05). Our data suggest that elevated static pressure modulates the hemostatic potential of vascular cells by modifying the molecular organization of the plasma membrane.


Assuntos
Endotélio Vascular/metabolismo , Proteínas Recombinantes/metabolismo , Tromboplastina/metabolismo , Aorta/citologia , Pressão Atmosférica , Permeabilidade da Membrana Celular/fisiologia , Células Cultivadas , Citocinas/farmacologia , Técnicas Citológicas/instrumentação , Endotélio Vascular/citologia , Corantes Fluorescentes/farmacocinética , Humanos , Pressão , Pirimidinonas/farmacocinética , RNA Mensageiro/metabolismo , Tromboplastina/genética , Veias Cavas/citologia
17.
Int J Dev Neurosci ; 17(1): 45-55, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10219960

RESUMO

The expression of phenylethanolamine N-methyl transferase (EC 2. 1.1.2.8, PNMT), the final enzyme in the cascade of catecholamine synthesis, is differentially regulated in adrenergic neurons in the brain and in adrenal chromaffin cells. Using reverse transcription-polymerase chain reaction-based techniques, we detected in the prenatal developing rat brainstem, two species of PNMT mRNA which were produced by a rare alternative splicing mechanism known as intron retention. The spliced, intronless message was downregulated postnatally, while the intron-retained mRNA species continued to be constitutively expressed through adulthood. By contrast in the adrenals, at all stages of development examined, only the intronless message was expressed. In line with previous reports on the failure of glucocorticoids to induce PNMT expression in the brain, the pattern of PNMT splicing in brainstem explants was not affected by the presence of the synthetic glucocorticoid dexamethasone. Undifferentiated sympathoadrenal PC12 pheochromocytoma cells expressed very low basal levels of both mRNA variants, accompanied by a very low basal PNMT enzymatic activity. Exposure of PC12 cells to dexamethasone resulted in the upregulation of only the spliced mRNA variant concomitant with a 3-fold increase in PNMT enzymatic activity. In contrast, treatment of PC 12 cells with nerve growth factor (NGF) enhanced the expression of both the intron-retained and the intronless mRNA species without changes in the basal enzyme activity. This latter result suggests that the translation of the intronless mRNA species may be regulated by the intron-retained mRNA species, which by itself may yield a truncated, yet enzymatically functional translational product. Our data suggest that the tissue-specific regulation of PNMT expression is based on a rare alternative splicing mechanism termed intron retention, and that in the adrenal, but not in the brain, this mechanism is sensitive to regulation by glucocorticoids. Thus, this system is uniquely suited for studying the hormonal control of tissue-specific splicing in the nervous system.


Assuntos
Medula Suprarrenal/enzimologia , Tronco Encefálico/enzimologia , Proteínas Fetais/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Íntrons/genética , Isoenzimas/genética , Proteínas do Tecido Nervoso/genética , Feniletanolamina N-Metiltransferase/genética , Splicing de RNA , RNA Mensageiro/metabolismo , Medula Suprarrenal/embriologia , Medula Suprarrenal/crescimento & desenvolvimento , Animais , Sequência de Bases , Tronco Encefálico/embriologia , Dexametasona/farmacologia , Eletroforese em Gel de Ágar , Indução Enzimática/efeitos dos fármacos , Proteínas Fetais/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/fisiologia , Isoenzimas/biossíntese , Dados de Sequência Molecular , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Fatores de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Especificidade de Órgãos , Células PC12/efeitos dos fármacos , Células PC12/enzimologia , Feniletanolamina N-Metiltransferase/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico
18.
Pancreas ; 18(1): 96-103, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9888665

RESUMO

Recent epidemiologic evidence suggests that patients with chronic pancreatitis (CP) have an increased risk of developing pancreatic carcinoma (PCA). In spite of numerous similarities in both diseases, mechanisms for progression from CP to PCA are poorly understood. We hypothesized that enhanced angiogenesis might play a pivotal role in the etiology and histopathology of both CP and PCA, and thus form a possible link between precancer and carcinoma. In surgical specimens of 18 patients with CP, 10 with PCA, and 18 controls, absolute numbers of blood vessels and relative blood vessel density were assessed after immunostaining of endothelial cells for von Willebrand factor and PECAM-1 (platelet/ endothelial cell adhesion molecule-1). Furthermore, the expression of cell adhesion molecules ICAM-1 (intercellular adhesion molecule-1) and VCAM-1 (vascular cell adhesion molecule-1) and of VEGF (vascular endothelial growth factor) was investigated in all specimens. Both CP and PCA exhibited areas of high vascular density ("hot spots"). The mean number of blood vessels in these areas in PCA was 132.2+/-16.8 per mm2, and in CP, 99.2+/-7.4 per mm2. The mean vessel count in controls was 25.1+/-5.1. Relative vessel density was increased in both PCA (41.3+/-3.5%) and CP (30.6+/-2.6%) versus controls (8.0+/-0.8%). Both absolute vessel count and relative vessel density were significantly higher (p<0.05) in PCA than in CP. Enhanced expression of ICAM-1 in CP and PCA was seen in ductal cells in CP and cancer cells. In controls, ICAM-1 and VCAM-1 were expressed only at low levels in endothelial cells. VCAM-1 was strongly expressed in acinar cells as well as in ductal cells. In CP and PCA, VEGF was strongly expressed in ductal cells in CP as well as in cancer cells. We show for the first time that angiogenic activity is increased in both CP and PCA. Based on this study, we suggest that antiangiogenesis might be a novel target for prevention or therapy in chronic pancreatic diseases.


Assuntos
Adenocarcinoma/patologia , Moléculas de Adesão Celular/análise , Fatores de Crescimento Endotelial/análise , Regulação da Expressão Gênica , Linfocinas/análise , Neovascularização Patológica/patologia , Neoplasias Pancreáticas/patologia , Pancreatite/patologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Moléculas de Adesão Celular/genética , Doença Crônica , Fatores de Crescimento Endotelial/genética , Humanos , Metástase Linfática , Linfocinas/genética , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Pancreatite/genética , Pancreatite/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula 1 de Adesão de Célula Vascular/análise , Molécula 1 de Adesão de Célula Vascular/genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Fator de von Willebrand/análise
19.
J Pharmacol Exp Ther ; 288(2): 399-406, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9918538

RESUMO

Pardaxin, an excitatory neurotoxin, induced dopamine release from pheochromocytoma (PC12) cells both in the presence and absence of extracellular calcium ([Ca]o). In the presence of extracellular calcium, nifedipine, an L-type calcium channel blocker, did not affect dopamine release, whereas 1,2-bis (2-aminophenoxy) ethane N,N, N'N'-tetra-acetic acid (BAPTA), a chelator of cytosolic calcium, and dantrolene, a blocker of calcium release from intracellular stores, inhibited only partially (30-40%) pardaxin-induced dopamine release. In the absence of [Ca]o, BAPTA and dantrolene were ineffective. Pardaxin stimulated the arachidonic acid (AA) cascade in PC12 cells independently of [Ca]o. The phospholipase inhibitors mepacrine and bromophenacyl bromide inhibited both pardaxin-induced AA release and pardaxin-induced dopamine release. Dopamine release induced by pardaxin also was blocked by the lipoxygenase inhibitors nordihydroguaiaretic acid, esculetin, and 2-(12-hydroxydodeca-5, 10-diynyl)-3,5,6-trimethyl-1,4-benzoquinone. Under these conditions, a parallel reduction in 5-hydroxyeicosatetranoic acid release also was observed. Suppression of pardaxin-induced dopamine release by inhibitors of phospholipase A2 and lipoxygenase was more pronounced in calcium-free medium. These results indicate the involvement of the lipoxygenase pathway in pardaxin-induced dopamine release and suggest the use of this toxin as a novel pharmacological tool for investigating the mechanism of calcium-independent neurotransmitter release.


Assuntos
Cálcio/fisiologia , Dopamina/metabolismo , Eicosanoides/fisiologia , Venenos de Peixe/farmacologia , Neurotoxinas/farmacologia , Células PC12/efeitos dos fármacos , Células PC12/metabolismo , Animais , Ácido Araquidônico/metabolismo , Cálcio/metabolismo , Eicosanoides/biossíntese , Ratos , Taxa Secretória/efeitos dos fármacos
20.
Endothelium ; 6(1): 33-44, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9832331

RESUMO

The diversity in the local manifestation of inflammatory vascular lesions might be partially attributable to heterogenous cell adhesion molecule (CAM) expression among endothelial cells (EC) derived from different anatomical locations. We compared basal and tumor necrosis factor-alpha (TNFalpha, 0-100 ng/ml, 0-48 h)-induced intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expression in cultured human aortic EC (HAEC), vena cava EC (HVCEC), dermal microvascular EC (HMVEC), and vena cava smooth muscle cells (HVCSM), using a fluorescent ELISA and the competitive quantitative RT-PCR. We found marked differences in basal ICAM-1 expression, both at the protein and mRNA levels, such that HAEC>HVCEC approximately equal to HMVEC>>HVCSM. Basal VCAM-1 mRNA levels were significantly lower in HVCEC than in HAEC and HVCSM, while protein levels were indistinguishable. TNFalpha-induced ICAM-1 and VCAM-1 levels in all EC were similar and significantly higher than in HVCSM (2.5- and 5-fold, respectively). Dissimilar levels of basal and TNFalpha-induced CAM expression in vascular cells may explain the varied predisposition of different blood vessels to developing certain vasculopathies.


Assuntos
Endotélio Vascular/metabolismo , Molécula 1 de Adesão Intercelular/biossíntese , Músculo Liso Vascular/metabolismo , Molécula 1 de Adesão de Célula Vascular/biossíntese , Células Cultivadas , Humanos , Especificidade de Órgãos , Reação em Cadeia da Polimerase , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...