Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
BMC Med ; 10: 127, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23098093

RESUMO

Since the first description of the case of Auguste Deter, presented in Tübingen in 1906 by Alois Alzheimer, there has been an exponential increase in our knowledge of the neuropathological, cellular, and molecular foundation of Alzheimer's disease (AD). The concept of AD pathogenesis has evolved from a static, binary view discriminating cognitive normality from dementia, towards a dynamic view that considers AD pathology as a long-lasting morbid process that takes place progressively over years, or even decades, before the first symptoms become apparent, and thus operating in a continuum between the two aforementioned extreme states. Several biomarkers have been proposed to predict AD-related cognitive decline, initially in cases with mild cognitive impairment, and more recently in cognitively intact individuals. These early markers define at-risk individuals thought to be in the preclinical phase of AD. However, the clinical relevance of this preclinical phase remains controversial. The fate of such individuals, who are cognitively intact, but positive for some early AD biomarkers, is currently uncertain at best. In this report, we advocate the point of view that although most of these preclinical cases will evolve to clinically overt AD, some appear to have efficient compensatory mechanisms and virtually never develop dementia. We critically review the currently available early AD markers, discuss their clinical relevance, and propose a novel classification of preclinical AD, designating these non-progressing cases as 'stable asymptomatic cerebral amyloidosis'.


Assuntos
Doença de Alzheimer/diagnóstico , Doença de Alzheimer/patologia , Doenças Assintomáticas , Biomarcadores , Angiopatia Amiloide Cerebral/diagnóstico , Demência/diagnóstico , Humanos , Medição de Risco
2.
PLoS One ; 7(6): e39995, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761942

RESUMO

Human papillomaviruses (HPV) cause a variety of mucosal and skin lesions ranging from benign proliferations to invasive carcinomas. The clinical manifestations of infection are determined by host-related factors that define the natural anti-HPV barrier. Key elements of this barrier are the EVER1 and EVER2 proteins, as deficiency in either one of the EVER proteins leads to Epidermodysplasia Verruciformis (EV), a genodermatosis associated with HPV-induced skin carcinoma. Although EVERs have been shown to regulate zinc homeostasis in keratinocytes, their expression and function in other cell types that may participate to the anti-HPV barrier remain to be investigated. In this work, we demonstrate that EVER genes are expressed in different tissues, and most notably in lymphocytes. Interestingly, in contrast to the skin, where EVER2 transcripts are hardly detectable, EVER genes are both abundantly expressed in murine and human T cells. Activation of CD4+ and CD8+ T cells via the TCR triggers a rapid and profound decrease in EVER expression, accompanied by an accumulation of free Zn(2+) ions. Thus, EVER proteins may be involved in the regulation of cellular zinc homeostasis in lymphocytes. Consistent with this hypothesis, we show that the concentration of Zn(2+) ions is elevated in lymphoblastoid cells or primary T cells from EVER2-deficient patients. Interestingly, we also show that Zn(2+) excess blocks T-cell activation and proliferation. Therefore, EVER proteins appear as key components of the activation-dependent regulation of Zn(2+) concentration in T cells. However, the impact of EVER-deficiency in T cells on EV pathogenesis remains to be elucidated.


Assuntos
Ativação Linfocitária , Proteínas de Membrana/fisiologia , Papillomaviridae/imunologia , Linfócitos T/imunologia , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Feminino , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase
3.
Microbiol Mol Biol Rev ; 73(2): 348-70, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19487731

RESUMO

Infections by human papillomaviruses (HPVs) are the most frequently occurring sexually transmitted diseases. The crucial role of genital oncogenic HPV in cervical carcinoma development is now well established. In contrast, the role of cutaneous HPV in skin cancer development remains a matter of debate. Cutaneous beta-HPV strains show an amazing ubiquity. The fact that a few oncogenic genotypes cause cancers in patients suffering from epidermodysplasia verruciformis is in sharp contrast to the unapparent course of infection in the general population. Our recent investigations revealed that a natural barrier exists in humans, which protects them against infection with these papillomaviruses. A central role in the function of this HPV-specific barrier is played by a complex of the zinc-transporting proteins EVER1, EVER2, and ZnT-1, which maintain cellular zinc homeostasis. Apparently, the deregulation of the cellular zinc balance emerges as an important step in the life cycles not only of cutaneous but also of genital HPVs, although the latter viruses have developed a mechanism by which they can break the barrier and impose a zinc imbalance. Herein, we present a previously unpublished list of the cellular partners of EVER proteins, which points to future directions concerning investigations of the mechanisms of action of the EVER/ZnT-1 complex. We also present a general overview of the pathogenesis of HPV infections, taking into account the latest discoveries regarding the role of cellular zinc homeostasis in the HPV life cycle. We propose a potential model for the mechanism of function of the anti-HPV barrier.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Proteínas de Membrana/metabolismo , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/virologia , Feminino , Predisposição Genética para Doença , Homeostase , Humanos , Papillomaviridae/genética , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/genética , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/genética , Neoplasias do Colo do Útero/etiologia , Neoplasias do Colo do Útero/genética , Zinco/metabolismo
6.
J Exp Med ; 205(1): 35-42, 2008 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-18158319

RESUMO

Epidermodysplasia verruciformis (EV) is a genodermatosis associated with skin cancers that results from a selective susceptibility to related human papillomaviruses (EV HPV). Invalidating mutations in either of two genes (EVER1 and EVER2) with unknown functions cause most EV cases. We report that EVER1 and EVER2 proteins form a complex and interact with the zinc transporter 1 (ZnT-1), as shown by yeast two-hybrid screening, GST pull-down, and immunoprecipitation experiments. In keratinocytes, EVER and ZnT-1 proteins do not influence intracellular zinc concentration, but do affect intracellular zinc distribution. EVER2 was found to inhibit free zinc influx to nucleoli. Keratinocytes with a mutated EVER2 grew faster than wild-type keratinocytes. In transiently and stably transfected HaCaT cells, EVER and ZnT-1 down-regulated transcription factors stimulated by zinc (MTF-1) or cytokines (c-Jun and Elk), as detected with luciferase assays. To get some insight into the control of EV HPV infection, we searched for interaction between EVER and ZnT-1 and oncoproteins of cutaneous (HPV5) and genital (HPV16) genotypes. HPV16 E5 protein binds to EVER and ZnT-1 and blocks their negative regulation. The lack of a functional E5 protein encoded by EV HPV genome may account for host restriction of these viruses.


Assuntos
Epidermodisplasia Verruciforme/metabolismo , Epidermodisplasia Verruciforme/virologia , Regulação da Expressão Gênica , Proteínas de Membrana/fisiologia , Papillomaviridae/metabolismo , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/prevenção & controle , Zinco/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Predisposição Genética para Doença , Genoma Viral , Humanos , RNA Interferente Pequeno/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Zinco/química
7.
Brain Dev ; 29(5): 273-80, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17071037

RESUMO

Tuberous sclerosis (TS), autosomal dominant disorder manifested by the formation of usually benign tumors in the brain, heart, kidneys and skin, results from an inactivating mutation in one of two tumor suppressor genes TSC1 or TSC2. Protein products of these genes, hamartin and tuberin, respectively, have been shown to participate in the mTOR pathway controlling translation of approx. 10-15% of all proteins. In the current paper, we aimed at verifying whether hamartin and tuberin may also be implicated in the control of gene transcription. Very recently it has been hypothesized that the pathway triggered by WNT, one of embryonic growth factors involved in cell differentiation and migration, could be disturbed in TS. In order to test this hypothesis we evaluated samples of four subependymal giant cell astrocytomas (SEGAs), brain tumors developing in the progress of TS. We found that beta-catenin, transcription factor and mediator of WNT pathway activity is indeed present and active in SEGAs. mRNA transcripts for c-Myc and N-Myc, proteins whose transcription is regulated by beta-catenin, were upregulated in two of four SEGAs, while cyclin D1 mRNA was significantly higher in three SEGAs. At the same time, c-Myc and N-Myc proteins were detected in the same two samples. Thus, we show for the first time that aberrant WNT signaling may contribute to the pathogenesis of TS-associated SEGAs.


Assuntos
Astrocitoma/complicações , Astrocitoma/fisiopatologia , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/fisiopatologia , Transdução de Sinais/fisiologia , Esclerose Tuberosa/etiologia , Esclerose Tuberosa/fisiopatologia , Regulação para Cima/fisiologia , Proteínas Wnt/fisiologia , Western Blotting , Núcleo Celular/genética , Ciclina D1/genética , Ciclina D1/fisiologia , Genes myc/fisiologia , Humanos , Imuno-Histoquímica , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Translocação Genética/genética , beta Catenina/biossíntese , beta Catenina/genética , beta Catenina/metabolismo
8.
Int J Mol Med ; 17(5): 779-83, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16596260

RESUMO

The parathormone (PTH) production is controlled by calcium and vitamin D, which interact with the calcium-sensing receptor (CaSR) and vitamin D receptor (VDR), respectively. All of these elements control calcium homeostasis, which is crucial for many physiological processes. Thus, impairment of the upstream component of this system, e.g. a decrease of CaSR and/or VDR, could result in hyperparathyroidism (HPTH). Therefore, the aim of this study was to assess the expression of CaSR and VDR in a tertiary form of HPTH (T-HPTH). The study involved 19 T-HPTH patients qualified for parathyroidectomy and 21 control parathyroids harvested from multi-organ cadaver donors. The small fragments of harvested glands were homogenized and used for Western blot analysis, whereas the remaining tissues underwent routine hematoxylin-eosin staining or immunostaining for CaSR and VDR. Among 64 T-HPTH parathyroids, 58 revealed the morphology of benign hyperplasia, 2 were identified as adenoma and 4 were classified as normal; some glands displayed a mixed histological phenotype. Western blot analysis revealed a decrease of CaSR and VDR in hyperplasia and adenoma-derived samples. However, no correlation between the types of hyperplasia and receptor expression was observed. On the other hand, microscopic analysis of CaSR- and VDR-immunostained sections revealed a highly differentiated and significantly decreased mean expression of both receptors, which correlated with parathyroid histology. The reason behind the impaired expression of CaSR and VDR in T-HPTH is unclear. It presumably results from constant parathyroid stimulation at the stage of S-HPTH, followed by further development of polyclonal autonomy. However, the verification of this thesis requires further study.


Assuntos
Hiperparatireoidismo/patologia , Receptores de Calcitriol/biossíntese , Receptores de Detecção de Cálcio/biossíntese , Adulto , Western Blotting , Feminino , Humanos , Hiperparatireoidismo/metabolismo , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Glândulas Paratireoides/química , Glândulas Paratireoides/patologia , Receptores de Calcitriol/análise , Receptores de Detecção de Cálcio/análise
9.
Int J Mol Med ; 17(1): 135-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16328022

RESUMO

Pentoxifylline (PTX) is an unspecific inhibitor of phosphodiesterase activity that increases intracellular concentration of cyclic nucleotides, mainly cAMP. Since PTX improves microcirculatory blood flow, it is commonly and often chronically used in peripheral vascular diseases. On the other hand PTX also displays a variety of immunomodulatory activities. PTX inhibits natural cytotoxicity and it has previously been suggested that it could partially act also through its influence on perforin/granzyme-dependent pathways. However, the underlying mechanisms are obscure and it remains unknown whether PTX inhibits natural cytotoxicity influencing only leukocytes or also acting on target cells. In this study, we show that PTX inhibits expression of granzyme A in human leukocytes probably due to suppression of phosphodiesterase activity. Contrary, PTX does not affect expression of granzyme B and H. On the other hand we hypothesized that PTX could inhibit natural cytotoxicity not only affecting leukocytes but also due to generation of resistance to leukocyte-mediated cytotoxicity in target cells e.g. through overexpression of PI-9, a specific granzyme B inhibitor. We found that at the mRNA level, PTX stimulates expression of PI-9 in K562 cells. However, we did not observe such an influence at the protein level, in either K562 cells or in human leukocytes. It may suggest that other PTX-triggered molecular events may interfere with PI-9 overexpression in these cells at the further, post-transcriptional levels. According to these results, PTX did not affect resistance of target cells to natural cytotoxicity. Altogether, PTX inhibits natural cytotoxicity affecting mainly effector but not target cells and in case of the effector cells, besides previously reported mechanisms, it can also inhibit granzyme A expression.


Assuntos
Pentoxifilina/metabolismo , Inibidores de Fosfodiesterase/metabolismo , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/metabolismo , Serpinas/metabolismo , Bucladesina/metabolismo , Granzimas , Humanos , Células K562 , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Toxina Pertussis/metabolismo , Serina Endopeptidases/genética
10.
Int J Mol Med ; 16(2): 215-9, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16012752

RESUMO

Precise localization of parathyroid glands using 99mTc-labeled hexakis-2-methoxyisobutylisonitrile (99mTc-MIBI) scintigraphy could be affected by various biological factors. There is increasing evidence that radiotracer retention could be controlled by members of multidrug resistance (MDR) system, especially P-glycoprotein (P-gp). Since the role of P-gp in tertiary hyperparathyroidism (T-HPTH) scintigraphic studies is poorly recognized, the aim of the study was to compare the correlation between parathyroid P-gp expression and results of their scintigraphy in T-HPTH versus primary hyperparathyroidism (P-HPTH). P-HPTH (n = 19) and T-HPTH (n = 18) patients were subjected to 99mTc-MIBI scintigraphy followed by surgical treatment. The parathyroid glands were assessed in routine hematoxylin-eosin staining and P-gp expression was analyzed using immunohistochemistry. Parathyroids collected during cadaver donor multi-organ harvesting were used as a control. It has been found that P-HPTH-derived parathyroid glands with predominating adenoma morphology expressed less P-gp, as compared to P-gp-rich T-HPTH glands, mainly displaying nodular or diffused hyperplasia phenotype. This finding reversely correlated with results of 99mTc-MIBI scintigraphy. However, we did not observe any difference in P-gp expression nor scintigraphy result between nodular or diffused hyperplasia. Altogether, these data suggest that P-gp overexpression in T-HPTH could be responsible for decreased sensitivity of 99mTc-MIBI scintigraphy in those patients. Therefore, the recently proposed reduced neck exploration or limited parathyroid resection on the basis of scintigraphy could create the risk of persisted/recurrent hyperparathyroidism. However, this problem requires further study.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Hiperparatireoidismo/diagnóstico por imagem , Tecnécio Tc 99m Sestamibi , Adenoma/diagnóstico por imagem , Adenoma/metabolismo , Adenoma/patologia , Humanos , Hiperparatireoidismo/classificação , Hiperparatireoidismo/metabolismo , Hiperplasia , Imuno-Histoquímica , Glândulas Paratireoides/diagnóstico por imagem , Glândulas Paratireoides/metabolismo , Glândulas Paratireoides/patologia , Cintilografia , Sensibilidade e Especificidade , Tecnécio Tc 99m Sestamibi/farmacocinética
11.
Neuromolecular Med ; 7(4): 287-96, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16391386

RESUMO

Tuberous sclerosis is an autosomal-dominant disorder caused by the mutation of one of the two tumor suppressor genes: TSC1 or TSC2, encoding protein products, hamartin, and tuberin, respectively. Both proteins form intracellular complexes exerting inhibitory activity on mammalian target of rapamycin (mTOR) kinase. It has been demonstrated that signal transduction from tuberin to mTOR is mediated by a G protein, Ras homologue enriched in brain (Rheb). In normal cells, tuberin having GTPase-activating protein properties toward Rheb controls signals of nutrient depletion, hypoxia, or stress, not allowing activation of mTOR and subsequent protein translation and cell proliferation. However, when environmental conditions change, tuberin is phosphorylated and it forms a complex with hamartin is degraded, and downstream targets of mTOR, S6K, and eEF2K, can be activated. In this review, we summarize very recent information contributing to our knowledge of TSC2 regulation by four cellular signaling pathways: PI3K/Akt, Ras/MAPK, LKB1/AMPK, and REDD1.


Assuntos
Proteínas Quinases/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais , Serina-Treonina Quinases TOR , Fatores de Transcrição/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/fisiologia , Proteínas ras/fisiologia
12.
Int J Mol Med ; 14(5): 937-42, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15492869

RESUMO

Human autoimmune lymphoproliferative syndrome has been described as a result of various mutations concerning genes encoding receptor CD95 and/or its ligand - CD178. However, recently, several cases with identical clinical manifestation, despite a normal structure of CD95 or CD178 have also been reported. In this study we analyzed PBMC obtained from patient with clinically overt lymphoproliferative disorder. Using in vitro assays as well as molecular methods we tested expression and biological activity of CD95 and CD178 molecules. We found that analyzed patient's lymphocytes displayed normal cytotoxic activity against CD95-bearing targets. However, CD95-dependent induction of apoptosis in analyzed lymphocytes was diminished, as compared to healthy control. Surprisingly, the molecular studies did not reveal any abnormalities in structure of patient-derived CD95 receptor molecule. Therefore, expression of other factors involved in CD95-mediated signaling pathway was estimated using RNase protection assay. The expression of FADD was comparable to that of healthy control. However, it has been found that patient-derived lymphocytes expressed reduced amount of caspase-8 mRNA, as compared to control subject cells. This report confirms previous observations that lymphoproliferative disorder could be associated not only with CD95 and/or CD178 mutations, but also with dysfunction of other components of apoptosis induction pathway. However, the detailed molecular mechanism of observed abnormalities in caspase-8 expression remains to be elucidated.


Assuntos
Apoptose/genética , Doenças Autoimunes/imunologia , Caspases/genética , Linfócitos/patologia , Transtornos Linfoproliferativos/imunologia , Receptor fas/genética , Caspase 8 , Caspases/deficiência , Sobrevivência Celular/imunologia , Pré-Escolar , Proteína Ligante Fas , Feminino , Humanos , Ativação Linfocitária , Linfócitos/imunologia , Glicoproteínas de Membrana/genética , Reação em Cadeia da Polimerase , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Int J Mol Med ; 14(4): 725-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15375608

RESUMO

Pentoxifylline (PTX) is a methylxanthine derivative that unspecifically inhibits phosphodiesterase activity and thus, it increases intracellular concentration of cyclic nucleotides. Currently, PTX is commonly and chronically used in peripheral blood vessel diseases. Besides its well-known influence on rheologic properties of blood, PTX has also been found to decrease secretion of some cytokines such as IL-12, TNF and IFN-gamma and thus it could exert immunomodulatory activity. Furthermore, PTX inhibits lymphocyte cytotoxicity affecting the perforin-dependent pathway, both in humans and animals. It has also been shown recently that in some murine models, PTX promotes tumor growth. Such a phenomenon, at least partially, could result from PTX-dependent inhibition of natural cytotoxicity. However, the detailed mechanism of PTX influence on cytotoxic activity in humans has not been established. We hypothesized that PTX-dependent inhibition of natural cytotoxicity could result from decrease in perforin expression. In this study, it was shown that pentoxifylline only moderately inhibits perforin expression at the mRNA level in human peripheral blood mononuclear cells (PBMC), and this effect seems to be independent of intracellular cAMP concentration. On the other hand, PTX did not significantly influence perforin expression at the protein level. These results suggest that in humans, contrary to murine models, inhibition of perforin-dependent natural cytotoxicity through pentoxifylline does not result from changes in perforin production. Presumably, influence of PTX on natural cytotoxicity could be caused by e.g. interference with lymphocyte degranulation. Moreover, also other possibilities, such as PTX influence on conformational changes of perforin molecules and/or its influence on susceptibility of target cells to pore-forming of polyperforins cannot be excluded.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Pentoxifilina/farmacologia , Células Cultivadas , Humanos , Glicoproteínas de Membrana/genética , Perforina , Proteínas Citotóxicas Formadoras de Poros , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
Oncol Rep ; 11(5): 1121-5, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15069556

RESUMO

Pentoxifylline (PTX), a methylxanthine derivative, is commonly and in most cases chronically used in patients suffering from peripheral vascular diseases. It increases erythrocyte flexibility, reduces blood viscosity and improves microcirculatory flow. Moreover, PTX has been found to enhance anticancer activity of some chemotherapeutic agents and ionizing irradiation. On the other hand, PTX has been recently shown to inhibit anticancer response and promote murine tumor growth in liver. In this study we show that PTX facilitates development of murine colon adenocarcinoma- but not melanoma-derived tumors also in lungs. It could suggest that tumor-promoting PTX activity is tissue-independent, although, it might depend on the cancer cell line.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Melanoma/patologia , Pentoxifilina/uso terapêutico , Adenocarcinoma/patologia , Animais , Neoplasias do Colo/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Clin Cancer Res ; 9(14): 5417-22, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14614028

RESUMO

PURPOSE: The aim of the present study was to potentiate the antitumor effectiveness of photodynamic therapy (PDT). A cDNA microarray analysis was used to evaluate the gene expression pattern after Photofrin-mediated PDT to find more effective combination treatment with PDT and inhibitor(s) of the identified gene product(s) overexpressed in tumor cells. EXPERIMENTAL DESIGN: Atlas Mouse Stress Array was used to compare the expression profile of control and PDT-treated C-26 cells. The microarray results have been confirmed using Western blotting. Cytostatic/cytotoxic in vitro assay as well as in vivo tumor models were used to investigate the antitumor effectiveness of PDT in combination with cyclooxygenase (COX) 2 inhibitors. RESULTS: PDT induced the expression of 5 of 140 stress-related genes. One of these genes encodes for COX-2, an enzyme important in the tumor progression. Inhibition of COX-2 in vitro with NS-398, rofecoxib, or nimesulide, or before PDT with nimesulide did not influence the therapeutic efficacy of the treatment. Administration of a selective COX-2 inhibitor after PDT produced potentiated antitumor effects leading to complete responses in the majority of treated animals. CONCLUSIONS: COX-2 inhibitors do not sensitize tumor cells to PDT-mediated killing. However, these drugs can be used to potentiate the antitumor effectiveness of this treatment regimen when administered after tumor illumination.


Assuntos
Neoplasias do Colo/terapia , Inibidores de Ciclo-Oxigenase/farmacologia , Perfilação da Expressão Gênica , Isoenzimas/antagonistas & inibidores , Fotoquimioterapia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Terapia Combinada , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Éter de Diematoporfirina/metabolismo , Humanos , Luz , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Prostaglandina-Endoperóxido Sintases , Taxa de Sobrevida , Células Tumorais Cultivadas
16.
Oncol Rep ; 10(6): 1805-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14534700

RESUMO

Pentoxifylline is commonly used in the treatment of peripheral vascular diseases. It improves microcirculatory flow and tissue perfusion. Moreover, pentoxifylline displays some immunomodulatory properties that presumably might affect the anticancer response. Therefore, the aim of the present study was to evaluate the influence of pentoxifylline on tumor development. Balb/c mice were injected with murine colon adenocarcinoma C-26 cells intravenously, into the vena portae, and divided into two groups. Mice from the experimental groups received daily intraperitoneal injections of pentoxifylline (30 mg/kg) while the controls were injected with 0.9% NaCl. Two weeks after C-26 cell inoculation mice were sacrificed and autopsy was performed. It was found that the livers of control animals revealed only several small tumor foci, whereas the livers of pentoxifylline-treated mice displayed numerous cancerous outgrowths. The mean liver weight in pentoxifylline group was 2.21+/-0.62 g as compared to 1.36+/-0.15 g in controls (P=0.004). Moreover, the influence of pentoxifylline on murine and human cell line proliferation in vitro was evaluated. It has been observed that pentoxifylline, at pharmacologically achievable concentrations, stimulated the proliferation of murine (C-26) and human (CaSki, U-937) cell lines. However, it did not stimulate human melanoma (WM-35) cell proliferation. Since there has been no evidence so far that pentoxifylline may promote tumor progression, it is still considered to be a safe drug. Moreover, some beneficial properties of pentoxifylline, which could be useful in cancer treatment, have been reported and a few clinical trials with oncological patients have been performed. Surprisingly, our study revealed that pentoxifylline significantly promoted C-26-derived metastatic tumor growth in liver. Although this model might be unique in its sensitivity to tumor-promoting effects of pentoxifylline, it cannot be excluded that similar effects might occur in some cases of tumors developing in humans. Such effects could be relevant, since the stimulatory influence of pentoxifylline on proliferation does not appear to be species- or tissue-specific.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Sequestradores de Radicais Livres/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Pentoxifilina/farmacologia , Adenocarcinoma/patologia , Animais , Divisão Celular , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Corantes/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Fígado/patologia , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Sais de Tetrazólio/farmacologia , Tiazóis/farmacologia , Células U937
17.
Int J Mol Med ; 12(3): 279-87, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12883642

RESUMO

In the 70s, several new, both peptidyl and non-peptidyl, derivatives that stimulate and amplify pulsatile growth hormone (GH) secretion, independently from growth hormone releasing hormone (GHRH), were synthesized. The family of these molecules have been named growth hormone secretagogues (GHSs). The subsequent discovery of the natural receptor for GHSs (GHS-R) suggested existence of a new regulatory system, participating in GH secretion control. Three years later a natural ligand for GHS-R was identified and was designated 'ghrelin'. Subsequently, it has been found that ghrelin exerts pleiotropic activity. It influences not only GH release but also food intake, function of gastrointestinal tract and cardiovascular system, sleep patterns as well as cancer proliferation. The current knowledge on ghrelin, its structure and function, is reviewed in this article.


Assuntos
Encéfalo/metabolismo , Sistema Digestório/metabolismo , Hormônio do Crescimento/metabolismo , Hormônios Peptídicos/fisiologia , Sistema Cardiovascular/metabolismo , Divisão Celular/fisiologia , Ingestão de Alimentos/fisiologia , Grelina , Neoplasias/metabolismo , Hormônios Peptídicos/sangue , Hormônios Peptídicos/química
18.
Oncol Rep ; 10(4): 839-43, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12792732

RESUMO

Pentoxifylline (PTX), a methylxanthine derivative, is commonly used in patients suffering from peripheral vascular diseases. Besides its well-known hemorheological properties PTX has been found to decrease the secretion of some inflammatory cytokines such as TNF, IL-12 and IFN-gamma. PTX also suppresses lymphocyte cytotoxicity, affecting mainly the perforin-dependent pathway. We investigated the influence of PTX on the splenocyte cytotoxicity and leukocyte infiltration in a murine colon adenocarcinoma model. We observed that PTX decreased the cytotoxic activity of isolated splenocytes against C-26 cells and reduced the inflammatory cell infiltration in the peritumoral tissue. Thus, our results strongly suggest the necessity of further studies concerning the safety of PTX use, especially in elderly patients or patients with already diagnosed cancer.


Assuntos
Adenocarcinoma/patologia , Neoplasias do Colo/patologia , Citotoxicidade Imunológica/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Pentoxifilina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Adenocarcinoma/imunologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/imunologia , Feminino , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Baço/imunologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA