Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Mol Cell Endocrinol ; 538: 111454, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34520813

RESUMO

Hypothyroidism is a protective factor against breast cancer but long-term exposure or overdoses of thyroid replacement therapy with thyroxine (T4) may increase breast cancer risk. OBJECTIVE: to study, in vivo and in vitro, the effects of T4 on the proliferation and apoptosis of mammary tumors of hypo- and euthyroid rats, and the possible mechanisms involved in these effects. MATERIAL AND METHODS: Female Sprague-Dawley rats were treated with a single dose of dimethylbenzathracene (15 mg/rat) at 55 days of age and were divided into three groups: hypothyroidism (HypoT; 0.01% 6-N-propyl-2-thiouracil -PTU- in drinking water, n = 20), hypothyroidism treated with T4 (HypoT + T4; 0.01% PTU in drinking water and 0.25 mg/kg/day T4 via sc; n = 20) and EUT (untreated control, n = 20). At sacrifice, tumor explants from HypoT and EUT rats were obtained and treated either with 10-10 M T4 in DMEM/F12 without phenol red with 1% Charcoalized Fetal Bovine Serum or DMEM/F12 only for 15 min to evaluate intracellular signaling pathways associated with T4, and 24 h to evaluate changes in the expression of hormone receptors and proteins related to apoptosis and proliferation by immunohistochemistry and Western Blot. RESULTS: In vivo, hypothyroidism retards mammary carcinogenesis but its treatment with T4 reverted the protective effects. In vitro, the proliferative and anti-apoptosis mechanisms of T4 were different regarding the thyroid status. In EUT tumors, the main signaling pathway involved was the cross-talk with other receptors, such as ERα, PgR, and HER2. In HypoT tumors, the non-genomic signaling pathway of T4 was the chief mechanism involved since αvß3 integrin, HER2, ß-catenin and, downstream, PI3K/AKT and ERK signaling pathways were activated. CONCLUSION: T4 can regulate mammary carcinogenesis by mainly activating its non-genomic signaling pathway and by interacting with other hormone or growth factor pathways endorsing that overdoses of thyroid replacement therapy with T4 can increase the risk of breast cancer.


Assuntos
Antracenos/efeitos adversos , Hipotireoidismo/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Piperidinas/efeitos adversos , Propiltiouracila/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Tiroxina/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hipotireoidismo/induzido quimicamente , Neoplasias Mamárias Experimentais/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Tiroxina/farmacologia
2.
Cell Physiol Biochem ; 30(1): 113-22, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22759960

RESUMO

BACKGROUND/AIMS: Adipose microenvironment is involved in signaling pathways that influence prostate cancer (PCa) progression. However, the role of human periprostatic adipose tissue (PPAT) from patients with benign prostatic hyperplasia (BPH) has not been studied and compared to that of PPAT from PCa patients. The aim of this paper was to investigate the influence of factors derived from both PPATs on the behavior of androgen-dependent and castration resistant PCa cells. METHODS: PPAT conditioned media (CM) were obtained from tissue samples from patients with clinically primary PCa (TPPAT) or BPH (BPPAT). Cell adhesion, proliferation, migration and metalloproteinase expression were evaluated following exposure of LNCaP (androgen dependent) and PC3 (androgen independent) prostate cancer cell lines to BPPAT or TPPAT CM. RESULTS: Proliferation or motility of LNCaP or PC3 cells were not significantly affected by TPPAT or BPPAT CM. The number of LNCaP but not PC3 cells attached to components of TPPAT CM significantly decreased compared to cells attached to BPPAT CM. PPAT produced and released pro-MMP-9. Zymograms demonstrated that TPPAT CM induced a significant increase in pro-MMP-9 activity compared to BPPAT CM in LNCaP cells but not in PC3 cells. CONCLUSIONS: We conclude that TPPAT released factors, such as pro-MMP-9, could induce the invasive capacity of LNCaP cells and speculate that PPAT derived factors could, in the early stages of prostate cancer, modulate disease progression.


Assuntos
Gordura Intra-Abdominal/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Próstata/patologia , Hiperplasia Prostática/patologia , Neoplasias da Próstata/patologia , Idoso , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Meios de Cultivo Condicionados , Humanos , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Microambiente Tumoral
3.
Mol Med Rep ; 3(3): 433-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21472258

RESUMO

Stromal tissue regulates the development and differentiation of breast epithelial cells, with adipocytes being the main stromal cell type. The aim of the present study was to evaluate the effect of adipocyte differentiation on proliferation and migration, as well as to assess the activity of heparanase and metalloproteinase-9 (MMP-9), in normal (NMuMG) and tumoral (LM3) murine breast epithelial cells. NMuMG and LM3 cells were grown on irradiated 3T3-L1 cells (stromal support, SS) at various degrees of differentiation [preadipocytes (preA), poorly differentiated adipocytes (pDA) and mature adipocytes (MA)] and/or were incubated in the presence of conditioned medium (CM) derived from each of these three types of differentiated cells. Cells grown on a plastic support or in fresh medium served as the controls. Cell proliferation was measured with a commercial colorimetric kit, and the motility of the epithelial cells was evaluated by means of a wound-healing assay. Heparanase activity was assessed by quantifying heparin degradation, and the expression of MMP-9 was determined using Western blotting. The results indicate that cell proliferation was increased after 24 and 48 h in the NMuMG and LM3 cells grown on preA, pDA and MA SS. In the NMuMG cells cultured on SS in the presence of all three types of CM, proliferation was enhanced. LM3 cell migration was increased in the presence of all three types of CM and in cells grown on preA SS. Heparanase activity was increased in the NMuMG cells incubated with all three types of CM, and in the LM3 cells incubated with the CM from pDA and MA. Both the NMuMG and LM3 cell lines presented basal expression of MMP-9; however, a significant increase in MMP-9 expression was observed in the LM3 cells incubated with each of the three types of CM. In conclusion, adipocyte differentiation influences normal and tumoral breast epithelial cell proliferation and migration. Heparanase and MMP-9 appear to be involved in this regulation. The experimental model presented in this study is in keeping with the characteristics of the physiological environment of breast epithelial cells, in terms of both the soluble and insoluble factors present and the stromal structure per se.

4.
Brain Res ; 1230: 320-33, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18675791

RESUMO

Shiga toxin (Stx) from enterohemorrhagic Escherichia coli (STEC) is the main cause of hemorrhagic colitis which may derive into Hemolytic Uremic Syndrome (HUS) and acute encephalopathy, one of the major risk factors for infant death caused by the toxin. We have previously demonstrated that intracerebroventricular administration of Stx2 causes neuronal death and glial cell damage in rat brains. In the present work, we observed that the intracerebroventricular administration of Stx2 increased the expression of glial fibrillary acidic protein (GFAP) leading to astrogliosis. Confocal microscopy showed reactive astrocytes in contact with Stx2-containing neurons. Immunocolocalization of increased GFAP and Stx2 in astrocytes was also observed. This insult in the brain was correlated with changes in the expression and activity of neuronal nitric oxide synthase (nNOS) by using the NADPH-diaphorase histochemical technique (NADPH-d HT). A significant decrease in NOS/NADPH-d-positive neurons and NOS/NADPH-d activity was observed in cerebral cortex and striatum, whereas an opposite effect was found in the hypothalamic paraventricular nucleus. We concluded that the i.c.v. administration of Stx2 promotes a typical pattern of brain injury showing reactive astrocytes and an alteration in the number and activity of nNOS/NADPH-d. According to the functional state of nNOS/NADPH-d and to brain cell morphology data, it could be inferred that the i.c.v. administration of Stx2 leads to either a neurodegenerative or a neuroprotective mechanism in the affected brain areas. The present animal model resembles the encephalopathy developed in Hemolytic Uremic Syndrome (HUS) patients by STEC intoxication.


Assuntos
Química Encefálica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/biossíntese , Óxido Nítrico Sintase Tipo I/biossíntese , Toxina Shiga II/toxicidade , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/genética , Imuno-Histoquímica , Injeções Intraventriculares , Masculino , Microscopia Confocal , Microscopia Eletrônica de Transmissão , NADPH Desidrogenase/metabolismo , Neostriado/fisiologia , Óxido Nítrico Sintase Tipo I/genética , Células Piramidais/efeitos dos fármacos , Células Piramidais/enzimologia , Células Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Toxina Shiga II/administração & dosagem , Toxina Shiga II/isolamento & purificação
5.
Brain Res ; 1161: 106-15, 2007 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-17610852

RESUMO

Shiga toxin (Stx) from enterohemorrhagic Escherichia coli (STEC) is the main cause of hemorrhagic colitis which may derive to hemolytic-uremic syndrome (HUS). HUS is characterized by acute renal failure, thrombocytopenia and microangiopathic hemolytic anemia. Mortality in the acute stage has been lower than 5% of total affected argentine children with endemic HUS. Common signs of severe CNS involvement leading to death included seizures, alteration of consciousness, hemiparesis, visual disturbances, and brainstem symptoms. The main purpose of the present work was to study the direct involvement of Stx2 in brain cells by intracerebroventricular (i.c.v.) administration of Stx2. Immunodetection of Stx2 was confirmed by immunoelectron cytochemistry in different subsets and compartments of affected caudate putamen cells of corpus striatum. Transmission electron microscopy (TEM) studies revealed apoptotic neurons, glial ultrastructural alterations and demyelinated fibers. The i.c.v. microinfusion was applied for the first time in rats to demonstrate the direct action of Stx2 in neurons and glial cells. The toxin may affect brain neuroglial cells without the involvement of proinflammatory or systemic neurotoxic elements.


Assuntos
Corpo Estriado/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/ultraestrutura , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Toxina Shiga II/administração & dosagem , Análise de Variância , Animais , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/metabolismo , Injeções Intraventriculares/métodos , Masculino , Microscopia Imunoeletrônica/métodos , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Toxina Shiga II/metabolismo
6.
Microbes Infect ; 8(2): 410-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16242986

RESUMO

Shiga toxin-producing Escherichia coli produces watery diarrhea, hemorrhagic colitis and hemolytic-uremic syndrome (HUS). In Argentina, HUS is the most common cause of acute renal failure in children. The purpose of the present study was to examine the cytotoxicity of Stx type 2 (Stx2 holotoxin) and its B subunit (Stx2 B subunit) on human renal tubular epithelial cells (HRTEC), in the presence and absence of inflammatory factors. Cell morphology, cell viability, protein synthesis and apoptosis were measured. HRTEC are sensitive to both Stx2 holotoxin and Stx2 B subunit in a dose- and time-dependent manner. IL-1, LPS and butyrate but not TNF, IL-6 and IL-8, increased the Stx mediated cytotoxicity. The effects of Stx2 B subunit appear at doses higher than those used for Stx2 holotoxin. Although the physiological importance of these effects is not clear, it is important to be aware of any potentially toxic activity in the B subunit, given that it has been proposed for use in a vaccine.


Assuntos
Células Epiteliais/efeitos dos fármacos , Túbulos Renais/citologia , Túbulos Renais/efeitos dos fármacos , Subunidades Proteicas/toxicidade , Toxina Shiga II/toxicidade , Adulto , Apoptose , Butiratos/farmacologia , Células Cultivadas , Escherichia coli/patogenicidade , Humanos , Interleucina-1/farmacologia , Lipopolissacarídeos/farmacologia
7.
Medicina (B Aires) ; 66 Suppl 3: 11-5, 2006.
Artigo em Espanhol | MEDLINE | ID: mdl-17354471

RESUMO

In the last years, infection associated with Shiga toxin-producing Escherichia coli (STEC) and subsequent Hemolitic-Uremic Syndrome (HUS) became relevant as a public health since it was considered as one of the most important emergent patogen present in the food contaminated by cattle feces. STEC infection may be asymptomatic or begins with a watery diarrhea that may or may not progress to bloody diarrhea (hemorrhagic colitis) and HUS. In Argentina, HUS is the most common pediatric cause of acute renal insufficiency and the second cause of chronic renal failure. Up to now, STEC infection lacks of known effective treatment strategies that diminish risk of progression to HUS. The mechanisms by which Shiga toxin (Stx) induce HUS may help to find strategies to prevent or ameliorate HUS. In this article, recent progress that has contributed to understanding the disease pathogenesis of STEC is reviewed. New strategies to prevent further uptake of Shiga from the gut, either during the diarrheal phase or once HUS has developed are discussed.


Assuntos
Infecções por Escherichia coli/microbiologia , Síndrome Hemolítico-Urêmica/microbiologia , Toxinas Shiga/metabolismo , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/microbiologia , Escherichia coli/metabolismo , Escherichia coli/patogenicidade , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/fisiopatologia , Vacinas contra Escherichia coli/administração & dosagem , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/fisiopatologia , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Rim/metabolismo , Rim/microbiologia , Toxinas Shiga/antagonistas & inibidores
8.
Medicina (B Aires) ; 65(2): 147-50, 2005.
Artigo em Espanhol | MEDLINE | ID: mdl-16075810

RESUMO

Shiga toxin (Stx)-producing E. coli causing watery diarrhea, hemorrhagic colitis and hemolytic-uremic syndrome (HUS). In Argentina, HUS is the most common cause of acute renal failure in children. The purpose of the present study was to examine the cytotoxicity of Stx type 2 (Stx2) and its B subunit (Stx2B) on human renal tubular epithelial cells (HRTEC), in the presence and absence of inflammatory factors. Cytotoxic effects were assessed in terms of functionality of the epithelium, histological damage, cell viability, protein synthesis and cellular apoptosis. Results show that Stx2 regulates the passage of water through the HRTEC within an incubation period of 1h. Within longer periods, up to 72 hours, the study of morphology, viability, protein synthesis and apoptosis shows that HRTEC were sensitive to the cytotoxic action of Stx2 and Stx2B in a dose- and time-dependent manner. These effects were potentiated by lipopolysaccharides (LPS), IL-1beta, and butirate.


Assuntos
Células Epiteliais/efeitos dos fármacos , Síndrome Hemolítico-Urêmica/fisiopatologia , Túbulos Renais/citologia , Toxina Shiga II/toxicidade , Adulto , Apoptose , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/patologia , Escherichia coli/patogenicidade , Síndrome Hemolítico-Urêmica/complicações , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Subunidades Proteicas/toxicidade , Insuficiência Renal/etiologia
9.
Medicina [B Aires] ; 65(2): 147-50, 2005.
Artigo em Espanhol | BINACIS | ID: bin-38323

RESUMO

Shiga toxin (Stx)-producing E. coli causing watery diarrhea, hemorrhagic colitis and hemolytic-uremic syndrome (HUS). In Argentina, HUS is the most common cause of acute renal failure in children. The purpose of the present study was to examine the cytotoxicity of Stx type 2 (Stx2) and its B subunit (Stx2B) on human renal tubular epithelial cells (HRTEC), in the presence and absence of inflammatory factors. Cytotoxic effects were assessed in terms of functionality of the epithelium, histological damage, cell viability, protein synthesis and cellular apoptosis. Results show that Stx2 regulates the passage of water through the HRTEC within an incubation period of 1h. Within longer periods, up to 72 hours, the study of morphology, viability, protein synthesis and apoptosis shows that HRTEC were sensitive to the cytotoxic action of Stx2 and Stx2B in a dose- and time-dependent manner. These effects were potentiated by lipopolysaccharides (LPS), IL-1beta, and butirate.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...