Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Front Mol Biosci ; 9: 842149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35281256

RESUMO

The family of the human small Heat Shock Proteins (HSPBs) consists of ten members of chaperones (HSPB1-HSPB10), characterized by a low molecular weight and capable of dimerization and oligomerization forming large homo- or hetero-complexes. All HSPBs possess a highly conserved centrally located α-crystallin domain and poorly conserved N- and C-terminal domains. The main feature of HSPBs is to exert cytoprotective functions by preserving proteostasis, assuring the structural maintenance of the cytoskeleton and acting in response to cellular stresses and apoptosis. HSPBs take part in cell homeostasis by acting as holdases, which is the ability to interact with a substrate preventing its aggregation. In addition, HSPBs cooperate in substrates refolding driven by other chaperones or, alternatively, promote substrate routing to degradation. Notably, while some HSPBs are ubiquitously expressed, others show peculiar tissue-specific expression. Cardiac muscle, skeletal muscle and neurons show high expression levels for a wide variety of HSPBs. Indeed, most of the mutations identified in HSPBs are associated to cardiomyopathies, myopathies, and motor neuropathies. Instead, mutations in HSPB4 and HSPB5, which are also expressed in lens, have been associated with cataract. Mutations of HSPBs family members encompass base substitutions, insertions, and deletions, resulting in single amino acid substitutions or in the generation of truncated or elongated proteins. This review will provide an updated overview of disease-related mutations in HSPBs focusing on the structural and biochemical effects of mutations and their functional consequences.

2.
Braz J Med Biol Res ; 53(11): e10067, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33053110

RESUMO

RU486 (mifepristone), a glucocorticoid and progesterone receptor antagonist, has been reported to exert antiproliferative effects on tumor cells. Experiments were performed to analyze the effects of RU486 on the proliferation of the human neuroblastoma, both in vitro and in vivo, using the human neuroblastoma SK-N-SH cell line. The exposure in vitro of SK-N-SH cells to RU486 revealed a dose-dependent inhibition of 3H-thymidine incorporation due to a rapid but persistent inhibition of MAPKinase activity and ERK phosphorylation. A significant decrease of SK-N-SH cell number was evident after 3, 6, and 9 days of treatment (up to 40% inhibition), without evident cell death. The inhibitory effect exerted by RU486 was not reversed by the treatment of the cells with dexamethasone or progesterone. Moreover, RU486 induced a shift in SK-N-SH cell phenotypes, with an almost complete disappearance of the neuronal-like and a prevalence of the epithelial-like cell subtypes. Finally, the treatment with RU486 of nude mice carrying a SK-N-SH cell xenograft induced a strong inhibition (up to 80%) of tumor growth. These results indicated a clear effect of RU486 on the growth of SK-N-SH neuroblastoma cells that does not seem to be mediated through the classical steroid receptors. RU486 acted mainly on the more aggressive component of the SK-N-SH cell line and its effect in vivo was achieved at a concentration already used to inhibit oocyte implantation.


Assuntos
Neuroblastoma , Animais , Glucocorticoides , Humanos , Camundongos , Camundongos Nus , Mifepristona/farmacologia , Neuroblastoma/tratamento farmacológico , Progesterona
3.
Braz. j. med. biol. res ; 53(11): e10067, 2020. graf
Artigo em Inglês | LILACS, Coleciona SUS | ID: biblio-1132493

RESUMO

RU486 (mifepristone), a glucocorticoid and progesterone receptor antagonist, has been reported to exert antiproliferative effects on tumor cells. Experiments were performed to analyze the effects of RU486 on the proliferation of the human neuroblastoma, both in vitro and in vivo, using the human neuroblastoma SK-N-SH cell line. The exposure in vitro of SK-N-SH cells to RU486 revealed a dose-dependent inhibition of 3H-thymidine incorporation due to a rapid but persistent inhibition of MAPKinase activity and ERK phosphorylation. A significant decrease of SK-N-SH cell number was evident after 3, 6, and 9 days of treatment (up to 40% inhibition), without evident cell death. The inhibitory effect exerted by RU486 was not reversed by the treatment of the cells with dexamethasone or progesterone. Moreover, RU486 induced a shift in SK-N-SH cell phenotypes, with an almost complete disappearance of the neuronal-like and a prevalence of the epithelial-like cell subtypes. Finally, the treatment with RU486 of nude mice carrying a SK-N-SH cell xenograft induced a strong inhibition (up to 80%) of tumor growth. These results indicated a clear effect of RU486 on the growth of SK-N-SH neuroblastoma cells that does not seem to be mediated through the classical steroid receptors. RU486 acted mainly on the more aggressive component of the SK-N-SH cell line and its effect in vivo was achieved at a concentration already used to inhibit oocyte implantation.


Assuntos
Humanos , Animais , Coelhos , Neuroblastoma/tratamento farmacológico , Progesterona , Mifepristona/farmacologia , Glucocorticoides , Camundongos Nus
4.
J Endocrinol Invest ; 36(9): 775-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23765505

RESUMO

Adrenal glucocorticoids (Gc) are among the most significant hormones in the mammalian organisms; these steroids may reach and penetrate all tissues where they interact with cytoplasmic/nuclear receptors, through which they exert multiple and very multifaceted actions. The effects of physiological concentrations of Gc on brain functions have not been completely clarified, even though Gc are recognized to influence behavioral responses, emotions, cognitive processes and to take part in the neuroendocrine control of body homeostasis. Developmental programming effects of Gc in animal models and humans have been proposed. Actually, pre-natal stress, or exposure to high Gc levels, would somehow affect neuronal developmental events in some structure and this can lead to central nervous system altered functions, as the impairment of neuroendocrine activities, cognitive processes, sleep and mood disorders. Interestingly, it has been observed that these abnormalities may not be limited to the first directly exposed individuals but transmissible across generations. The establishment of animal models with localized pre-natal glucocorticoid receptors deficiency led to the accumulation of data on the possible roles of these hormones on development of the central and peripheral nervous system. The most recent findings on the effects of Gc on neuroblast development, with particular attention to neuronal migration, will be presented.


Assuntos
Glucocorticoides/fisiologia , Sistema Nervoso/embriologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Proteínas de Ligação a Calmodulina/genética , Movimento Celular , Sistema Nervoso Central/embriologia , Humanos , Erros Inatos do Metabolismo/fisiopatologia , Sistema Nervoso/efeitos dos fármacos , Neurônios/fisiologia , Sistema Nervoso Periférico/embriologia , Receptores de Glucocorticoides/biossíntese , Receptores de Glucocorticoides/deficiência
5.
Int J Oncol ; 32(1): 185-91, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18097558

RESUMO

Urokinase-type plasminogen activator (uPA) and its specific membrane receptor (uPAR) control extracellular matrix proteolysis, cell migration, invasion and cell growth in several cancers. The uPAR released from human cancers is detected in blood as soluble uPAR (suPAR). No information is available on the mechanism(s) of action of suPAR on prostate cancer (PCa) cell growth and invasion. In order to clarify this issue, we tested the effect of a treatment with the human recombinant suPAR (comprising amino acids l-303) on the proliferation, migration and invasion of DU145 cells, a PCa cell line expressing a potent autocrine uPA-uPAR signalling system. The results indicate that suPAR significantly inhibits cell growth, promotes apoptosis and decreases both migration and Matrigel invasion of DU145 cells. The mechanism of action of suPAR seems to be linked to a decrease of ERK and FAK activation. Cleavage of suPAR by chymotripsin reverses these effects. When added to the uPA-negative LNCaP cells, suPAR was ineffective; on the contrary, when LNCaP cells were cultured on fibronectin-coated plates in order to stimulate uPA expression, suPAR significantly decreased cell proliferation. In conclusion, our data suggest that suPAR can function as a potent molecule scavenger for uPA in human PCa cells characterized by high levels of uPA/uPAR as in DU145 cells, while it is ineffective in uPA-deficient LNCaP cells. The molecular mechanism(s) through which suPAR participates in the control of PCa progression may bear relevance for the long-term goal to identify new therapeutic targets aimed at silencing tumours in vivo.


Assuntos
Neoplasias da Próstata/patologia , Receptores de Superfície Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Masculino , Invasividade Neoplásica , Fosforilação , Neoplasias da Próstata/terapia , Receptores de Ativador de Plasminogênio Tipo Uroquinase
6.
Braz J Med Biol Res ; 39(9): 1233-40, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16981049

RESUMO

Glucocorticoids (Gc) influence the differentiation of neural crest-derived cells such as those composing sympathoadrenal tumors like pheochromocytomas, as well as neuroblastomas and gangliomas. In order to obtain further information on the effects of Gc on cells evolving from the neural crest, we have used the human neuroblastoma cell line SK-N-SH to analyze: 1) the presence and the binding characteristics of Gc receptors in these cells, 2) the effect of dexamethasone (Dex) on the migration of SK-N-SH cells, and 3) the effect of Dex on the organization of the cytoskeleton of SK-N-SH cells. We show that: 1) receptors that bind [(3)H]-Dex with high affinity and high capacity (Kd of 9.6 nM, Bmax of 47 fmol/mg cytosolic protein, corresponding to 28,303 sites/cell) are present in cytosolic preparations of SK-N-SH cells, and 2) treatment with Dex (in the range of 10 nM to 1 microM) has an inhibitory effect (from 100% to 74 and 43%, respectively) on the chemotaxis of SK-N-SH cells elicited by fetal bovine serum. This inhibition is completely reversed by the Gc receptor antagonist RU486 (1 microM), and 3) as demonstrated by fluorescent phalloidin-actin detection, the effect of Dex (100 nM) on SK-N-SH cell migration is accompanied by modifications of the cytoskeleton organization that appear with stress fibers. These modifications did not take place in the presence of 1 microM RU486. The present data demonstrate for the first time that Dex affects the migration of neuroblastoma cells as well as their cytoskeleton organization by interacting with specific receptors. These findings provide new insights on the mechanism(s) of action of Gc on cells originating in the neural crest.


Assuntos
Movimento Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Neuroblastoma/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Forma Celular , Quimiotaxia , Humanos , Neuroblastoma/química , Receptores de Glucocorticoides/análise
7.
Braz. j. med. biol. res ; 39(9): 1233-1240, Sept. 2006. ilus, graf
Artigo em Inglês | LILACS | ID: lil-435420

RESUMO

Glucocorticoids (Gc) influence the differentiation of neural crest-derived cells such as those composing sympathoadrenal tumors like pheochromocytomas, as well as neuroblastomas and gangliomas. In order to obtain further information on the effects of Gc on cells evolving from the neural crest, we have used the human neuroblastoma cell line SK-N-SH to analyze: 1) the presence and the binding characteristics of Gc receptors in these cells, 2) the effect of dexamethasone (Dex) on the migration of SK-N-SH cells, and 3) the effect of Dex on the organization of the cytoskeleton of SK-N-SH cells. We show that: 1) receptors that bind [³H]-Dex with high affinity and high capacity (Kd of 9.6 nM, Bmax of 47 fmol/mg cytosolic protein, corresponding to 28,303 sites/cell) are present in cytosolic preparations of SK-N-SH cells, and 2) treatment with Dex (in the range of 10 nM to 1 æM) has an inhibitory effect (from 100 percent to 74 and 43 percent, respectively) on the chemotaxis of SK-N-SH cells elicited by fetal bovine serum. This inhibition is completely reversed by the Gc receptor antagonist RU486 (1 æM), and 3) as demonstrated by fluorescent phalloidin-actin detection, the effect of Dex (100 nM) on SK-N-SH cell migration is accompanied by modifications of the cytoskeleton organization that appear with stress fibers. These modifications did not take place in the presence of 1 æM RU486. The present data demonstrate for the first time that Dex affects the migration of neuroblastoma cells as well as their cytoskeleton organization by interacting with specific receptors. These findings provide new insights on the mechanism(s) of action of Gc on cells originating in the neural crest.


Assuntos
Humanos , Movimento Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Neuroblastoma/patologia , Forma Celular , Quimiotaxia , Linhagem Celular Tumoral/efeitos dos fármacos , Neuroblastoma/química , Receptores de Glucocorticoides/análise
8.
Oncol Rep ; 15(2): 393-400, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16391860

RESUMO

Prostate cancer (PCa) growth initially depends on circulating androgens. Gonadotropin-releasing hormone (GnRH) agonists are currently used for the treatment of PCa. However, after an initial responsiveness to hormonal deprivation, PCa progresses and metastasizes. Recently, also GnRH antagonists have been used for clinical trials in patients with PCa and the results seem promising. The components of the plasminogen activator (PA) system (urokinase-type PA, uPA; PA inhibitors, PAI-1/2; uPA receptor, uPAR) have been implicated in the local degradation of the extracellular matrix (ECM) and PCa progression. The aim of this study was to test the possible effects of the treatment with an agonist (Leuprolide, GnRH-A) and an antagonist (Cetrorelix, GnRH-ANT) of GnRH on the expression and activity of uPA and PAI-1 in the conditioned media of DU145 and PC3, two PCa androgen-independent cell lines. The involvement of the PA system in the control of cellular migration was also investigated. The results obtained in DU145 and PC3 cells show that both GnRH-A and GnRH-ANT: i) inhibit cell proliferation; ii) significantly decrease the enzymatic activity and the secretion of uPA; iii) significantly increase the protein levels of PAI-1; iv) induce a significant decrease of the migratory and invasion PCa capabilities. This study suggests that GnRH analogues exhibit not only an antiproliferative effect, but also an anti-metastatic action exerted through the inhibition of the activity of PA system and might provide a rational basis for the development of clinical strategies for those tumours that progress towards an androgen-independent condition characterized by a higher metastatic potential.


Assuntos
Antineoplásicos Hormonais/farmacologia , Antagonistas de Hormônios/farmacologia , Invasividade Neoplásica/fisiopatologia , Ativadores de Plasminogênio/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Leuprolida/farmacologia , Masculino
9.
J Exp Biol ; 206(Pt 10): 1717-26, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12682103

RESUMO

European eels were exposed for 6 weeks to water CO(2) partial pressures (P(CO)(2)) from ambient (approx. 0.8 mmHg), through 15+/-1 mmHg and 30+/-1 mmHg to 45+/-1 mmHg in water with a total hardness of 240 mg l(-1) as CaCO(3), pH 8.2, at 23+/-1 degrees C. Arterial plasma P(CO)(2) equilibrated at approximately 2 mmHg above water P(CO)(2) in all groups, and plasma bicarbonate accumulated up to 72 mmol l(-1) in the group at a water P(CO)(2) of 45 mmHg. This was associated with an equimolar loss of plasma Cl(-), which declined to 71 mmol l(-1) at the highest water P(CO)(2). Despite this, extracellular acid-base compensation was incomplete; all hypercapnic groups tolerated chronic extracellular acidoses and reductions in arterial blood O(2) content (Ca(O)(2)), of progressive severity with increasing P(CO)(2). All hypercapnic eels, however, regulated the intracellular pH of heart and white muscle to the same levels as normocapnic animals. Hypercapnia had no effect on such indicators of stress as plasma catecholamine or cortisol levels, plasma osmolality or standard metabolic rate. Furthermore, although Ca(O)(2) was reduced by approximately 50% at the highest P(CO)(2), there was no effect of hypercapnia on the eels' tolerance of hypoxia, aerobic metabolic scope or sustained swimming performance. The results indicate that, at the levels tested, chronic hypercapnia was not a physiological stress for the eel, which can tolerate extracellular acidosis and extremely low Cl(-) levels while compensating tissue intracellular pH, and which can meet the O(2) requirements of routine and active metabolism despite profound hypoxaemia.


Assuntos
Anguilla/metabolismo , Hipercapnia/metabolismo , Equilíbrio Ácido-Base , Aerobiose , Anguilla/sangue , Animais , Epinefrina/sangue , Hidrocortisona/sangue , Hipercapnia/sangue , Hipóxia/sangue , Hipóxia/metabolismo , Modelos Biológicos , Norepinefrina/sangue , Oxigênio/sangue , Esforço Físico/fisiologia
10.
Pharmacol Res ; 44(4): 265-80, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11592860

RESUMO

Fish represent the oldest and most diverse classes of vertebrates, comprising around the 48% of the known member species in the subphylum Vertebrata. There are many scientific fields that use fish as models in research, including respiratory and cardiovascular research, cell culture, ecotoxicology, ageing, pharmacological and genetic studies.


Assuntos
Modelos Animais de Doenças , Peixes/fisiologia , Modelos Animais , Projetos de Pesquisa , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Testes de Carcinogenicidade/métodos , Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/fisiopatologia , Técnicas de Cultura de Células/métodos , Substâncias Perigosas/toxicidade , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/fisiologia , Vertebrados/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...