Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 23(6): 1410-1420, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28894299

RESUMO

Dehydroepiandrosterone (DHEA) is the most abundant circulating steroid hormone in humans, produced by the adrenals, the gonads and the brain. DHEA was previously shown to bind to the nerve growth factor receptor, tropomyosin-related kinase A (TrkA), and to thereby exert neuroprotective effects. Here we show that DHEA reduces microglia-mediated inflammation in an acute lipopolysaccharide-induced neuro-inflammation model in mice and in cultured microglia in vitro. DHEA regulates microglial inflammatory responses through phosphorylation of TrkA and subsequent activation of a pathway involving Akt1/Akt2 and cAMP response element-binding protein. The latter induces the expression of the histone 3 lysine 27 (H3K27) demethylase Jumonji d3 (Jmjd3), which thereby controls the expression of inflammation-related genes and microglial polarization. Together, our data indicate that DHEA-activated TrkA signaling is a potent regulator of microglia-mediated inflammation in a Jmjd3-dependent manner, thereby providing the platform for potential future therapeutic interventions in neuro-inflammatory pathologies.


Assuntos
Desidroepiandrosterona/farmacologia , Inflamação/metabolismo , Microglia/efeitos dos fármacos , Animais , Proteína de Ligação a CREB/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor trkA/efeitos dos fármacos , Receptores de Fator de Crescimento Neural/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
2.
Data Brief ; 4: 636-40, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26401519

RESUMO

This article contains data related to the research article entitled "Laser fabricated discontinuous anisotropic microconical substrates as a new model scaffold to control the directionality of neuronal network outgrowth" in the Biomaterials journal [1]. Scanning electron microscopy (SEM) analysis is performed to investigate whether Schwann cells and sympathetic neurons alter their morphology according to the underlying topography, comprising arrays of silicon microcones with anisotropic geometrical characteristics [1]. It is observed that although soma of sympathetic neurons always preserves its round shape, this is not the case for Schwann cells that become highly polarized in high roughness microconical substrates.

3.
Biomaterials ; 67: 115-28, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26210178

RESUMO

Patterning of neuronal outgrowth in vitro is important in tissue engineering as well as for the development of neuronal interfaces with desirable characteristics. To date, this has been achieved with the aid of micro- and nanofabrication techniques giving rise to various anisotropic topographies, either in the form of continuous or discontinuous structures. In this study we propose a currently unexplored geometry of a 3D culture substrate for neuronal cell growth comprising discontinuous subcellular microstructures with anisotropic geometrical cross-section. Specifically, using laser precision 3D micro/nano fabrication techniques, silicon substrates comprising arrays of parallel oriented elliptical microcones (MCs) were fabricated to investigate whether a discontinuous geometry comprising anisotropic features at the subcellular level could influence the alignment of peripheral nervous system cell populations. It was shown that both Schwann cells and axons of sympathetic neurons were parallel oriented onto the MCs of elliptical shape, while they exhibited a random orientation onto the MCs of arbitrary shape. Notably, this topography-induced guidance effect was also observed in more complex cell culture systems, such as the organotypic culture whole dorsal root ganglia (DRG) explants. Our results suggest that a discontinuous topographical pattern could promote Schwann cell and axonal alignment, provided that it hosts anisotropic geometrical features, even though the sizes of those range at the subcellular lengthscale. The laser-patterned arrays of MCs presented here could potentially be a useful platform for patterning neurons into artificial networks, allowing the study of neuronal cells interactions under 3D ex-vivo conditions.


Assuntos
Lasers , Neurônios/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Anisotropia , Axônios/efeitos dos fármacos , Axônios/metabolismo , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Fibronectinas/metabolismo , Gânglios Espinais/citologia , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley , Células de Schwann/citologia , Células de Schwann/efeitos dos fármacos , Silício/farmacologia , Propriedades de Superfície
4.
Transl Psychiatry ; 5: e685, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26795749

RESUMO

Fingolimod (FTY720) was the first per os administered disease-modifying agent approved for the treatment of relapsing-remitting multiple sclerosis. It is thought that fingolimod modulates the immune response by activating sphingosine-1 phosphate receptor type 1 (S1P1) on lymphocytes following its in vivo phosphorylation. In addition to its immune-related effects, there is evidence that fingolimod exerts several other effects in the central nervous system, including regulation of the proliferation, survival and differentiation of various cell types and their precursors. In the present study, we have investigated the effect of fingolimod on the production of new neurons in the adult mouse hippocampus and the association of this effect with the ability for pattern separation, an established adult neurogenesis-dependent memory function. Immunofluorescence analysis after chronic administration of a physiologic dose of fingolimod (0.3 mg kg(-1)) revealed a significant increase in both the proliferation and the survival of neural progenitors in the area of dentate gyrus of hippocampus, compared with control animals. These effects were replicated in vitro, in cultures of murine hippocampal neural stem/precursor cells that express S1P1 receptor, suggesting cell-autonomous actions. The effects of fingolimod on neurogenesis were correlated to enhanced ability for context discrimination after fear conditioning. Since impairment of adult hippocampal neurogenesis and memory is a common feature of many neuropsychiatric conditions, fingolimod treatment may be beneficial in therapeutic armamentarium of these disorders.


Assuntos
Medo/efeitos dos fármacos , Cloridrato de Fingolimode/farmacologia , Hipocampo/efeitos dos fármacos , Memória/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Imunossupressores/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Cell Mol Life Sci ; 64(13): 1638-55, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17453142

RESUMO

Corticotropin-releasing factor (CRF), also termed corticotropin-releasing hormone (CRH) or corticoliberin, is the major regulator of the adaptive response to internal or external stresses. An essential component of the adaptation mechanism is the adrenal gland. CRF regulates adrenal function indirectly through the central nervous system (CNS) via the hypothalamic-pituitary-adrenal (HPA) axis and via the autonomic nervous system by way of locus coeruleus (LC) in the brain stem. Accumulating evidence suggests that CRF and its related peptides also affect the adrenals directly, i.e. not through the CNS but from within the adrenal gland where they form paracrine regulatory loops. Indeed, CRF and its related peptides, the urocortins (UCNs: UCN1, UCN2 and UCN3), their receptors CRF type 1 (CRF(1)) and 2 (CRF(2)) as well as the endogenous pseudo-receptor CRF-binding protein (CRF-BP) are all expressed in adrenal cortical, medullary chromaffin and resident immune cells. The intra-adrenal CRF-based regulatory system is complex and depends on the balance between the local concentration of CRF ligands and the availability of their receptors.


Assuntos
Glândulas Suprarrenais/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Peptídeos/metabolismo , Doenças das Glândulas Suprarrenais/metabolismo , Animais , Humanos , Sistema Imunitário/metabolismo
6.
Endocrinology ; 148(4): 1524-38, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17194738

RESUMO

Corticotropin-releasing factor (CRF) affects catecholamine production both centrally and peripherally. The aim of the present work was to examine the presence of CRF, its related peptides, and their receptors in the medulla of human and rat adrenals and their direct effect on catecholamine synthesis and secretion. CRF, urocortin I (UCN1), urocortin II (UCN2), and CRF receptor type 1 (CRF1) and 2 (CRF2) were present in human and rat adrenal medulla as well as the PC12 pheochromocytoma cells by immunocytochemistry, immunofluorescence, and RT-PCR. Exposure of dispersed human and rat adrenal chromaffin cells to CRF1 receptor agonists induced catecholamine secretion in a dose-dependent manner, an effect peaking at 30 min, whereas CRF2 receptor agonists suppressed catecholamine secretion. The respective effects were blocked by CRF1 and CRF2 antagonists. CRF peptides affected catecholamine secretion via changes of subplasmaliminal actin filament polymerization. CRF peptides also affected catecholamine synthesis. In rat chromaffin and PC12 cells, CRF1 and CRF2 agonists induced catecholamine synthesis via tyrosine hydroxylase. However, in human chromaffin cells, activation of CRF1 receptors induced tyrosine hydroxylase, whereas activation of CRF2 suppressed it. In conclusion, it appears that a complex intraadrenal CRF-UCN/CRF-receptor system exists in both human and rat adrenals controlling catecholamine secretion and synthesis.


Assuntos
Glândulas Suprarrenais/efeitos dos fármacos , Catecolaminas/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Glândulas Suprarrenais/metabolismo , Animais , Catecolaminas/biossíntese , Células Cultivadas , Células Cromafins/metabolismo , Feminino , Humanos , Células PC12 , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Distribuição Tecidual , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Urocortinas
7.
Endocrinology ; 146(8): 3309-18, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15860555

RESUMO

Adrenal cortical cells of zona reticularis produce the neuroactive steroids dehydroepiandrosterone (DHEA), its sulfate ester dehydroepiandrosterone sulfate (DHEAS), and allopregnanolone (ALLO). An interaction between zona reticularis and adrenal medulla has been postulated based on their close proximity and their interwoven borders. The aim of this paper was to examine in vitro the possible paracrine effects of these steroids on catecholamine production from adrenomedullary chromaffin cells, using an established in vitro model of chromaffin cells, the PC12 rat pheochromocytoma cell line. We have found the following: 1) DHEA, DHEAS, and ALLO increased acutely (peak effect between 10-30 min) and dose-dependently (EC50 in the nanomolar range) catecholamine levels (norepinephrine and dopamine). 2) It appears that the acute effect of these steroids involved actin depolymerization/actin filament disassembly, a fast-response cellular system regulating trafficking of catecholamine vesicles. Specifically, 10(-6) m phallacidin, an actin filament stabilizer, completely prevented steroid-induced catecholamine secretion. 3) DHEAS and ALLO, but not DHEA, also affected catecholamine synthesis. Indeed, DHEAS and ALLO increased catecholamine levels at 24 h, an effect blocked by L-2-methyl-3-(-4-hydroxyphenyl)alanine and 3-(hydrazinomethyl)phenol hydrochloride, inhibitors of tyrosine hydroxylase and L-aromatic amino acid decarboxylase, respectively, suggesting that this effect involved catecholamine synthesis. The latter hypothesis was confirmed by finding that DHEAS and ALLO increased both the mRNA and protein levels of tyrosine hydroxylase. In conclusion, our findings suggest that neuroactive steroids exert a direct tonic effect on adrenal catecholamine synthesis and secretion. These data associate the adrenomedullary malfunction observed in old age and neuroactive steroids.


Assuntos
Actinas/metabolismo , Catecolaminas/genética , Sulfato de Desidroepiandrosterona/farmacologia , Pregnanolona/farmacologia , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Catecolaminas/biossíntese , Primers do DNA , Desidroepiandrosterona/farmacologia , Indução Enzimática , Nicotina/farmacologia , Células PC12 , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tirosina 3-Mono-Oxigenase/metabolismo , Zona Reticular/fisiologia
8.
Artigo em Inglês | MEDLINE | ID: mdl-15032629

RESUMO

The hypothalamic neuropeptide corticotropin-releasing hormone (CRH) is produced by several tissues of the female reproductive system. It is also secreted at inflammatory sites and possesses potent pro-inflammatory properties influencing both innate and acquired immune processes. Uterine CRH participates in local immune early pregnancy phenomena, such as decidualization of endometrial strom a and protection of the fetus from maternal immune system. This is maintained through induction of the expression of apoptotic FasL on invasive extravillous trophoblast and maternal decidual cells at the fetal-maternal interface. Furthermore, CRH increases apoptosis of activated T lymphocytes through FasL induction participating in the process of implantation and early pregnancy. Female rats treated with the non-peptidic CRH receptor 1 (CRHR1) specific antagonist antalarmin, in the first 6 days of gestation, have undergone a decrease of endometrial implantation sites and live embryos and markedly diminished endometrial FasL expression.


Assuntos
Hormônio Liberador da Corticotropina/antagonistas & inibidores , Hormônio Liberador da Corticotropina/fisiologia , Implantação do Embrião/efeitos dos fármacos , Pirimidinas/farmacologia , Pirróis/farmacologia , Animais , Implantação do Embrião/fisiologia , Feminino , Humanos , Gravidez , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/fisiologia
9.
Mol Hum Reprod ; 9(2): 91-5, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12569178

RESUMO

Transforming growth factor beta1 (TGFbeta1) is expressed in human endometrium. It regulates epithelial cell proliferation and apoptosis. The aim of the present work was to examine the role of TGFbeta1 on human endometrial stromal cell apoptosis. Primary cultures of isolated stromal cells were obtained from biopsies of late secretory phase endometrium. We have found the following: (i) TGFbeta1 induced apoptosis of stromal cells in a time- and dose-dependent manner; (ii) blockade of TGFbeta1's autocrine/paracrine effect by TGFbeta1-neutralizing antibodies diminished the basal rate of stromal cell apoptosis; (iii) semi-quantitative Western blot analysis showed that TGFbeta1 caused a rapid but transient elevation of the pro-apoptotic FasL protein, without affecting the levels of Fas receptor; (iv) TGFbeta1 increased the levels of the anti-apoptotic Bcl-2 and Bcl-xL proteins, while having no significant effects on the pro-apoptotic proteins Bax and Bak, suggesting the activation of a transient survival mechanism activated in stromal cells as a parallel rescue response to the apoptosis-inducing FasL protein. In conclusion, our data provide evidence that TGFbeta1 exerts an autocrine pro-apoptotic effect on human endometrial stroma, via the FasL/Fas system.


Assuntos
Apoptose/efeitos dos fármacos , Endométrio/citologia , Glicoproteínas de Membrana/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Células Estromais/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Apoptose/fisiologia , Células Cultivadas , Endométrio/fisiologia , Proteína Ligante Fas , Feminino , Humanos , Cinética , Glicoproteínas de Membrana/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Células Estromais/citologia
10.
J Clin Endocrinol Metab ; 88(1): 478-83, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12519893

RESUMO

The presence of CRH and urocortin (Ucn), members of the CRH family of neuropeptides, was examined in human gastric biopsies from normal controls and in patients with active gastritis from Helicobacter pylori (H. pylori) and after eradication treatment. RT-PCR analysis showed the presence of the Ucn transcript in biopsies (obtained by gastroscopy) from normal and inflamed gastric mucosa, whereas the CRH transcript was not detectable. Immunoreactive (ir-) Ucn was localized (by immunohistochemistry) in gastric epithelial cells and in inflammatory elements of the surrounding negative for Ucn gastric stroma. The level of ir-Ucn was higher in gastric biopsies from the group of patients with active H. pylori gastritis than in normal controls (10.4 +/- 1.8 vs. 2.0 +/- 1.3 pg/ micro g total protein; P < 0.001). After the apparent eradication of H. pylori infection (by clinical and morphological criteria) ir-Ucn levels increased dramatically to 43.1 +/- 9.8 pg/ micro g total protein, (P < 0.001) compared with pretreatment values. Interestingly, nonresponders to the eradication treatment did not show any significant change in ir-Ucn levels (18.7 +/- 12.3 pg/ micro g total protein) compared with their pretreatment values. In conclusion, our data suggest that in human gastric epithelium Ucn is present and plays an important physiological role, whereas CRH is absent. In addition, and in contrast to what has been found for CRH in ulcerative colitis, a highly significant, but negative, correlation has been found between Ucn levels and gastric inflammation, suggesting that Ucn may exert an antiinflammatory effect in gastric mucosa.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Mucosa Gástrica/metabolismo , Gastrite/metabolismo , Hormônio Liberador da Corticotropina/genética , Gastrite/microbiologia , Infecções por Helicobacter , Helicobacter pylori , Humanos , RNA Mensageiro/metabolismo , Distribuição Tecidual , Urocortinas
11.
J Neurochem ; 80(1): 81-90, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11796746

RESUMO

Corticotropin-releasing hormone (CRH) is present in the adrenal gland acting as a paracrine factor via stimulation of the locally expressed CRH receptors. In this study, we examined if the adrenal CRH system also contains a key component of the neuronal CRH-containing system, the CRH-binding protein (CRH-BP). Our data show that: (i) the CRH-BP transcript is detectable using RT-PCR in total RNA isolated from rat adrenals, and (ii) its protein product is also found by western blot analysis in cell lysates. (iii) Immunohistochemical staining showed that adrenomedullary chromaffin cells produce the bulk of adrenal CRH-BP, an ability retained by the PC12 rat pheochromocytoma cell line. (iv) Regulation of adrenal CRH-BP expression by major modulators of the CRH system was also examined. Protein expression appears to be under the positive control of CRH itself, protein kinase A effector cAMP, glucocorticoids and interleukin (IL)-6. It is thus evident that CRH-BP may play a role in mediating their effects in the adrenal. (v) Differentiation of PC12 into neuron-like cells resulted in a significant increase in CRH-BP, parallel to the induction of the CRH peptide itself. In conclusion, CRH-BP mRNA and protein are present in normal rat adrenomedullary chromaffin cells and in the PC12 rat pheochromocytoma cell line, making the adrenal CRH system directly comparable with those described in the CNS.


Assuntos
Glândulas Suprarrenais/metabolismo , Proteínas de Transporte/metabolismo , Animais , Encéfalo/metabolismo , Proteínas de Transporte/genética , Diferenciação Celular , Feminino , Humanos , Células PC12/metabolismo , Células PC12/patologia , Placenta/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
12.
Nat Immunol ; 2(11): 1018-24, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11590404

RESUMO

The semi-allograft embryo in the blastocyst stage implants itself in the endometrium, yet no immune rejection processes are activated. Embryonic trophoblast and maternal decidua produce corticotropin-releasing hormone (CRH) and express Fas ligand (FasL), a proapoptotic cytokine. We found that antalarmin, a CRH receptor type 1 antagonist, decreased FasL expression and promoted apoptosis of activated T lymphocytes, an effect which was potentiated by CRH and inhibited by antalarmin. Female rats treated with antalarmin showed a marked decrease in implantation sites and live embryos and diminished endometrial FasL expression. Embryos from mothers that lacked T cells or from syngeneic matings were not rejected when the mothers were given antalarmin. These findings suggested that locally produced CRH promotes implantation and maintenance of early pregnancy primarily by killing activated T cells.


Assuntos
Blastocisto/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Blastocisto/imunologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Decídua/imunologia , Implantação do Embrião/efeitos dos fármacos , Endométrio/imunologia , Proteína Ligante Fas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Histocompatibilidade , Humanos , Inflamação , Tamanho da Ninhada de Vivíparos/efeitos dos fármacos , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Gravidez , Pirimidinas/farmacologia , Pirimidinas/toxicidade , Pirróis/farmacologia , Pirróis/toxicidade , Ratos , Ratos Endogâmicos F344 , Ratos Nus , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Trofoblastos/imunologia , Receptor fas/fisiologia
13.
Mol Hum Reprod ; 7(9): 867-74, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11517294

RESUMO

Human endometrium expresses specific kappa-opioid binding sites and their endogenous ligands, the dynorphins. In neural crest-derived tissues, kappa-opioids affect apoptosis, a phenomenon of major significance in endometrial stroma physiology. Our hypothesis was that endometrial kappa-opioids may play a role in endometrial stromal cell apoptosis. Thus, we examined the effect of the synthetic kappa-opioid agonist, U69593, on the apoptotic rate of human endometrial stromal cells in primary culture. Apoptosis of endometrial stromal cells was elevated after 3 h exposure to 100 nmol/l U69593, and remained elevated for up to 3 days. This effect was dose-dependent and was reversed by the general opioid antagonist, naloxone, suggesting that it is mediated via opioid receptors. In parallel, semi-quantitative Western blot and flow cytometry analysis showed that U69593 caused a rapid but transient up-regulation of Fas protein, suggesting that its effect on apoptosis is mediated by activation of the Fas/FasL apoptotic pathway. Additionally, U69593 increased the content of the anti-apoptotic members of the Bcl-2 family of proteins, the Bcl-2 and Bcl-x(L), whereas it had no significant effect on the apoptosis-promoting homologues Bax, Bcl-x(S) and Bak. This implies that a transient survival mechanism is activated in stromal cells as a parallel rescue response to the apoptosis-inducing factor. In conclusion, our data suggest that endometrial opioid dynorphins may participate in the apoptotic processes related to endometrial tissue remodelling during early pregnancy or menstruation.


Assuntos
Apoptose , Benzenoacetamidas , Endométrio/citologia , Glicoproteínas de Membrana/fisiologia , Receptores Opioides kappa/fisiologia , Transdução de Sinais , Receptor fas/metabolismo , Apoptose/efeitos dos fármacos , Células Cultivadas , Endométrio/efeitos dos fármacos , Proteína Ligante Fas , Feminino , Humanos , Ligantes , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pirrolidinas/farmacologia , Receptores Opioides kappa/agonistas , Transdução de Sinais/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos
14.
Peptides ; 22(5): 785-93, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11337092

RESUMO

In human endometrium, both epithelial and stroma cells produce corticotropin-releasing hormone (CRH). Both types of cells also possess specific CRH-binding sites indicating a local effect of endometrial CRH. The transcription of the CRH gene in human endometrium is under the control of steroid hormones and locally produced prostanoids and interleukins. Endometrial CRH interacts with locally produced prostaglandins and interleukins. Based on these observations it can be hypothesized that CRH, prostaglandins and interleukins form a network responsible for the communication between epithelial and stromal cells, at the level of the endometrium, and between endometrial and myometrial cells at the level of uterus. The net product of these interaction is the micro-regulation of the decidualizing process and the preparation of endometrium for the implantation/nidation of the conceptus. Indeed, this network may represent the core of the intrauterine neuroendocrine-immune interactions involved in the decidualization of stroma and implantation of blastocyst. In addition, this network appears to be essential for the fine-tuning of myometrial tone.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Implantação do Embrião/fisiologia , Endométrio/fisiologia , Miométrio/fisiologia , Útero/metabolismo , Animais , Comunicação Celular , Endométrio/citologia , Células Epiteliais/metabolismo , Feminino , Glucocorticoides/metabolismo , Humanos , Interleucinas/metabolismo , Miométrio/citologia , Células Estromais/metabolismo , Útero/citologia
15.
Gynecol Endocrinol ; 15(2): 98-102, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11379015

RESUMO

Corticotropin-releasing hormone (CRH) is synthesized in most female reproductive tissues such as the ovaries and the uterus. In the non-pregnant uterus, it is mainly produced by epithelial cells of the endometrium. Recent in vitro experimental findings show that endometrial CRH is under the positive control of progesterone, participating in the decidualization process of endometrial stroma and the progression of blastocyst implantation. CRH is also produced in the thecal compartment of the human ovary, controlling ovarian steroid hormone biosynthesis. In the present study we compared the concentration of immunoreactive CRH (ir-CRH) in biopsies from proliferative and secretory human endometria, and from pre- and postmenopausal human ovaries. We found that the concentration of ir-CRH was significantly higher in the secretory (92 +/- 8 pg/mg protein; n = 10) than the proliferative (75 +/- 9 pg/mg protein; n = 12; p < 0.05) endometria. This observation supports the experimental in vitro findings associating endometrial CRH in intrauterine phenomena of the secretory phase of the menstrual cycle (decidualization and implantation). Additionally, we have shown that the concentration of ir-CRH was significantly higher in the premenopausal (125 +/- 12 pg/mg protein; n = 14) than the postmenopausal (100 +/- 12 pg/mg protein; n = 12; p < 0.05) ovaries, suggesting that ovarian CRH is related to normal ovarian function during the reproductive lifespan.


Assuntos
Envelhecimento , Hormônio Liberador da Corticotropina/análise , Endométrio/química , Ciclo Menstrual/fisiologia , Ovário/química , Biópsia , Feminino , Humanos
16.
Endocrinology ; 142(5): 2022-31, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11316769

RESUMO

Catecholamine secretion and actin filament disassembly are closely coupled in chromaffin cells. Opioid suppression of catecholamine secretion is fast and transient, both characteristics of actin filament involvement. The aim of the present work was to test the hypothesis that opioids suppress catecholamine secretion via an inhibitory effect on actin filament disassembly. For this purpose we used the PC12 rat pheochromocytoma cell line. Norepinephrine and dopamine were measured by enzyme-linked immunosorbent assay or RIA. Polymerized actin was measured by rhodamine-phalloidin and visualized by confocal laser scanning microscopy. Opioids suppressed basal catecholamine secretion. The onset of this effect was fast and transient, peaking at 2 min, and was reversible by opioid antagonists. Synchronously, opioids suppressed actin filament disassembly; this was also reversible by opioid antagonists. Cytochalasin B prevented the inhibitory effect of opioids on catecholamine secretion. In addition, opioids suppressed the stimulatory effect of nicotine on catecholamine secretion and actin depolymerization. Changes in actin cytoskeleton in neuron-like PC12 cells make them resistant to both effects of opioids, i.e. on catecholamine secretion and actin disassembly. In conclusion, our data suggest that the suppressive effect of opioids on basal and nicotine-induced catecholamine secretion may result from an opioid-provoked stabilization of cortical actin. It also appears that basal catecholamine secretion is associated with opioid-sensitive machinery regulating the continuous formation of short-lived areas of cortical actin filament disassembly.


Assuntos
Actinas/metabolismo , Benzenoacetamidas , Entorpecentes/farmacologia , Nicotina/farmacologia , Norepinefrina/metabolismo , Animais , Citocalasina B/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Leucina Encefalina-2-Alanina/farmacologia , Humanos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Células PC12 , Pirrolidinas/farmacologia , Ratos
17.
J Cell Physiol ; 187(3): 386-91, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11319762

RESUMO

The primary neuroendocrine carcinoma of the skin or Merkel cell carcinoma (MCC) is a skin tumor with aggressive biological behaviour. Experimental models for investigating the biological properties of the tumor are prerequisite for developing new therapeutic approaches. In this study, we report the establishment and characterisation of a cell line derived from the lymph-node metastasis of a patient with highly aggressive MCC. Merkel carcinoma cells (MCC-1) grew as floating aggregates in suspension cultures for more than two years and over 70 subcultures. The proliferation rate in suspension cultures was rather moderate with a population doubling time of 69 h. The immunocytochemical pattern of the cultured MCC-1 was similar to that of the original tumor with expression of cytokeratin 18, neuron-specific enolase, neurofilaments, and synaptophysin. In addition, reverse transcriptase polymerase chain reaction (RT-PCR) revealed presence of chromogranin A mRNA in the MCC-1 cell line. Furthermore, electron microscopy yielded the rare finding of neuroendocrine granules in the cytoplasm of the cultured cells. The cell line MCC-1 was able to form colonies in soft agar. Nude mice developed solid tumors with similar histology to the original tumor after subcutaneous and intravenous injections of cultured MCC-1, and malignant ascites was seen after intraperitoneal injection. Also, two MCC-1 sublines were established by reculturing cells from the xenografts grown in vivo and immunocytochemistry confirmed their neuroendocrine origin. The MCC-1 line may thus serve as a model for studying the biology and the metastatic potential of Merkel cell carcinoma.


Assuntos
Carcinoma de Célula de Merkel/metabolismo , Neoplasias Cutâneas/metabolismo , Células Tumorais Cultivadas , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos de Diferenciação/biossíntese , Carcinoma de Célula de Merkel/patologia , Divisão Celular , Cromogranina A , Cromograninas/biossíntese , Cromograninas/genética , Humanos , Queratinas/biossíntese , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fosfopiruvato Hidratase/biossíntese , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura , Neoplasias Cutâneas/patologia , Sinaptofisina/biossíntese , Transplante Heterólogo
18.
Mol Med ; 6(4): 303-18, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10949911

RESUMO

BACKGROUND: The brief incubation of opossum kidney (OK) cells with low P(i) results in Na+/P(i) cotransport up-regulation and in substantial, but transient, cytoskeletal reorganization. In this study, we examined signaling events involved in the depolymerization of microfilaments. RESULTS: Confocal laser scanning microscopy, immunoblot and immunoprecipitation experiments revealed villin co-localization with mainly actin short filaments and monomers, indicating that under the conditions used, villin acted as an actin-severing protein. Further analysis revealed that low concentrations of extracellular phosphate resulted in phospholipase Cgammal (PLC-gammal) translocation to the actin cytoskeleton, without increases in its tyrosine phosphorylation. Additionally, tyrosine phosphorylation of a portion of insoluble villin was increased; whereas, only tyrosine phosphorylated villin associated with PLC-gammal. Although, tyrosine phosphorylation of PLC-gammal was not observed during Na+/P(i) cotransport up-regulation, genistein treatment abolished the enzyme's translocation to the actin cytoskeleton, as well as its association with villin. In addition, villin was found to associate with the 85-KDa subunit (p85) of phosphatidylinositol (PI)-3 kinase, concomitant with PLC-gammal, in the cytoskeletal fraction of Na+/P(i) cotransport up-regulated cells. CONCLUSIONS: Our observations suggest a signaling mechanism linking low ambient P(i) levels to the acute up-regulation of its cotransport with sodium and the depolymerization of the subcortical actin cytoskeleton.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Citoesqueleto/metabolismo , Isoenzimas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais , Simportadores , Fosfolipases Tipo C/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/enzimologia , Ativação Enzimática/efeitos dos fármacos , Genisteína/farmacologia , Isoenzimas/antagonistas & inibidores , Microscopia Confocal , Gambás , Fosfatos/metabolismo , Fosfatos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama , Fosforilação/efeitos dos fármacos , Fosfotirosina/antagonistas & inibidores , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sódio/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato , Solubilidade , Fosfolipases Tipo C/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos
19.
Mol Hum Reprod ; 6(7): 602-9, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10871647

RESUMO

The transforming growth factor beta1 (TGFbeta1) is a major regulator of human endometrial function. Human endometrium possesses specific opioid binding sites, the majority of which belong to the kappa type, for which the prodynorphin-derived opioids are the endogenous ligands. Since these two systems interact in several other tissues we postulated that opioids may affect the production of TGFbeta1 in human endometrium. We have found that kappa opioids exerted a time- and dose-dependent inhibitory effect on TGFbeta1 production from endometrial stromal and epithelial cells and from the Ishikawa human endometrial adenocarcinoma cell line. This effect was reversible by the specific opioid antagonist diprenorphine. To examine if this effect represents a paracrine endometrial response to locally produced kappa opioids we searched for the presence of the endogenous kappa opioid receptor ligands. Indeed, the prodynorphin transcript was detectable on Northern blots from normal and tumoral human endometrial cells; its size was that of the pituitary transcript, i.e. approximately 2.4 kb long. Most immunoreactive dynorphin from human endometrium had a molecular weight of 8 kDa. Finally, immunofluorescence staining of normal and tumoral human endometrial cells revealed the presence of dynorphin-positive cytoplasmic secretory granules. Taken together, our data suggest that in human endometrium, kappa opioids and the TGFbeta1 form a paracrine network which appears to be retained by the Ishikawa human endometrial adenocarcinoma cell line.


Assuntos
Endométrio/metabolismo , Encefalinas/farmacologia , Precursores de Proteínas/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Células Cultivadas , Diprenorfina/farmacologia , Relação Dose-Resposta a Droga , Encefalinas/metabolismo , Feminino , Humanos , Entorpecentes/metabolismo , Entorpecentes/farmacologia , Precursores de Proteínas/metabolismo , Receptores Opioides kappa/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Mol Hum Reprod ; 6(4): 344-51, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10729317

RESUMO

The hypothalamic neuropeptide, corticotrophin-releasing hormone (CRH), which is also produced by human endometrium, has been shown to induce its decidualization in vitro. This process, induced mainly by progesterone, has characteristics of an aseptic inflammatory reaction, and is modulated by locally produced pro-inflammatory factors. In humans, prostaglandin E(2) (PGE(2)) enhances while interleukin (IL)-1 inhibits the decidualizing effect of progesterone. The aim of the present work was to test the hypothesis that CRH might affect the decidualization of human endometrium interacting with these factors. Therefore, we studied its effects on the production of pro-inflammatory interleukins IL-1, IL-6 and of PGE(2) from human endometrial stromal cells in primary culture. The results strongly suggest that CRH decidualizes stromal cells, as judged by the appearance of cytokeratins and the production of prolactin, two established markers of decidualization. In parallel to its effect on decidualization, CRH also decreased the production of PGE(2), while it increased the production of IL-1 and IL-6. Exposure of endometrial stromal cells to IL-6 also caused decidualization. The data presented here suggest that endometrial CRH regulates the production of local modulators of decidualization, i.e. PGE(2), IL-1 and IL-6. We postulate that, through the regulation of these factors, CRH acts as a local fine-tuner of decidualization initiated by progesterone.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Dinoprostona/metabolismo , Endométrio/citologia , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Células Estromais/metabolismo , Diferenciação Celular , Hormônio Liberador da Corticotropina/farmacologia , Decídua/citologia , Dinoprostona/biossíntese , Feminino , Humanos , Interleucina-1/biossíntese , Interleucina-6/biossíntese , Interleucina-6/farmacologia , Queratinas/biossíntese , Prolactina/biossíntese , Células Estromais/citologia , Células Estromais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...