Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Cell Res ; 333(1): 136-46, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25724897

RESUMO

BACKGROUND: Ephrin-B2 is the sole physiologically-relevant ligand of the receptor tyrosine kinase EphB4, which is over-expressed in many epithelial cancers, including 66% of prostate cancers, and contributes to cancer cell survival, invasion and migration. Crucially, however, the cancer-promoting EphB4 signalling pathways are independent of interaction with its ligand ephrin-B2, as activation of ligand-dependent signalling causes tumour suppression. Ephrin-B2, however, is often found on the surface of endothelial cells of the tumour vasculature, where it can regulate angiogenesis to support tumour growth. Proteolytic cleavage of endothelial cell ephrin-B2 has previously been suggested as one mechanism whereby the interaction between tumour cell-expressed EphB4 and endothelial cell ephrin-B2 is regulated to support both cancer promotion and angiogenesis. METHODS: An in silico approach was used to search accessible surfaces of 3D protein models for cleavage sites for the key prostate cancer serine protease, KLK4, and this identified murine ephrin-B2 as a potential KLK4 substrate. Mouse ephrin-B2 was then confirmed as a KLK4 substrate by in vitro incubation of recombinant mouse ephrin-B2 with active recombinant human KLK4. Cleavage products were visualised by SDS-PAGE, silver staining and Western blot and confirmed by N-terminal sequencing. RESULTS: At low molar ratios, KLK4 cleaved murine ephrin-B2 but other prostate-specific KLK family members (KLK2 and KLK3/PSA) were less efficient, suggesting cleavage was KLK4-selective. The primary KLK4 cleavage site in murine ephrin-B2 was verified and shown to correspond to one of the in silico predicted sites between extracellular domain residues arginine 178 and asparagine 179. Surprisingly, the highly homologous human ephrin-B2 was poorly cleaved by KLK4 at these low molar ratios, likely due to the 3 amino acid differences at this primary cleavage site. CONCLUSION: These data suggest that in in vivo mouse xenograft models, endogenous mouse ephrin-B2, but not human tumour ephrin-B2, may be a downstream target of cancer cell secreted human KLK4. This is a critical consideration when interpreting data from murine explants of human EphB4+/KLK4+ cancer cells, such as prostate cancer cells, where differential effects may be seen in mouse models as opposed to human clinical situations.


Assuntos
Efrina-B2/química , Calicreínas/química , Calicreínas/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Calicreínas/fisiologia , Masculino , Camundongos , Dados de Sequência Molecular , Transplante de Neoplasias , Neoplasias da Próstata , Proteólise , Células Sf9
2.
Growth Factors ; 32(6): 207-13, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25413947

RESUMO

Several Eph receptor tyrosine kinases (RTKs) are commonly over-expressed in epithelial and mesenchymal cancers and are recognized as promising therapeutic targets. Although normal interaction between Eph receptors and their ephrin ligands stimulates kinase activity and is generally tumor suppressive, significant Eph over-expression allows activation of ligand- and/or kinase-independent signaling pathways that promote oncogenesis. Single-agent kinase inhibitors are widely used to target RTK-driven tumors but acquired and de novo resistance to such agents is a major limitation to effective clinical use. Accumulating evidence suggests that Ephs can be inhibited by "leaky" or low-specificity kinase inhibitors targeted at other RTKs. Such off-target effects may therefore inadvertently promote ligand- and/or kinase-independent oncogenic Eph signaling, thereby providing a new mechanism by which resistance to the RTK inhibitors can emerge. We propose that combining specific, non-leaky kinase inhibitors with tumor-suppressive stimulators of Eph signaling may provide more effective treatment options for overcoming treatment-induced resistance and clinical failure.


Assuntos
Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Receptores da Família Eph/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo
3.
Endocr Relat Cancer ; 12(4): 839-50, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16322325

RESUMO

While oestrogen, progesterone and growth factors, including growth hormone (GH), are clearly implicated in the pathogenesis of breast cancer, there is now evidence that the newly described ghrelin axis is also involved. The aims of this study were to investigate the expression of the ghrelin axis in breast cancer tissues and cell lines and to examine the effect of ghrelin on breast cancer cell proliferation in vitro. Ghrelin and its functional receptor, the growth hormone secretagogue receptor (GHSR) type 1a, were expressed in normal breast tissue and breast cancer specimens and cell lines. In contrast, the truncated GHSR type 1b isoform was exclusively expressed in breast carcinoma, suggesting that it has potential as a diagnostic marker. Ghrelin treatment significantly increases the proliferation of the MDA-MB-435 and MDA-MB-231 breast cancer cell lines in vitro. In addition, we have described the expression of a human preproghrelin isoform, exon 3-deleted preproghrelin, which encodes mature ghrelin plus a novel C-terminal peptide. Quantitative RT-PCR was used to demonstrate that this mRNA isoform is highly expressed in the MDA-MB-435 metastatic breast cancer cell line relative to the benign MCF-10A breast epithelial cell line. The unique C-terminal peptide of exon 3-deleted preproghrelin is expressed in the glandular epithelium of breast cancer tissues, with high-grade carcinoma exhibiting the strongest immunoreactivity. The data presented here suggest that components of the ghrelin axis may represent novel markers for breast cancer and potential therapeutic targets.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma/metabolismo , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/farmacologia , Hormônios Peptídicos/fisiologia , Sequência de Aminoácidos , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/patologia , Carcinoma/diagnóstico , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Grelina , Humanos , Imuno-Histoquímica , Dados de Sequência Molecular , Hormônios Peptídicos/análise , Hormônios Peptídicos/genética , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/genética , Receptores de Grelina , Deleção de Sequência , Transcrição Gênica
4.
Endocr Relat Cancer ; 12(3): 631-43, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16172196

RESUMO

Prostate-specific antigen (PSA) and the related kallikrein family of serine proteases are current or emerging biomarkers for prostate cancer detection and progression. Kallikrein 4 (KLK4/hK4) is of particular interest, as KLK4 mRNA has been shown to be elevated in prostate cancer. In this study, we now show that the comparative expression of hK4 protein in prostate cancer tissues, compared with benign glands, is greater than that of PSA and kallikrein 2 (KLK2/hK2), suggesting that hK4 may play an important functional role in prostate cancer progression in addition to its biomarker potential. To examine the roles that hK4, as well as PSA and hK2, play in processes associated with progression, these kallikreins were separately transfected into the PC-3 prostate cancer cell line, and the consequence of their stable transfection was investigated. PC-3 cells expressing hK4 had a decreased growth rate, but no changes in cell proliferation were observed in the cells expressing PSA or hK2. hK4 and PSA, but not hK2, induced a 2.4-fold and 1.7-fold respective increase, in cellular migration, but not invasion, through Matrigel, a synthetic extracellular matrix. We hypothesised that this increase in motility displayed by the hK4 and PSA-expressing PC-3 cells may be related to the observed change in structure in these cells from a typical rounded epithelial-like cell to a spindle-shaped, more mesenchymal-like cell, with compromised adhesion to the culture surface. Thus, the expression of E-cadherin and vimentin, both associated with an epithelial-mesenchymal transition (EMT), was investigated. E-cadherin protein was lost and mRNA levels were significantly decreased in PC-3 cells expressing hK4 and PSA (10-fold and 7-fold respectively), suggesting transcriptional repression of E-cadherin, while the expression of vimentin was increased in these cells. The loss of E-cadherin and associated increase in vimentin are indicative of EMT and provides compelling evidence that hK4, in particular, and PSA have a functional role in the progression of prostate cancer through their promotion of tumour cell migration.


Assuntos
Caderinas/metabolismo , Calicreínas/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/patologia , Divisão Celular , Linhagem Celular Tumoral , Movimento Celular , Células Epiteliais/patologia , Humanos , Calicreínas/genética , Masculino , Mesoderma/patologia , Invasividade Neoplásica
5.
Endocrinology ; 146(1): 432-40, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15471962

RESUMO

Ghrelin, an n-octanoylated 28-amino-acid peptide capable of inducing GH secretion and food intake in humans and rats, is the endogenous ligand for the GH secretagogue receptor (GHS-R). Here we describe the expression and tissue distribution of the ghrelin/GHS-R axis in the mouse. We also report for the first time the identification of a novel mouse ghrelin mRNA variant in which there is a complete deletion of exon 4. Translation of this variant mRNA yields a protein containing ghrelin and an alternative C-terminal domain with a unique C-terminal peptide sequence. RT-PCR with primers specific for mouse ghrelin was used to demonstrate the mRNA expression of the full preproghrelin transcript and the exon 4-deleted variant in multiple mouse tissues. Real-time PCR was also employed to quantitate mRNA expression of ghrelin, the novel isoform and a previously reported ghrelin gene variant, ghrelin gene-derived transcript. We also demonstrated the tissue expression of the functional GHS-R in the mouse. Immunohistochemistry, employing antibodies raised against the mature human n-octanoylated ghrelin peptide and the putative C-terminal peptide encoded by the exon 4-deleted proghrelin variant, was used to demonstrate protein expression of ghrelin and the variant in multiple mouse tissues including stomach, kidney, and reproductive tissues. The coexpression of ghrelin and its receptor in a wide range of murine tissues suggests varied autocrine/paracrine roles for these peptides. Exon 4-deleted proghrelin, a novel mouse proghrelin isoform with a unique C-terminal peptide sequence, is also widely expressed in the mouse and thus may possess biological activity in these tissues.


Assuntos
Éxons , Deleção de Genes , Expressão Gênica , Variação Genética , Hormônios Peptídicos/genética , Precursores de Proteínas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Sistemas Computacionais , Mucosa Gástrica/metabolismo , Grelina , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Hormônios Peptídicos/metabolismo , Reação em Cadeia da Polimerase , Precursores de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Grelina , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
6.
J Clin Endocrinol Metab ; 90(3): 1798-804, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15585554

RESUMO

Ghrelin, the endogenous ligand of the GH secretagogue receptor (GHS-R), is a newly identified, ubiquitously expressed molecule that has been involved in a wide array of endocrine and nonendocrine functions, including cell proliferation. In this context, our group recently reported the expression of ghrelin and its functional receptor, the GHS-R type 1a, in the human ovary and testis as well as several testicular tumors. Ovarian malignancies, however, remain unexplored. Notably, a vast majority of ovarian tumors derive from the surface epithelium, which originates from the celomic epithelium. Considering the proven expression of ghrelin in the human ovary, and its reported effects in the proliferative activity of different cancer cell lines, we aimed at evaluating whether the ovarian surface epithelium as well as related reproductive structures and tumors are potential targets of ghrelin. To this end, expression of GHS-R1a was analyzed by immunohistochemistry in a panel of normal, metaplastic, and neoplastic tissues. Uniform GHS-R1a immunostaining was detected throughout the ovarian surface epithelium. Likewise, ciliated cells within the fallopian tube epithelium showed strong GHS-R1a expression. In contrast, other celomic derivatives, such as endometrium and endocervix, were negative for GHS-R1a immunoreactivity. In keeping with data from normal tissues, inclusion cysts from the surface epithelium expressed GHS-R1a. Similarly, benign serous tumors resembling fallopian tube epithelium were also positive, whereas serous cystadenocarcinomas showed GHS-R1a expression only in highly differentiated specimens. In contrast, other neoplasms, such as mucinous cystadenomas and cystadenocarcinomas, endometrioid tumors, clear cell carcinomas, and Brenner tumors, did not express GHS-R1a. In conclusion, our results demonstrate that the ovarian surface epithelium and related tumors are potential targets for systemic or locally produced ghrelin because they express the functional type 1a GHS-R. Considering the relevant role of the ovarian surface epithelium in key physiological events (such as ovulation) and neoplastic transformation of the ovary, the potential actions of ghrelin in those phenomena merit further investigation.


Assuntos
Tubas Uterinas/metabolismo , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Colo do Útero/embriologia , Colo do Útero/metabolismo , Endométrio/embriologia , Endométrio/metabolismo , Células Epiteliais/metabolismo , Tubas Uterinas/embriologia , Feminino , Humanos , Imuno-Histoquímica , Ductos Paramesonéfricos/embriologia , Neoplasias Ovarianas/patologia , Ovário/embriologia , Receptores de Grelina
7.
J Clin Endocrinol Metab ; 89(1): 400-9, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14715878

RESUMO

Ghrelin, the endogenous ligand for the GH secretagogue receptor (GHS-R), has been primarily linked to the central neuroendocrine regulation of GH secretion and food intake, although additional peripheral actions of ghrelin have also been reported. In this context, the expression of ghrelin and its cognate receptor has been recently demonstrated in rat testis, suggesting a role for this molecule in the direct control of male gonadal function. However, whether this signaling system is present in human testis remains largely unexplored. In this study we report the expression and cellular location of ghrelin and its functional receptor, the type 1a GHS-R, in adult human testis. In addition, evaluation of ghrelin and GHS-R1a immunoreactivity in testicular tumors and dysgenetic tissue is presented. The expression of the mRNAs encoding ghrelin and GHS-R1a was demonstrated in human testis specimens by RT-PCR, followed by direct sequencing. In normal testis, ghrelin immunostaining was demonstrated in interstitial Leydig cells and, at lower intensity, in Sertoli cells within the seminiferous tubules. In contrast, ghrelin was not detected in germ cells at any stage of spermatogenesis. The cognate ghrelin receptor showed a wider pattern of cellular distribution, with detectable GHS-R1a protein in germ cells, mainly in pachytene spermatocytes, as well as in somatic Sertoli and Leydig cells. Ghrelin immunoreactivity was absent in poorly differentiated Leydig cell tumor, which retained the expression of GHS-R1a peptide. In contrast, highly differentiated Leydig cell tumors expressed both the ligand and the receptor. The expression of ghrelin and GHS-R1a was also detected in dysgenetic Sertoli cell-only seminiferous tubules, whereas germ cell tumors (seminoma and embryonal carcinoma) were negative for ghrelin and were weakly positive for GHS-R1a. In conclusion, our results demonstrate that ghrelin and the type 1a GHS-R are expressed in adult human testis and testicular tumors. Overall, the expression of ghrelin and its functional receptor in human and rat testis, with roughly similar patterns of cellular distribution, is highly suggestive of a conserved role for this newly discovered molecule in the regulation of mammalian testicular function.


Assuntos
Expressão Gênica , Hormônios Peptídicos/genética , Receptores Acoplados a Proteínas G/genética , Neoplasias Testiculares/química , Testículo/química , Adulto , Idoso , Carcinoma Embrionário/química , Grelina , Humanos , Imuno-Histoquímica , Tumor de Células de Leydig/química , Células Intersticiais do Testículo/química , Masculino , Pessoa de Meia-Idade , Hormônios Peptídicos/análise , RNA Mensageiro/análise , Receptores Acoplados a Proteínas G/análise , Receptores de Grelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Túbulos Seminíferos/química , Seminoma/química , Células de Sertoli/química
8.
Eur J Clin Invest ; 33(7): 574-81, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12814394

RESUMO

BACKGROUND: Peroxisome proliferator activated receptor gamma (PPARgamma) is a ligand-activated transcription factor known to be central to both adipose tissue development and insulin action. Growth of adipose tissue requires differentiation of preadipocytes with acquisition of specific cellular functions including insulin sensitivity, leptin secretion and the capacity to store triglyceride. Dietary fatty acids and members of the thiazolidinedione class of compounds have been reported to influence adipogenesis at the transcriptional level. Here, we compare the effects of a dietary fatty acid, linoleic acid, and a thiazolidinedione, rosiglitazone, on biochemical and functional aspects of human preadipocyte differentiation in vitro. MATERIALS AND METHODS: Human omental and subcutaneous preadipocytes were subcultured 2-3 times and subsequently differentiated for 21 days in the presence of either linoleic acid or rosiglitazone. Differentiation was assessed using a number of biochemical and functional parameters. RESULTS: Omental and subcutaneous preadipocytes differentiated in the presence of linoleic acid showed marked cytoplasmic triacylglycerol accumulation however, no biochemical markers of differentiation (LPL expression, G3PDH gene expression and enzyme activity and leptin expression or secretion) were detected. In contrast, treatment of these cells with rosiglitazone induced full biochemical differentiation as judged by all markers assessed, despite comparatively little lipid accumulation. The rosiglitazone effects were subcutaneous depot-specific. Cells treated with linoleic acid showed decreased glucose uptake cf rosiglitazone-treated cells. A luciferase reporter assay demonstrated that rosiglitazone potently activates h-peroxisome proliferator activated receptor gamma while linoleic acid had no effect. CONCLUSIONS: These studies demonstrate that (a) human preadipocytes have the potential to accumulate triacylglycerol irrespective of their stage of biochemical differentiation; (b) while omental preadipocytes are refractory to biochemical differentiation in vitro, they are able to accumulate triacylglycerol; and (c) rosiglitazone and linoleic acid may exert their effects via different biochemical pathways.


Assuntos
Adipócitos/citologia , Tecido Adiposo/citologia , Diferenciação Celular/efeitos dos fármacos , Ácido Linoleico/uso terapêutico , Tiazóis/uso terapêutico , Tiazolidinedionas , Adipócitos/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Adulto , Idoso , Diferenciação Celular/fisiologia , Feminino , Humanos , Hipoglicemiantes/uso terapêutico , Masculino , Pessoa de Meia-Idade , Receptores Citoplasmáticos e Nucleares/metabolismo , Rosiglitazona , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
9.
J Clin Endocrinol Metab ; 88(2): 879-87, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12574228

RESUMO

Ghrelin is a novel 28-amino acid peptide identified as the endogenous ligand for the GH secretagogue receptor (GHS-R). Besides its hallmark central neuroendocrine effects in the control of GH secretion and food intake, an unexpected reproductive facet of ghrelin has recently emerged because expression of this molecule and its cognate receptor has been demonstrated in rat testis. However, whether this signaling system is present in human gonads remains to be evaluated. In this study, we have assessed the presence and cellular location of ghrelin and its functional receptor, namely the type 1a GHS-R, in the cyclic human ovary by means of immunohistochemistry using specific polyclonal antibodies. Strong ghrelin immunostaining was demonstrated in ovarian hilus interstitial cells. In contrast, ghrelin signal was not detected in ovarian follicles at any developmental stage, nor was it present in newly formed corpora lutea (CL) at very early development. However, specific ghrelin immunoreactivity was clearly observed in young and mature CL, whereas expression of the peptide disappeared in regressing luteal tissue. Concerning the cognate receptor, ovarian expression of GHS-R1a protein showed a wider pattern of tissue distribution, with detectable specific signal in oocytes as well as somatic follicular cells; luteal cells from young, mature, old, and regressing CL; and interstitial hilus cells. Of particular note, follicular GHS-R1a peptide expression paralleled follicle development with stronger immunostaining in granulosa and theca layers of healthy antral follicles. In conclusion, our results are the first to demonstrate that ghrelin and its functional type 1a receptor are expressed in the cyclic human ovary with distinct patterns of cellular location. The presence of both components (ligand and receptor) of the ghrelin signaling system within the human ovary opens up the possibility of a potential regulatory role of this novel molecule in ovarian function under physiological and pathophysiological conditions.


Assuntos
Folículo Ovariano/química , Hormônios Peptídicos/análise , Receptores de Superfície Celular/análise , Receptores Acoplados a Proteínas G , Especificidade de Anticorpos , Feminino , Grelina , Humanos , Ciclo Menstrual/fisiologia , Hormônios Peptídicos/imunologia , Receptores de Superfície Celular/imunologia , Receptores de Grelina
10.
Growth Horm IGF Res ; 12(2): 126-36, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12175650

RESUMO

Prostate cancer is a significant cause of morbidity and mortality in Western males. While it is known that androgens play a central role in prostate cancer development and progression, other hormones and growth factors are also involved in prostate growth. Insulin-like growth factor-I (IGF-I) plasma levels have been associated with prostate cancer risk, and growth hormone (GH), a major factor regulating IGF levels, also appears to have a role in prostate cancer cell growth. Most significantly, GH has been shown to increase the rate of cell proliferation in prostate cancer cell lines. We have now demonstrated the co-expression of GH and GH receptor (GHR) mRNA isoforms in the ALVA41, PC3, DU145, LNCaP prostate cancer cells by reverse transcription polymerase chain reaction. Sequence analysis has confirmed that these cell lines express the pituitary form of GH mRNA and also the placental mRNA isoform. These prostate cancer cell lines also express the full-length mRNA for the GHR and the exon 3 deleted isoform. We have also demonstrated the presence of GH and GHR proteins in these cell lines by immunohistochemistry. GH expression has not been described previously in human prostate cancer cells. The co-expression of GH and its receptor would enable an autocrine-paracrine pathway to exist in the prostate that would be capable of stimulating prostate growth, either directly via the GHR or indirectly via IGF production. The GH axis in the prostate could therefore be an important additional target for the future development of prostate cancer therapies.


Assuntos
Hormônio do Crescimento/biossíntese , Hormônio do Crescimento/química , Neoplasias da Próstata/metabolismo , Receptores da Somatotropina/biossíntese , Receptores da Somatotropina/química , Northern Blotting , Southern Blotting , Divisão Celular , Núcleo Celular/metabolismo , Meios de Cultivo Condicionados/farmacologia , Primers do DNA/farmacologia , DNA Complementar/metabolismo , Éxons , Humanos , Imuno-Histoquímica , Masculino , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
11.
J Endocrinol ; 172(3): R7-11, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11874717

RESUMO

This study has examined the expression of two new facets of the growth hormone axis, the growth hormone secretagogue receptor (GHS-R) and its recently identified putative natural ligand ghrelin, in prostate cancer cells. GHS-R 1a and 1b isoforms and ghrelin mRNA expression were detected by RT-PCR in the ALVA-41, LNCaP, DU145 and PC3 prostate cancer cell lines. A normal prostate cDNA library expressed GHS-R1a, but not the 1b isoform or ghrelin. Immunohistochemical staining for the GHS-R 1a isoform and ghrelin was positive in the four cell lines studied. PC3 cells showed increased cell proliferation in vitro in response to ghrelin to levels 33% above untreated controls, implying a potential tumour-promoting role for ghrelin in this tissue. This study is the first to demonstrate the co-expression of the GHS-R and ghrelin in prostate cancer cells. It is also the first study to provide evidence that a previously unrecognised autocrine/paracrine pathway involving ghrelin, that is capable of stimulating growth, exists in prostate cancer.


Assuntos
Hormônios Peptídicos , Peptídeos/metabolismo , Neoplasias da Próstata/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G , Grelina , Humanos , Masculino , Peptídeos/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/análise , Receptores de Superfície Celular/genética , Receptores de Grelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
12.
Prostate ; 49(2): 116-21, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11582590

RESUMO

BACKGROUND: Recent studies have shown that GHRH antagonists inhibit prostate tumour growth and IGF-II production both in vivo and in vitro. The mechanism underlying these observations is unknown, but may involve an interaction with a prostatic GHRH receptor (GHRH-R), raising the possibility of an autocrine pathway for the GHRH axis in the prostate. METHODS: GHRH and GHRH-R mRNA expression was examined by RT-PCR in human prostate cancer cell lines, and the authenticity of PCR products was confirmed by Southern analysis and cDNA sequencing. Immunohistochemical techniques were used to examine the expression of GHRH protein in prostate cancer cell lines. RESULTS: GHRH-R (mRNA) and GHRH (mRNA and protein) are co-expressed in the ALVA-41, DU145, LNCaP and PC3 human prostate cancer cell lines. CONCLUSIONS: These observations suggest the presence of an intact prostatic GHRH autocrine pathway which may stimulate prostate cell proliferation. This pathway may be disrupted by GHRH antagonists.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/biossíntese , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Neuropeptídeos/biossíntese , Receptores de Hormônios Reguladores de Hormônio Hipofisário/biossíntese , Southern Blotting , Divisão Celular/fisiologia , DNA de Neoplasias/biossíntese , DNA de Neoplasias/genética , Hormônio Liberador de Hormônio do Crescimento/genética , Humanos , Imuno-Histoquímica , Masculino , Neoplasias da Próstata/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Células Tumorais Cultivadas
13.
Am J Physiol Endocrinol Metab ; 281(5): E1037-44, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11595661

RESUMO

Adipogenesis is preceded by development of a microvascular network, and optimal functioning of adipose tissue as an energy store and endocrine organ is dependent on extensive vascularization. We have examined the role of endothelial cell-derived factors that influence the proliferation of human preadipocytes. Microvascular endothelial cells and preadipocytes were isolated from human omental and subcutaneous adipose tissue biopsies by use of a developed procedure of collagenase digest, immunoselection, and differential trypsinization. Conditioned medium from microvascular endothelial cell cultures promoted the proliferation of preadipocytes (P = <0.001) and (to a lesser extent) other cell types. No depot-specific differences in mitogenic capacity of microvascular endothelial cell medium or of preadipocyte response were observed. These results indicate that adipose tissue endothelial cells secrete soluble adipogenic factor(s).


Assuntos
Adipócitos/citologia , Tecido Adiposo/irrigação sanguínea , Divisão Celular , Endotélio Vascular/fisiologia , Células-Tronco/citologia , Adulto , Idoso , Anticorpos Monoclonais , Biópsia , Separação Celular , Células Cultivadas , Colágeno/metabolismo , Meios de Cultivo Condicionados , Feminino , Imunofluorescência , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Masculino , Microesferas , Pessoa de Meia-Idade , Omento , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Tripsina/metabolismo
14.
Biol Reprod ; 64(5): 1331-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11319136

RESUMO

Embryo implantation in humans and rodents is a highly invasive yet tightly controlled process involving extracellular matrix (ECM) degradation. Matrix metalloproteinase 9 (MMP-9) has been implicated as the major facilitator of this ECM degradation. MMP-9 is expressed by the embryo's trophoblast cells, whereas tissue inhibitor of metalloproteinases 3 (TIMP-3) is expressed by the maternal uterine cells immediately adjacent to the trophoblast. We examined the functional roles of MMP-9 and TIMP-3 during in vitro ECM degradation by mouse embryos. Blastocysts were treated with either MMP-9 antisense or sense oligonucleotides and incubated on an ECM gel. The extent of ECM degradation exhibited by the blastocysts due to proteinase secretion was quantified. Embryos exposed to MMP-9 antisense oligonucleotides exhibited reduced ECM-degrading activity as compared with controls, and this reduced activity was correlated with the level of MMP-9 secreted by the embryos. The functional role of TIMP-3 was then examined by incubating blastocysts on an ECM gel that had been impregnated with various amounts of TIMP-3. In a dose-dependent manner, increases in TIMP-3 resulted in a reduction in ECM degradation and were correlated with diminished MMP-9 activity. These results provide important functional evidence that in vitro ECM degradation is regulated by embryo-derived MMP-9 and ECM-derived TIMP-3.


Assuntos
Embrião de Mamíferos/ultraestrutura , Matriz Extracelular/metabolismo , Metaloproteinase 9 da Matriz/fisiologia , Inibidor Tecidual de Metaloproteinase-3/fisiologia , Animais , Blastocisto/fisiologia , Blastocisto/ultraestrutura , Linhagem Celular , Cricetinae , Meios de Cultivo Condicionados , Técnicas de Cultura , Implantação do Embrião , Embrião de Mamíferos/fisiologia , Inibidores Enzimáticos , Rim , Inibidores de Metaloproteinases de Matriz , Camundongos , Oligonucleotídeos/farmacologia , Oligonucleotídeos Antissenso/farmacologia
15.
Anat Embryol (Berl) ; 204(6): 477-83, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11876533

RESUMO

Several extracellular matrix (ECM)-degrading proteinases are hypothesised to play important roles during early mammalian development. In particular, urokinase-type plasminogen activator (uPA) and matrix metalloproteinase 9 (MMP-9) are expressed in peri-implantation mouse, sheep, and pig embryos and are implicated in the implantation process. These proteinases are not expressed in early (pre-blastocyst) mouse, sheep or pig embryos. The aim of this study was to establish the gene expression and proteolytic activity of uPA and MMP-9 in in vitro-produced (IVP) cow embryos. Using RT-PCR, mRNA transcripts for uPA and MMP-9 were detected during the first 7 days of development. To investigate the activity of these proteinases, conditioned media from various stages of development (days 2, 3, 4, 5 and 7) were assayed for uPA activity by chromogenic assay and MMP-9 activity by gelatin zymography. Both uPA and MMP-9 activities were detected in the media samples indicating the production and secretion of these proteinases. This pattern of proteinase expression is novel in comparison to the mouse where uPA and MMP-9 are only expressed from the blastocyst stage onwards. The results of this study suggest that these ECM proteinases have a role prior to implantation in the cow, in contrast to that exhibited by mouse, sheep and pig embryos.


Assuntos
Embrião de Mamíferos/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Bovinos , Meios de Cultivo Condicionados/metabolismo , Desenvolvimento Embrionário e Fetal/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas In Vitro , Masculino , Metaloproteinase 9 da Matriz/genética , RNA/análise , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Fatores de Tempo , Ativador de Plasminogênio Tipo Uroquinase/genética
16.
Growth Horm IGF Res ; 10(5): 275-89, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11042024

RESUMO

These studies have established the presence of two major classes of high affinity growth hormone binding proteins in pregnant rat serum, designated GHBPa and GHBPb, with apparent native Mr of 257 K and 98 K respectively. GHBPa, which has not been identified previously, exhibits a binding affinity (2-5 nM(-1)) that is up to 20-fold higher than GHBPb (0.2-0.8 nM(-1)) and is the least abundant form, being approximately 15-20% of total serum GH-binding capacity. Western immunoblot analysis revealed that each GHBP is composed of several immunoreactive proteins which were reactive with carboxy-terminal (RB1615) and/or N-terminal (MAb263) domain antibodies, suggesting the presence of GHBPs with and without the hydrophilic tail. Of importance is that GHBPa exhibited significantly higher Mr (78-182 K, +DTT) than that predicted by GHBP cloning, suggesting that they may be covalently bound to other non-GH-binding proteins or may be distinct entities. GHBPb, on the other hand, was composed of smaller Mr (43/48 K, +DTT) "hydrophilic" tail-containing proteins, some of which were disulphide linked to a larger complex of approximately 110 K. These novel findings challenge the current view of the mechanism for generation of the rat serum GHBP and raise the intriguing possibility that the two classes of GHBP may play distinct and important roles in GH physiology.


Assuntos
Proteínas de Transporte/sangue , Prenhez/sangue , Animais , Anticorpos Monoclonais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Feminino , Hormônio do Crescimento/sangue , Hormônio do Crescimento Humano/metabolismo , Cinética , Fígado/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley
17.
Mol Cell Endocrinol ; 167(1-2): 11-21, 2000 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-11000516

RESUMO

The ADAMs are a multi-functional gene family, some of which have been shown to play a role in diverse biological processes such as fertilization, myogenesis, neurogenesis and the activation of growth factors/immune regulators such as TNF-alpha. So-named because they possess both A Disintegrin And Metalloprotease domain, the ADAMs have potential implications for the metastasis of human tumour cells via cell adhesion and protease activities. However, no studies have yet comprehensively examined the expression or regulation of ADAMs in solid tumours. Therefore, the aim of this study was to examine the expression of the ADAMs in human prostate cancer cell lines and to examine their possible regulation by androgen, a primary hormonal regulator of prostate cancer cell proliferation and metastasis. Applying RT-PCR, ADAM-9, -10, -11, -15 and -17 mRNA expression was found in the androgen-dependent prostate cancer cell lines, LNCaP and ALVA-41 and the androgen-independent cell lines, DU-145 and PC-3. Northern blotting of LNCaP cell total RNA revealed transcripts for ADAM-9 (3.8 kb), ADAM-10 (4.4, 3.2 and 0.54 kb), ADAM-15 (3 kb) and ADAM-17 (4 and 2.6 kb). ADAM-11 transcript was not detected by Northern blotting possibly due to low levels of ADAM-11 mRNA expression. This is the first report of ADAM expression in prostate cancer cell lines. Since androgens are implicated in prostate cancer cell growth and maintenance, the regulation of ADAMs by dihydrotestosterone (DHT) was investigated in the androgen-dependent cell line LNCaP. It was shown by quantitative RT-PCR using continuous fluorescence monitoring that ADAM-10 mRNA expression was regulated in a bell shaped, dose-dependent manner by DHT. Maximum stimulation was observed at 1.0 nM DHT (5-fold significant increase). For ADAM-9 mRNA, a significant upregulation was found at 1.0 and 10 nM (1.5-1.7-fold increase). In contrast, ADAM-17 mRNA, was significantly inhibited at 0.1 and 1.0 nM (1.7-fold decrease). This is the first report, to our knowledge, illustrating hormonal regulation of ADAM mRNA. The novel data described here also provide a strong stimulus to the development of specific quantitative and functional assays for particular ADAMs. These assays, which are not yet available, are required to enable subsequent investigation, both in vitro and in vivo, of the specific roles of each ADAM in prostate cancer cell proliferation, cell motility and invasion.


Assuntos
Di-Hidrotestosterona/farmacologia , Glicoproteínas de Membrana/genética , Metaloendopeptidases/metabolismo , Neoplasias da Próstata/genética , Northern Blotting , Divisão Celular , Relação Dose-Resposta a Droga , Formazans/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/genética , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sais de Tetrazólio/metabolismo , Células Tumorais Cultivadas
18.
Horm Metab Res ; 31(2-3): 216-25, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10226805

RESUMO

IGFBP-3 proteolysis clears IGFBP-3 from body fluids and increases IGF bioavailability. As shown here, native human IGFBP-3 was cleaved by proteases in media conditioned by hamster and insect cells. This proteolysis was less pronounced for IGFBP-3 containing a mutated heparin binding domain, and was prevented by purifying IGFBP-3 on an IGF-I affinity column in the presence of 2 M sodium chloride, suggesting that the responsible protease(s) binds the IGFBP-3 heparin binding domain. To determine if any human proteases act this way, we first studied plasma prekallikrein since it can copurify with IGFBP-3, and found: 1) [125]IGFBP-3 binds to prekallikrein immobilized either on nitrocellulose or on immunocapture plates; 2) the IGFBP-3 heparin binding domain participates in forming the IGFBP-3/prekallikrein complex; 3) the binary IGFBP-3/prekallikrein complex can bind IGF-I to form a ternary complex; and 4) activation of prekallikrein to alpha-kallikrein by Factor XIIa resulted in proteolysis of bound IGFBP-3. This work suggests: 1) cleavage of IGFBP-3 by a protease may be aided by the ability of the protease zymogen to directly bind the IGFBP-3 heparin binding domain; and 2) direct binding of protease zymogens to IGFBP-3 may explain some instances where IGFBP-3 is preferentially proteolyzed in the presence of other IGFBPs.


Assuntos
Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina , Calicreínas/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Fator XII/metabolismo , Expressão Gênica/fisiologia , Heparina/metabolismo , Humanos , Immunoblotting , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/química , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like II/genética , Radioisótopos do Iodo , Dados de Sequência Molecular , Mutagênese/fisiologia , Sondas de Oligonucleotídeos , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína
19.
Endocrinology ; 138(9): 3577-86, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9275038

RESUMO

Previous workers have suggested that guinea pig serum does not contain a GH-binding protein (GHBP) or that it is defective. The current studies, however, have identified and characterized the presence of GH-binding activity in guinea pig serum using gel chromatography to separate bound and free hormone. The detection of GH-binding activity is critically reliant on the type of radioligand used to measure binding. Clear identification of GH-binding activity was demonstrated with [125I]ovine GH (oGH), but specific binding could not be measured with [125I]human GH. The novel specificity was also shared by guinea pig liver membrane GH receptor (GHR) and cytosol GHBP, suggesting structural similarity in the GH-binding domain between the GHR and soluble GHBPs. The binding of oGH was dependent on serum concentration (5 microl serum produced 16.03 +/- 0.5% specific binding; mean +/- SEM; n = 11) and incubation time (equilibrium was reached by approximately 6 h at 21 C) and was completely reversible (t(1/2), approximately 2 h). Scatchard analysis revealed linear plots with an affinity constant (Ka) of 0.59 +/- 0.09 x 10(9) M(-1) and a capacity of 23,181 +/- 4,474 fmol/ml serum. Similar association constants were obtained for liver membrane GHR (0.79 +/- 0.22 x 10(9) M(-1)) and cytosol GHBPs (0.99 +/- 0.15 x 10(9) M(-1)), but the capacity, when expressed as femtomoles per g tissue, was significantly increased (4-fold) in cytosol (4,303 +/- 505) over that in membranes (1,071 +/- 257). There was no sex difference in Ka or level of GHBP in guinea pig serum. Surprisingly, the level of GH-binding activity was very low to undetectable in pregnant guinea pig serum. Characterization of the native structure of guinea pig GHBPs has indicated the presence of several proteins that are structurally distinct. Although the distribution of GH-binding activity covered a large Mr range (approximately 70-350 kDa) the major form of the circulating GHBP identified by gel chromatography had an apparent native Mr of 150-170 kDa. Partially purified GHBP (approximate Mr, 170 kDa) was covalently cross-linked to [125I]oGH and subjected to nonreducing SDS-PAGE. Specific GHBP complexes of 158 and 85 kDa were detected, suggesting that the partially purified GHBP complex may be composed of a smaller GHBP associated noncovalently with a non-GH-binding protein. "Pore limit" native PAGE (cathodic and anodic) revealed the presence of specific GHBPs of 363, 158, 74, and 55 kDa, which cross-hybridized with the rat liver membrane GHR monoclonal antibody mAb 263 but not with the rat serum GHBP-specific mAb 4.3. Interestingly, although GH binding was undetectable in pregnant guinea pig serum, Western immunoblot analysis with mAb 263 demonstrated the presence of a major immunoreactive GHBP band of 105 kDa in addition to 158- and 55-kDa GHBPs. The data indicate that the GHBPs are immunologically related to the rat membrane GHR, but provide no evidence to support the presence of a hydrophilic tail sequence homologous to that in the rat GHBP. These studies have identified in guinea pig serum GHBPs that exhibit novel ligand specificity, structural heterogeneity, and an immunological relationship to the rat liver membrane GHR. The identification of serum GHBP and the novel ligand specificity, which is also expressed by the liver membrane GHR, argue against the view that the guinea pig has a defective GHBP.


Assuntos
Proteínas de Transporte/sangue , Cobaias/sangue , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Cromatografia em Gel , Reagentes de Ligações Cruzadas , Feminino , Hormônio do Crescimento/metabolismo , Hormônio do Crescimento Humano/metabolismo , Humanos , Ligantes , Fígado/metabolismo , Masculino , Peso Molecular , Gravidez , Ligação Proteica , Ratos , Especificidade da Espécie
20.
J Biol Chem ; 272(8): 5024-30, 1997 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-9030565

RESUMO

The hepatic expression and serum levels of insulin-like growth factor-binding protein-3 (IGFBP-3) are decreased in insulin-dependent and insulin-resistant diabetes. Insulin increases hepatic IGFBP-3 expression by enhancing gene transcription. This report identifies sequences within the IGFBP-3 promoter that are necessary and sufficient for the response to insulin in hepatic nonparenchymal cells. By transient transfection, we mapped the insulin response element to the -1150 to -1124 base pair (bp) region of the rat IGFBP-3 promoter. Three tandem repeats of the -1150 to -1117 bp region conferred insulin responses in a heterologous promoter. Gel shift analyses revealed a 3-fold increase in DNA-protein complex formation with nuclear extracts obtained from insulin-stimulated nonparenchymal cells compared with cells incubated without insulin and revealed 3-4-fold decrease in complex formation with nuclear extracts obtained from the livers of streptozotocin-diabetic rats compared with control rats. Mutational analysis of this 34-bp region showed a core sequence of 10 bp (-1148 to -1139) that is critical for interaction with insulin-induced trans-acting factors. Southwestern blotting revealed a approximately 90-kDa protein that was increased 2-3-fold by the addition of insulin. Thus, we have identified cis-acting DNA sequences that are responsible for regulation of IGFBP-3 transcription by insulin and essential for binding of insulin-responsive nuclear factors.


Assuntos
Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Insulina/metabolismo , Animais , Sítios de Ligação/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...