11 research outputs found

    Intraepithelial Alterations In The Guinea Pig Lateral Prostate At Different Ages After Estradiol Treatment

    No full text
    The prostate is an accessory gland of the mammal reproductive system with great volume and high functional importance. Many works infer that, in addition to the androgenic ones, the estrogen can be associated with benign prostatic hyperplasia and prostatic cancer, but no conclusive evidence exists on the role of estrogen in normal prostatic and neoplastic tissue. The objective of this work was to evaluate the effects of chronic administration of estradiol benzoate on the lateral prostate of guinea pigs in the pre-pubescent, pubescent, post-pubescent and adult phases, with emphasis on the modifications provoked by this hormone on the glandular epithelium. The analyses of the estradiol-treated and control groups were investigated using histological procedures and transmission electron microscopy. The histopathological analysis of the lateral prostate in the treated group revealed areas where epithelial dysplasia was observed, assuming at some places a pattern of epithelial stratification characteristic of prostatic intraepithelial neoplasia. After ultrastructural analysis, the following were observed: enlargement of the internal membranes, heterogeneity in the cellular types, hypertrophy of the basal cells and apparent decrease of cytoplasmic organelles in some cells of the prostatic intraepithelial neoplasia. Still, a loss of cellular polarity was observed, along with nuclei of various forms, sizes and heights - as well as irregular chromatin distribution patterns. Such alterations were found mainly in pubescent, post-pubescent and adult animals subject to the chronic administration of estradiol. These findings reinforce the already existent data in understanding the role of estrogen in the etiology of prostatic diseases.362141148Bosland, M.C., The role of steroid hormones in prostate carcinogenesis (2000) J. Natl. Cancer Inst. Monogr., 27, pp. 39-66Bosland, M.C., Ford, H., Horton, L., Induction at high incidence of ductal prostate adenocarcinomas in NBL/Cr and Sprague-Dawley Hsd: SD rats treated with a combination of testosterone and estradiol-17B or diethylstilbestrol (1995) Carcinogenesis, 16, pp. 1311-1317Burfeind, P., Chernicky, C.L., Rinisland, F., Ilan, J., Ilan, J., Antisense RNA to the type I Insulin-like growth factor receptor supresses tumor growth and prevents invasion by rat prostate cancer cells in vivo (1996) Proc. Natl. Acad. Sci. USA, 93, pp. 7263-7268Carvalho, H.F., Recco-Pimentel, S.M., (2001) A Célula 2001. 1st. Ed., 287p. , Edit. Manole, São Paulo, BrasilChan, J.M., Stampfer, M.J., Giovanucci, E., Gann, P.H., Ma, J., Wilkinson, P., Hennekens, C.H., Pollak, M., Plasma insulin-like growth factor-I and prostate cancer risk: A prospective study (1998) Science, 279, pp. 563-566Corrales, J.J., Kadohama, N., Chai, L.S., Hoisaeter, P.A., Hampton, M.T., Murphy, G.P., Sandberg, A.A., Fluid imbibition as a factor in estrogen-induced increase of prostate weight in castrated rats (1981) Prostate, 2, pp. 337-358Cotta-Pereira, G., Rodrigo, F.G., David-Ferreira, J.F., The use of tannic acid-glutaraldehyde in the study of elastic related fibers (1976) Stain. Technol., 51, pp. 7-11De Carvalho, H.F., Lino Neto, J., Taboga, S.R., Microfibrils: Neglected components of pressure-bearing tendons (1994) Ann. Anat., 176, pp. 155-159De Marzo, A.M., Nelson, W.G., Meeker, A.K., Coffey, D.S., Stem cell features of benign and malignant prostate epithelial cells (1998) J. Urol., 160, pp. 2381-2392Di Giovanni, J., Kiguchi, K., Frijhoff, A., Wilker, E., Bol, D.K., Beltran, L., Moats, S., Conti, C., Deregulated expression of insulin-like growth factor 1 in prostate epithelium leads to neoplasia in transgenic mice (2000) Proc. Natl. Acad. Sci. USA, 97, pp. 3455-3460Droller, M.J., Medical approaches in the management of prostatic disease (1997) Br. J. Urol., 79 (2 SUPPL.), pp. 42-52Echeverria, O., Effects of testosterone on nuclear ribonucleoprotein components of prostate epithelial cells (1991) Biol. Cell., 72, pp. 223-229Glauert, A.M., (1975) Fixation, Dehydration and Embedding of Biological Specimens. Pratical Methods in Electron Microscopy, 3, pp. 1-207. , Glauert A.M. ed., N. Holland Publishing Company, NYHo, S.M., Lee, K.F., Lane, K., Neoplastic transformation of the prostate (1997) Prostate: Basic and Clinical Aspects, pp. 73-113. , Naz R.K. ed., CRC. Press, Boca RatonHorsfall, D.J., Mayne, K., Ricciardelli, C., Rao, M., Skinner, J.M., Henderson, D.W., Marshall, V.R., Tilley, W.D., Age-related prostatic stroma in guinea pig (1994) Lab. Invest., 70, pp. 753-763Huggins, C., Hodges, C.V., Studies on prostatic cancer. I. The effect of castration, of estrogen and androgen injection on serum phosphatases in metastatic carcinoma of the prostate (1972) CA. Cancer J. Clin., 22, pp. 232-240Huynh, H., Alpert, L., Alaoui-Jamali, M.A., Ng, C.Y., Chan, T.W.M., Co-administration of finasteride and the pure anti-oestrogen ICI 182,780 act synergistically in modulating the IGF system in rat prostate (2001) J. Endocr., 171, pp. 109-118Lane, K.E., Leav, I., Ziar, J., Bridges, R.S., Rand, W.M., Ho, S.M., Suppression of testosterone and estradiol-17Beta-induced dysplasia in the dorsolateral prostate of Noble rats by bromocriptine (1997) Carcinogenesis, 18, pp. 1505-1510Leav, I., Merk, F.B., Ofner, P., Goodrich, G., Kwan, P.W., Stein, B.M., Sar, M., Stumpf, W.E., Bipotentiality of response to sex hormones by the prostate of castrated or hypophysectomized dogs. Direct effects of estrogen (1978) Am. J. Pathol., 93, pp. 69-92Leav, L., Ho, S.M., Ofner, P., Merk, F.B., Kwan, P.W.L., Damassa, D., Biochemical alterations in sex hormone induced hyperplasia and dysplasia of the dorsolateral prostates of Noble rats (1988) J. Natl. Cancer Inst., 80, pp. 1045-1053Levine, A.C., Kirschenbaum, A., Droller, M., Gabrilove, J.L., Effect of the addition of estrogen to medical castration on prostatic siza, symptoms, histology and serum prostate specific antigen in 4 men with benign prostatic hypertrophy (1991) J. Urol., 146, pp. 790-793McNeal, J.E., Alroy, J., Leav, I., Redwine, E.A., Freiha, F.S., Stamey, T.A., Immunohistochemical evidence for impaired cell differentiation in the premalignant phase of prostate carcinogenesis (1988) Am. J. Clin. Pathol., 90, pp. 23-32Montenegro, M.R., Franco, M., (1995) Patologia: Processos Gerais. 3rd Ed., 263p. , Editora Atheneu. São PauloNeil, J.D., Freeman, M.E., Tillson, S.A., Control of the proestrus surge of prolactin in luteineizing hormone secretion by estrogen in the rat (1971) Endocrinology, 98, pp. 1448-1453Pylkkanen, L., Makela, S., Santti, R., Animal models for the preneoplastic lesions of the prostate (1996) Eur. Urol., 30, pp. 243-248Pylkkanen, L., Makela, S., Valve, E., Harkonen, P., Toikkanen, S., Santti, R., Prostatic dysplasia associated with increased expression of C-MYC in neonatally estrogenized mice (1993) Journ. Urol., 149, pp. 1593-1601Rhodes, L., Ding, V.D., Kemp, R.K., Khan, M.S., Nakhla, A.M., Pikounis, B., Rosner, W., Feeney, W.P., Estradiol causes a dose-dependent stimulation of prostate growth in castred beagle dogs (2000) Prostate, 44, pp. 8-18Santti, R., Newbold, R.R., Makela, S., Pylkkanen, L., McLachlan, J.A., Developmental estrogenization and prostatic neoplasia (1994) Prostate, 24, pp. 67-78Schacht, M.J., Niederberger, C.S., Garnett, J.E., Sensibar, J.A., Lee, C., Grayhack, J.T., A local direct effect of pituitary graft on growth of the lateral prostate in rats (1992) Prostate, 20, pp. 51-58Shibata, M.A., Ward, J.M., Devor, D.E., Liu, M.L., Green, J.E., Progression of prostatic intraepithelial neoplasia to invasive carcinoma in C3(1)/SV40 large T antigen transgenic mice: Histopathological and molecular biological alterations (1996) Cancer Res., 56, pp. 4894-4903Tam, C.C., Wong, Y.C., Ultrastructural study of the effects of 17β-Oestradiol on the lateral prostate and seminal vesicle of the castred Guinea pig (1991) Acta Anat., 141, pp. 51-62Thompson, A.S., Rowley, D.R., Heidger Jr., P.M., Effects of estrogen upon the fine structure of epithelium and stroma in the rat ventral prostate gland (1979) Invest. Urol., 17, pp. 83-89Weibel, E.R., Principles and methods for the morphometric study of the lung and other organs (1978) Lab. Invest., 12, pp. 131-15

    Testosterone Therapy Differently Regulates The Anti- And Pro-inflammatory Cytokines In The Plasma And Prostate Of Rats Submitted To Chronic Ethanol Consumption (uchb)

    No full text
    Problem: Ethanol consumption damages the prostate, and testosterone is known by anti-inflammatory role. Methods of Study: The cytokines were investigated in the plasma and ventral prostate of UChB rats submitted or not to testosterone therapy by ELISA and Western blot, respectively. Additionally, inflammatory foci and mast cells were identified in the ventral prostate slides stained by hematoxylin and eosin and toluidine blue, respectively. Results: Inflammatory foci were found in the ethanol-treated animals and absent after testosterone therapy. Plasma levels of IL-6 and IL-10 were not changed while TNFα and TFG-β1 were increased in the animals submitted testosterone therapy. Regarding to ventral prostate, IL-6 did not alter, while IL-10, TNFα, and TFG-β1 were increased after testosterone therapy. Ethanol increases NFR2 in addition to high number of intact and degranulated mast cell which were reduced after testosterone therapy. Conclusions: So, ethanol and testosterone differentially modulates the cytokines in the plasma and prostate. © 2014 John Wiley & Sons A/S

    Key Participants Of The Tumor Microenvironment Of The Prostate: An Approach Of The Structural Dynamic Of Cellular Elements And Extracellular Matrix Components During Epithelial-stromal Transition

    No full text
    Cancer is a multistep process that begins with the transformation of normal epithelial cells and continues with tumor growth, stromal invasion and metastasis. The remodeling of the peritumoral environment is decisive for the onset of tumor invasiveness. This event is dependent on epithelial-stromal interactions, degradation of extracellular matrix components and reorganization of fibrillar components. Our research group has studied in a new proposed rodent model the participation of cellular and molecular components in the prostate microenvironment that contributes to cancer progression. Our group adopted the gerbil Meriones unguiculatus as an alternative experimental model for prostate cancer study. This model has presented significant responses to hormonal treatments and to development of spontaneous and induced neoplasias. The data obtained indicate reorganization of type I collagen fibers and reticular fibers, synthesis of new components such as tenascin and proteoglycans, degradation of basement membrane components and elastic fibers and increased expression of metalloproteinases. Fibroblasts that border the region, apparently participate in the stromal reaction. The roles of each of these events, as well as some signaling molecules, participants of neoplastic progression and factors that promote genetic reprogramming during epithelial-stromal transition are also discussed.1171413Adisetiyo, H., Liang, M., Liao, C.P., Jeong, J.H., Cohen, M.B., Roy-Burman, P., Dependence of castration-resistant prostate cancer (CRPC) stem cells on CRPC-associated fibroblasts (2014) J Cell Physiol, 229, pp. 1170-1176Ardi, V.C., Kupriyanova, T.A., Deryugina, E.I., Quigley, J.P., Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis (2007) Proc Natl Acad Sci, 104, pp. 20262-20267Bacac, M., Provero, P., Mayran, N., Stehle, J.C., Fusco, C., Stamenkovic, I., (2006) A mouse stromal response to tumor invasion predicts prostate and breast cancer patient survival PLoS One, , 101371/journalpone0000032Bauvois, B., New facets of matrix metalloproteinases MMP-2 and MMP-9 as cell surface transducers: outside-in signaling and relationship to tumor progression (2012) Biochim Biophys Acta, 1825, pp. 29-36Berry, P.A., Maitland, N.J., Collins, A.T., Androgen receptor signalling in prostate: effects of stromal factors on normal and cancer stem cells (2008) Mol Cell Endocrinol, 288, pp. 30-37Bhowmick, N.A., Moses, H.L., Tumor-stroma interactions (2005) Curr Opin Genet Dev, 15, pp. 97-101Bissell, M.J., Hines, W.C., Why don't we get more cancer?. A proposed role of the microenvironment in restraining cancer progression (2011) Nat Med, 17, pp. 320-329Bruni-Cardoso, A., Rosa-Ribeiro, R., Pascoal, V.D., De Thomaz, A.A., Cesar, C.L., Carvalho, H.F., MMP-2 regulates rat ventral prostate development in vitro (2010) Dev Dyn, 239, pp. 737-746Campos, S.G., Zanetoni, C., Scarano, W.R., Vilamaior, P.S., Taboga, S.R., Age-related histopathological lesions in the Mongolian gerbil ventral prostate as a good model for studies of spontaneous hormone-related disorders (2008) Int J Exp Pathol, 89, pp. 13-24Campos, S.G., Gonçalves, B.F., Scarano, W.R., Corradi, L.S., Santos, F.C., Custodio, A.M., Tissue changes in senescent gerbil prostate after hormone deprivation leads to acquisition of androgen insensitivity (2010) Int J Exp Pathol, 91, pp. 394-407Campos, S.G.P., Gonçalves, B.F., Scarano, W.R., Ribeiro, D.L., Goes, R.M., Taboga, S.R., Prostatic stromal cells of old gerbils respond to steroidal blockades creating a microenvironment similar to reactive stroma (2011) Biomed Aging Pathol, 1, pp. 97-106Carvalho, H.F., Vilamaior, P.S.L., Taboga, S.R., Elastic system of the ventral prostate and its modifications following orchiectomy (1997) Prostate, 32, pp. 27-34Celià-Terrassa, T., Meca-Cortés, O., Mateo, F., de Paz, A.M., Rubio, N., Arnal-Estapé, A., Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells (2012) J Clin Invest, 122, pp. 1849-1868Cheng, J.C., Leung, P.C., Type I collagen down-regulates E-cadherin expression by increasing PI3KCA in cancer cells (2011) Cancer. Lett., 304, pp. 107-116Chung, L., Baseman, A., Assikis, V., Zhau, H., Molecular insights into prostate cancer progression: the missing link of tumor microenvironment (2005) J Urol, 173, pp. 10-20Cirri, P., Chiarugi, P., Cancer-associated-fibroblasts and tumour cells: a diabolic liaison driving cancer progression (2012) Cancer Metastasis Ver, 31, pp. 195-208Clark, A.K., Taubenberger, A.V., Taylor, R.A., Niranjan, B., Chea, Z.Y., Zotenko, E., A bioengineered microenvironment to quantitatively measure the tumorigenic properties of cancer-associated fibroblasts in human prostate cancer (2013) Biomaterials, 34, pp. 85-4777Cordeiro, R.S., Scarano, W.R., Campos, S.G., Santos, F.C., Vilamaior, P.S., Góes, R.M., Androgen receptor in the Mongolian gerbil ventral prostate: evaluation during different phases of postnatal development and following androgen blockage (2008) Micron, 39, pp. 1312-1324Corn, P.G., The tumor microenvironment in prostate cancer: elucidating molecular pathways for therapy development (2012) Cancer Manage Res, 4, pp. 183-193Corradi, L.S., Goes, R.M., Carvalho, H.F., Taboga, S.R., Inhibition of 5-alpha reductase activity induces stromal remodeling and smooth muscle differentiation in adult gerbil ventral prostate (2004) Differentiation, 72, pp. 198-208Corradi, L.S., Góes, R.M., Vilamaior, P.S., Taboga, S.R., Increased androgen receptor and remodeling in the prostatic stroma after the inhibition of 5-alpha reductase and aromatase in gerbil ventral prostate (2009) Microsc Res Tech, 72, pp. 939-950Cunha, G.R., Hayward, S.W., Dahiya, R., Foster, B.A., Smooth muscle-epithelial interactions in normal and neoplastic prostatic development (1996) Acta Anat (Basel), 155, pp. 63-72Custodio, A.M., Santos, F.C., Campos, S.G., Vilamaior, P.S.L., Góes, R.M., Taboga, S.R., Aging effects on the mongolian gerbil female prostate (Skene's paraurethral glands): structural, ultrastructural, quantitative, and hormonal evaluations (2008) Anat Rec (Hoboken), 291, pp. 463-474De Wever, O., Mareel, M., Role of tissue stroma in cancer cell invasion (2003) J Pathol, 200, pp. 429-447Egeblad, M., Werb, Z., New functions for the matrix metalloproteinases in cancer progression (2002) Nat Rev Cancer, 2, pp. 61-74Fávaro, W.J., Hetzl, A.C., Reis, L.O., Ferreira, U., Billis, A., Cagnon, V.H., Periacinar retraction clefting in nonneoplastic and neoplastic prostatic glands: artifact or molecular involvement (2012) Pathol Oncol Res, 18, pp. 285-292Fülöp, T., Jacob, M.P., Wallach, J., Hauck, M., Seres, I., Varga, Z., The elastin-laminin receptor (2001) J Soc Biol, 195, pp. 157-164Fülöp, T., Larbi, A., Putative role of 67kDa elastin-laminin receptor in tumor invasion (2002) Semin Cancer Biol, 12, pp. 219-229Fülöp, T., Khalil, A., Larbi, A., The role of elastin peptides in modulating the immune response in aging and age-related diseases (2012) Pathol Biol, 60, pp. 28-33Gobbo, M.G., Taboga, S.R., Ribeiro, D.L., Góes, R.M., Short-term stromal alterations in the rat ventral prostate following alloxan-induced diabetes and the influence of insulin replacement (2012) Micron, 43, pp. 326-333Gonçalves, B.F., Zanetoni, C., Scarano, W.R., Góes, R.M., Vilamaior, P.S.L., Taboga, S.R., Prostate carcinogenesis induced by N-methyl-N-nitrosourea (MNU) in gerbils: histopathological diagnosis and potential invasiveness mediated by extracellular matrix components (2010) Exp Mol Pathol, 88, pp. 96-106Gonçalves, B.F., Campos, S.G.P., Zanetoni, C., Scarano, W.R., Falleiros Júnior, L.R., Amorin, R.L., A new proposed rodent model of chemically induced prostate carcinogenesis: distinct time-course prostate cancer progression in the dorsolateral and ventral lobes (2013) Prostate, 73, pp. 1202-1213Gordon, M., Hahn, R., Collagens (2010) Cell Tissue Res, 339, pp. 247-257Goulas, A., Hatzichristou, D.G., Karakiulakis, G., Mirtsou-Fidani, V., Kalinderis, A., Papakonstantinou, E., Benign hyperplasia of the human prostate is associated with tissue enrichment in chondrotin sulphate of wide size distribution (2000) Prostate, 44, pp. 104-110Hanahan, D., Weinberg, R.A., Hallmarks of cancer: the next generation (2011) Cell, 144, pp. 646-674Hance, M.W., Dole, K., Gopal, U., Bohonowych, J.E., Jezierska-Drutel, A., Neumann, C.A., Secreted Hsp90 is a novel regulator of the epithelial to mesenchymal transition (EMT) in prostate cancer (2012) J Biol Chem, 287, pp. 37732-37744Haslam, S.Z., Woodward, T.L., Host microenvironment in breast cancer development: epithelial-cell-stromal-cell interactions and steroid hormone action in normal and cancerous mammary gland (2003) Breast Cancer Res, 5, pp. 208-215Hatfield, K.J., Reikvam, H., Bruserud, O., The crosstalk between the matrix metalloprotease system and the chemokine network in acute myeloid leukemia (2010) Curr Med Chem, 17, pp. 4448-4461Hayashi, N., Sugimura, Y., Kamamura, J., Doncajour, A.A., Cunha, G.R., Morphological and functional hetereogeneity in the rat prostatic gland (1991) Biol Reprod, 45, pp. 308-321Hensley, P.J., Kyprianou, N., Modeling prostate cancer in mice: limitations and opportunities (2012) J Androl, 33, pp. 133-144Hua, H., Li, M., Luo, T., Yin, Y., Jiang, Y., Matrix metalloproteinases in tumorigenesis an evolving paradigm (2011) Cell Mol Life Sci, 68, pp. 3853-3868Iyoda, T., Fukai, F., Modulation of tumor cell survival, proliferation, and differentiation by the peptide derived from tenascin-C: implication of β1-integrin activation (2012) Int J Cell Biol, p. 647594. , 101155/2012/647594Johansson, J.E., Holmberg, L., Johansson, S., Bergstrom, R., Adami, H.O., Fifteen-year survival in prostate cancer A prospective, population-based study in Sweden (1997) JAMA, 277, pp. 467-471Justulin, L.A., Acquaro, C., Carvalho, R.F., Silva, M.D., Felisbino, S.L., Combined effect of the finasteride and doxazosin on rat ventral prostate morphology and physiology (2010) Int J Androl, 33, pp. 489-499Justulin, L.A., Della-Coleta, H.H., Taboga, S.R., Felisbino, S.L., Matrix metalloproteinase (MMP)-2 and MMP-9 activity and localization during ventral prostate atrophy and regrowth (2010) Int J Androl, 33, pp. 696-708Kalluri, R., Basement membranes: structure, assembly and role in tumour angiogenesis (2003) Nat Rev Cancer, 3, pp. 422-433Kalluri, R., Han, Y., Targeting TGF-β and the extracellular matrix in Marfan's syndrome (2008) Dev Cell, 15, pp. 1-2Katenkamp, K., Berndt, A., Hindermann, W., Wunderlich, H., Haas, K.M., Borsi, L., MRNA expression and protein distribution of the unspliced tenascin-C isoform in prostatic adenocarcinoma (2004) J Pathol, 203, pp. 771-779Kessenbrock, K., Plaks, V., Werb, Z., Matrix metalloproteinases: regulators of the tumor microenvironment (2010) Cell, 141, pp. 52-67Kosir, M.A., Quinn, C.C., Sorting of heparan sulfate proteoglycan into matrix compartments of prostate adenocarcinoma cells (1995) J Surg Res, 58, pp. 46-52Kraning-Rush, C.M., Califano, J.P., Reinhart-King, C.A., Cellular traction stresses increase with increasing metastatic potential (2012) PLoS One, , 101371/journalpone0032572Kunz-Schughart, L.A., Knuechel, R., Tumor-associated fibroblasts (part I): Active stromal participants in tumor development and progression? (2002) Histol Histopathol, 17, pp. 599-621Kunz-Schughart, L.A., Knuechel, R., Tumor-associated fibroblasts (part II): Functional impact on tumor tissue (2002) Histol Histopathol, 17, pp. 623-637LeBleu, V.S., MacDonald, B., Kalluri, R., Structure and function of basement membranes (2007) Exp Biol Med, 232, pp. 1121-1129Littlepage, L.E., Egeblad, M., Werb, Z., Coevolution of cancer and stromal cellular responses (2005) Cancer Cell, 7, pp. 499-500Menon, S., Beningo, K.A., Cancer cell invasion is enhanced by applied mechanical stimulation (2011) PLoS One, 6, p. e17277Micalizzi, D.S., Farabaugh, S.M., Ford, H.L., Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression (2010) J Mammary Gland Biol Neoplasia, 15, pp. 117-134Morrison, C., Thornhill, J., Gaffney, E., The connective tissue framework in the normal prostate, BPH and prostate cancer: analysis by scanning electron microscopy after cellular digestion (2000) Urol Res, 28, pp. 304-307Murphy, G., The ADAMs: signaling scissors in the tumour microenvironment (2008) Nat Rev Cancer, 8, pp. 932-941Murphy-Ullrich, J.E., The de-adhesive activity of matricellular proteins: is intermediate cell adhesion an adaptive state? (2001) J Clin Invest, 107, pp. 785-790Narunsky, L., Oren, R., Bochner, F., Neeman, M., Imaging aspects of the tumor stroma with therapeutic implications (2014) Pharmacol Ther., 141, pp. 192-208Nemeth, J.A., Lee, C., Prostatic ductal system in rats: regional variation in stromal organization (1996) Prostate, 28, pp. 124-128Oliveira, S.M., Leite Vilamaior, P.S., Corradi, L.S., Góes, R.M., Taboga, S.R., Cellular and extracellular behavior in the gerbil (Meriones unguiculatus) ventral prostate following different types of castration and the consequences of testosterone replacement (2007) Cell Biol Int, 31, pp. 235-245Parsons, D.F., Tumor cell interactions with stromal elastin and type I collagen: the consequences of specific adhesion and proteolysis (1993) Tumour Biol, 14, pp. 137-143Paulsson, M., Basement membrane proteins: structure,assembly, and cellular interaction (1992) Crit. Rev. Biochem. Mol. Biol., 27, pp. 93-127Pegorin de Campos, S.G., Zanetoni, C., Goes, R.M., Taboga, S.R., Biological behavior of the gerbil ventral prostate in three phases of postnatal development (2006) Anat Rec, A: Discovery Mol Cell Evol Biol, 288, pp. 723-733Pezzato, E., Sartor, L., Dell'Aica, I., Dittadi, R., Gion, M., Belluco, C., Prostate carcinoma and green tea: PSA-triggered basement membrane degradation and MMP-2 activation are inhibited by (-)epigallocatechin-3-gallate (2004) Int J Cancer, 112, pp. 787-792Pienta, K.J., Abate-Shen, C., Agus, D.B., Attar, R.M., Chung, L.W., Greenberg, N.M., The current state of preclinical prostate cancer animal models (2008) Prostate, 68, pp. 629-639Pietras, K., Ostman, A., Hallmarks of cancer: interactions with the tumor stroma (2010) Exp Cell Res, 316, pp. 1324-1331Prud'homme, G.J., Pathobiology of transforming growth factor β in cancer, fibrosis and immunologic disease, and therapeutic considerations (2007) Lab Invest, 87, pp. 1077-1091Reich, R., Thompson, E.W., Iwamoto, Y., Martin, G.R., Deason, J.R., Fuller, G.C., Effects of inhibitors of plasminogen activator, serine proteinases, and collagenase IV on the invasion of basement membranes by metastatic cells (1988) Cancer Res, 48, pp. 3307-3312Ribeiro, D.L., Marques, S.F., Alberti, S., Spadella, C.T., Manzato, A.J., Taboga, S.R., Malignant lesions in the ventral prostate of alloxan-induced diabetic rats (2008) Int J Exp Pathol, 89, pp. 276-283Ribeiro, D.L., Taboga, S.R., Góes, R.M., Diabetes induces stromal remodelling and increase in chondroitin sulphate proteoglycans of the rat ventral prostate (2009) Int J Exp Pathol, 90, pp. 400-411Ricard-Blum, S., The collagen family (2011) Cold Spring Harbor Perspect Biol, 3, p. a004978Ricard-Blum, S., Ballut, L., Matricryptins derived from collagens and proteoglycans (2011) Front Biosci (Landmark Ed), 16, pp. 674-697Rochel, S.S., Bruni-Cardoso, A., Taboga, S.R., Vilamaior, P.S., Góes, R.M., Lobe identity in the Mongolian gerbil prostatic complex: a new rodent model for prostate study (2007) Anat Rec (Hoboken), 290, pp. 1233-1247Rowe, R.G., Weiss, S.J., Navigating ECM barriers at the invasive front: the cancer cell-stroma interface (2009) Annu Rev Cell Dev Biol, 25, pp. 567-595Roy, R., Yang, J., Moses, M.A., Matrix metalloproteinases as novel biomarkers and potential therapeutic targets in human cancer (2009) J Clin Oncol, 27, pp. 5287-5297Sakko, A.J., Butler, M.S., Byers, S., Reinboth, B.J., Stahl, J., Kench, J.G., Immunohistochemical level of unsulfated chondroitin disaccharides in the cancer stroma is an independent predictor of prostate cancer relapse (2008) Cancer Epidemiol Biomarkers Prev, 17, pp. 2488-2497Santos, F.C.A., Carvalho, H.F., Góes, R.M., Taboga, S.R., Structure, histochemistry and ultrastructure of the epithelium and stroma in the gerbil (Meriones unguiculatus) female prostate (2003) Tissue Cell, 35, pp. 447-457Santos, F.C., Leite, R.P., Custodio, A.M., Carvalho, K.P., Monteiro-Leal, L.H., Santos, A.B., Testosterone stimulates growth and secretory activity of the female prostate in the adult gerbil (Meriones unguiculatus) (2006) Biol Reprod, 75, pp. 370-379Scarano, W.R., Vilamaior, P.S., Taboga, S.R., Tissue evidence of the testosterone role on the abnormal growth and aging effects reversion in the gerbil (Meriones unguiculatus) prostate (2006) Anat Rec, A: Discovery Mol Cell Evol Biol, 288, pp. 1190-2000Scarano, W.R., Sousa, D.E., Campos, S.G., Corradi, L.S., Vilamaior, P.S.L., Taboga, S.R., Oestrogen supplementation following castration promotes stromal remodelling and histopathological alterations in the Mongolian gerbil ventral prostate (2008) Int J Exp Pathol, 89, pp. 25-37Seandel, M., Noack-Kunnmann, K., Zhu, D., Aimes, R.T., Quigley, J.P., Growth factor induced angiogenesis in vivo requires specific cleavage of fibrillar type I collagen (2001) Blood, 97, pp. 2323-2332Sprenger, C.C., Plymate, S.R., Reed, M.J., Aging-related alterations in the extracellular matrix modulate the microenvironment and influence tumor progression (2010) Int J Cancer, 127, pp. 2739-2748Suwiwat, S., Ricciardelli, C., Tammi, R., Tammi, M., Auvinen, P., Kosma, V.M., Expression of extracellular matrix components versican, chondroitin sulfate, tenascin, and hyaluronan, and their association with disease outcome in node-negative breast cancer (2004) Clin Cancer Res, 10, pp. 2491-2498Taboga, S.R., Vidal, B.C., Collagen fibers in human prostatic lesions: histochemistry and anisotropies (2003) J Submicrosc Cytol Pathol, 35, pp. 11-16Taboga, S.R., Scortegagna, E., Siviero, M.P., Carvalho, H.F., Anatomy of smooth muscle cells in nonmalignant and malignant human prostate tissue (2008) Anat Rec (Hoboken), 291, pp. 1115-1123Tanjore, H., Kalluri, R., The role of type IV collagen and basement membranes in cancer progression and metastasis (2006) Am J Pathol, 168, pp. 715-717Taylor, M.A., Parvani, J.G., Schiemann, W.P., The pathophysiology of epithelial-mesenchymal transition induced by transforming growth factor-β in normal and malignant mammary epithelial cells (2010) J Mammary Gland Biol Neoplasia, 15, pp. 169-190Taylor, R.A., Risbridger, G.P., Prostatic tumor stroma a key player in cancer progression (2008) Curr Cancer Drug Targets, 8, pp. 490-497Thiery, J.P., Acloque, H., Huang, R.Y., Nieto, M.A., Epithelial-mesenchymal transitions in development and disease (2009) Cell, 139, pp. 871-890Timpl, R., Macromolecular organization of basement membranes (1996) Curr Opin Cell Biol, 8, pp. 618-624Tlsty, T.D., Coussens, L.M., Tumor stroma and regulation of cancer development (2005) Annu Rev Pathol Mech Dis, 1, pp. 119-150Tomas, D., Ulamec, M., Hudolin, T., Bulimbasić, S., Belicza, M., Kruslin, B., Myofibroblastic stromal reaction and expression of tenascin-C and laminin in prostate adenocarcinoma (2006) Prostate Cancer Prostatic. Dis., 9, pp. 414-419Tuxhorn, J.A., Ayala, G.E., Rowley, D.R., Reactive stroma in prostate cancer progression (2001) J Urol, 166, pp. 2472-2483Valastyan, S., Weinberg, R.A., Tumor metastasis: molecular insights and evolving paradigms (2011) Cell, 147, pp. 275-292Van Hoorde, L., Van Aken, E., Mareel, M., Collagen type I: a substrate and a signal for invasion (2000) Prog Mol Subcell Biol, 25, pp. 105-134Vilamaior, P.S.L., Taboga, S.R., Carvalho, H.F., Modulation of smooth muscle cell function: morphological evidence for a contractile to synthetic transition in the rat ventral protate after castration (2005) Cell Biol Int, 29, pp. 809-816Wendt, M.K., Tian, M., Schiemann, W.P., Deconstructing the mechanisms andconsequences of TGF-induced EMT during cancer progression (2012) Cell Tissue Res., 347, pp. 85-101Yip, G.W., Smollich, M., Gotte, M., Therapeutic value of glycosaminoglycans in cancer (2006) Mol Cancer Ther, 5, pp. 2139-2148Yurchenco, P.D., Schittny, J.C., Molecular architecture of basement membranes (1990) FASEB J, 4, pp. 1577-1590Yurchenco, P.D., Smirnov, S., Mathus, T., Analysis of basement membrane self-assembly and cellular interactions with native and recombinant glycoproteins (2002) Methods Cell Biol, 69, pp. 111-144Zhang, J., Liu, J., Tumor stroma as targets for cancer therapy (2013) Pharmacol Ther, 137, pp. 200-215Zhang, L., Shi, J., Feng, J., Klocker, H., Lee, C., Zhang, J., Type IV collagenase (matrix metalloproteinase-2 and -9) in prostate cancer (2004) Prostate Cancer Prostatic Dis, 7, pp. 327-332Zou, M., Jiao, J., Zou, Q., Xu, Y., Cheng, M., Xu, J., Multiple metastases in a novel LNCaP model of human prostate cancer (2013) Oncol Rep, 30, pp. 615-62

    Androgen Receptor In The Mongolian Gerbil Ventral Prostate: Evaluation During Different Phases Of Postnatal Development And Following Androgen Blockage

    No full text
    The normal growth, differentiation and maintenance of the morphofunctional integrity of the prostate gland are dependent on the interaction of constant levels of androgens with their receptors. The need to study the responses to hormones under several conditions and the effect of their blockage is due to the fact that the human prostate is the site of a great number of age-related diseases, and the ones with a major medical importance are prostate cancer (CaP) and benign prostatic hyperplasia (BPH), which can both be treated with androgen suppression. Seventy-five male gerbils were divided, randomly, into 3 groups of 25 animals each, where each group corresponded to one phase of postnatal development. In each phase, it was possible to morphologically and stereologically analyze the compartments of prostatic ventral lobe, as well as to immunohistochemically analyze the degree of expression of androgen receptors (ARs) after the androgen blockage therapies. In addition, it was possible to establish the hormonal dosage of serum testosterone levels given the comparative approach of the expression of androgen receptors. There is a pattern of AR distribution in the prostatic ventral lobe throughout postnatal development, in which the younger the animal is the higher, the interaction of circulating androgens that stimulate the AR expression in both the epithelial and stromal compartments. The androgen blockage therapies decreased AR expression in the prostatic compartments, but the androgen reposition after these blockages was not sufficient to recover the glandular structure or stimulate the AR expression up to normal physiological conditions. Both the regulation and distribution of androgen receptors along the gerbil prostatic tissues are complex mechanisms that are likely to be genetically regulated by androgens prenatally or by other factors that are still unknown. This rodent species seems to be a valuable model in the attempt to improve the understanding of the morphophysiological and pathological behavior of this important gland in humans throughout aging and to stimulate new therapeutic ideas to fight prostate cancer. © 2008 Elsevier Ltd. All rights reserved.39813121324Antoniolli, E., Della-Colleta, H.H.M., Carvalho, H.F., Smooth muscle cell behavior in the ventral prostate of castrated rats (2004) J. Androl., 25 (1), pp. 50-56Banerjee, P.P., Banerjee, S., Brown, T.R., Increased androgen receptor expression correlates with development of age-dependent, lobe-specific spontaneous hyperplasia of the brown Norway rat prostate (2001) Endocrinology, 142 (9), pp. 4066-4075Berthold, D.R., Moore, M.J., Novel targets in prostate cancer (2006) Expert Opin. Ther. Target, 10 (5), pp. 777-780Bonkhoff, H., Remberger, K., Differentiation pathways and histogenetic aspects of normal and abnormal prostatic growth: a stem cell model (1996) Prostate, 28, pp. 98-106Brum, I.S., Spritzer, P.M., Brentani, M.M., Biologia Molecular das Neoplasias de Próstata (2005) Arq. Bras. Endocrinol. Metab., 49 (5), pp. 797-804Buchanan, G., Irvine, R.A., Coetzee, G.A., Tilley, W.D., Contribution of the androgen receptor to prostate cancer predisposition and progression (2001) Cancer Metastasis Rev., 20, pp. 207-223Carvalho, H.F., Line, S.R.P., Basement membrane associated changes in the rat ventral prostate following castration (1996) Cell Biol. Int., 20, pp. 809-819Carvalho, H.F., Taboga, S.R., Vilamaior, P.S.L., Collagen type VI is a component of the extracellular matrix microfibril network of the prostatic stroma (1997) Tissue Cell, 29, pp. 163-170Carvalho, H.F., Vilamaior, P.S.L., Taboga, S.R., Elastic system of the rat ventral prostate and its modifications following orchiectomy (1997) Prostate, 32, pp. 27-34Chan, T.Y., Mikolajczyk, S.D., Lecksell, K., Shue, M.J., Rittenhouse, H.G., Partin, A.W., Epstein, J.I., Immunohistochemical staining of prostate cancer with monoclonal antibodies to the precursor of prostate-specific antigen (2003) Urology, 62 (1), pp. 177-181Cordeiro, R.S., Scarano, W.R., Góes, R.M., Taboga, S.R., Tissue alterations in the guinea pig lateral prostate following antiandrogen flutamide therapy (2004) BioCell, 28 (1), pp. 21-30Corradi, L.S., Góes, R.M., Carvalho, H.F., Taboga, S.R., Inhibition of 5-α-reductase activity induces stromal remodeling and smooth muscle de-differentiation in adult gerbil ventral prostate (2004) Differentiation, 72 (5), pp. 198-208Culing, Z., Hobisch, A., Cronauer, M.V., Androgen receptor activation in prostatic tumor cell lines by insulin-like growth factor-I, keratinocyte growth factor, and epidermal growth factor (1994) Cancer Res., 54, pp. 5474-5478Cunha, G.R., Bigsby, R.M., Coople, P.S., Sugimura, Y., Stromal-epithelial interactions in adult organs (1985) Cell Diff., 17, pp. 137-148Cunha, G.R., Hayward, S.W., Dahiya, R., Foster, B.A., Smooth muscle-epithelial interactions in normal and neoplastic prostatic development (1996) Acta Anat., 155, pp. 63-72Dehm, S.M., Tindall, D.J., Molecular regulation of androgen action in prostate câncer (2006) J. Cell. Biochem., 99, pp. 333-344Dounjacour, A.A., Cunha, G.R., The effect of androgen deprivation on branching morphogenesis in the mouse prostate (1988) Dev. Biol., 128, pp. 1-14Farla, P., Hersmus, R., Trapman, J., Houtsmuller, A.B., Antiandrogens prevent stable DNA-binding of the androgen receptor (2005) J. Cell Sci., 118, pp. 4187-4198Feldman, B.J., Feldman, D., The development of androgen-independent prostate cancer (2001) Nat. Rev. Cancer, 1, pp. 34-45Foster, P.M.D., Harris, M.W., Changes in androgen-mediated reproductive development in male rat offspring following exposure to a single oral dose of flutamide at different gestational ages (2005) Toxicol. Sci., 85, pp. 1024-1032Gail, S.P., Lynn, B., Geoffrey, L.G., Androgen receptor localization in different cell types of the adult rat prostate (1991) Endocrinology, 129 (6), pp. 3187-3199Galbraith, S.M., Duchesne, G.M., Androgens and prostate cancer: biology, pathology and hormonal therapy (1997) Eur. J. Cancer, 33 (4), pp. 545-554Gao, W., Kim, J., Dalton, J.T., Pharmacokinetics and pharmacodynamics of nonsteroidal androgen receptor ligands (2006) Pharm. Res., 23 (8), pp. 1641-1658Goto, K., Koizumi, K., Takauri, H., Fojii, Y., Furuyama, Y., Saika, H., Suzuki, H., Suzuki, K., Effects of flutamide on sex maturation behavior of offspring born to male rats treated during late pregnancy (2004) J. Toxicol. Sci., 29 (5), pp. 517-534Grossmann, M.E., Huang, H., Tindall, D.J., Androgen receptor signaling in androgen-refractory prostate cancer (2001) J. Natl. Cancer Inst., 93 (22), pp. 1687-1697Hayashi, N., Sugimura, Y., Kamamura, J., Doncajour, A.A., Cunha, G.R., Morphological and functional heterogeneity in the rat prostatic gland (1991) Biol. Reprod., 45, pp. 308-321Hayward, S.W., Rosen, M.A., Cunha, G.R., Stromal-epithelial interactions in the normal and neoplasic prostate (1997) Br. J. Urol., 79 (SUPPL. 2), pp. 18-26Heinlein, C.A., Chang, C., Androgen receptor in prostate cancer (2004) Endocr. Rev., 25 (2), pp. 276-308Horsfall, D.J., Mayne, K., Ricciardelli, C., Rao, M., Skinner, J.M., Henderson, D.W., Marshall, V.R., Tilley, W.D., Age-related changes in guinea pig prostatic stroma (1994) Lab. Invest., 70, pp. 753-763Hudson, D.L., Guy, A.T., Fry, P., O'hare, M.J., Watt, F.M., Masters, J.N.R., Epithelial cell differentiation pathways in the human prostate: identification of intermediate phenotypes by keratin expression (2001) J. Histochem. Cytochem., 49 (2), pp. 271-278Hughes, I.A., Lim, H.N., Martin, H., Mongan, N.P., Dovey, L., Ahmed, S.F., Hawkins, J.R., Developmental aspects of androgen action (2001) Mol. Cell. Endocrinol., 185, pp. 33-41Huttunen, E., Romppanen, T., Helminen, H.J., A histoquantitative study on the effects of castration on the rat ventral prostate lobe (1981) J. Anat., 132, pp. 357-370Leav, I., Schelling, K.H., Adams, J.Y., Merck, F.B., Alroy, J., Role of canine basal cells in postnatal prostatic development, induction of hyperplasia, and sex-hormone-stimulated growth, and ductal origin of carcinoma (2001) Prostate, 48, pp. 210-224Lee, D.K., Chang, C., Expression and degradation of androgen receptor: mechanism and clinical implication (2003) J. Clin. Endocrinol. Metab., 88 (9), pp. 4043-4054Marker, P.C., Donjacour, A.A., Dahiya, R., Cunha, G.R., Hormonal, cellular and molecular control of prostatic development (2003) Dev. Biol., 253, pp. 165-174Mirosevich, J., Bentel, J.M., Zeps, N., Redmound, S.L., D'antuono, M.F., Dawkins, H.J.S., Androgen receptor expression of proliferating basal and luminal cells in adult murine ventral prostate (1999) J. Endocrinol., 162, pp. 341-350Miyata, K., Yabushita, S., Sano, M., Miyashita, K., Okuno, Y., Matsuo, M., Effects of perinatal exposure to flutamide on sex hormonal responsiveness in F1 male rats (2003) J. Toxicol. Sci., 28 (3), pp. 149-163Nemeth, J.A., Lee, C., Prostatic ductal system in rats: regional variation in stromal organization (1996) Prostate, 28, pp. 124-128O'neill, A.J., Boran, S.A., O'keane, C., Coffey, R.N.T., Hegarty, N.J., Hegarty, P., Gaffney, E.F., Watson, R.W.G., Caspase 3 expression in benign prostatic hyperplasia and prostate carcinoma (2001) Prostate, 47, pp. 183-188Paris, F., Weinbauer, G.F., Blüm, V., Nieschlag, E., The effect of androgen and antiandrogens on the immunohistochemical localization of the androgen receptor in accessory reproductive organs of male rats (1994) J. Steroid Biochem. Mol. Biol., 48, pp. 129-137Peehl, D.M., Cellular biology of prostatic growth factors (1996) Prostate, 6 (SUPPL), pp. 74-78Powell, S.M., Brooke, G.N., Whitaker, H.C., Reebye, V., Gamble, S.C., Chotai, D., Dart, D.A., Bevan, C.L., Mechanisms of androgen receptor repression in prostate cancer (2006) Biochem. Soc. Trans., 34 (PART 6), pp. 1124-1127Prins, G.S., Birch, L., Greene, G., Androgen receptor localization in different cell type of the adult rat prostate (1991) Endocrinology, 129 (6), pp. 3187-3199Prins, G.S., Birch, L., Immunocytochemical analysis of androgen receptor along the ducts of the separate rat prostate lobes after androgen withdrawal and replacement (1993) Endocrinology, 132, pp. 169-178Prins, G.S., Cooke, P.S., Birch, L., Androgen receptor expression and 5α-reductase activity along the proximal-distal axis of the rat prostatic duct (1992) Endocrinology, 130, pp. 3066-3073Prins, G.S., Woodham, C., Autologous regulation of androgen receptor messenger ribonucleic acid in the separate lobes of the rat prostate gland (1995) Biol. Reprod., 53, pp. 609-619Raghow, S., Kuliyev, E., Steakley, M., Greenberg, N., Steiner, M.S., Efficacious chemoprevention of primary prostate cancer by flutamide in an autochthonous transgenic model (2000) Cancer Res., 60, pp. 4093-4097Rochel, S.S., Bruni-Cardoso, A., Taboga, S.R., Vilamaior, P.S., Góes, R.M., Lobe identity in the Mongolian gerbil prostatic complex: a new rodent model for prostate study (2007) Anat. Rec. (Hoboken), 290 (10), pp. 1233-1247Ruijter, E., Van De Kaa, C., Miller, G., Ruiter, D., Debruyne, F., Schalken, J., Molecular genetics and epidemiology of prostate carcinoma (1999) Endocrinol. Rev., 20 (1), pp. 22-45Sanchez, D., Rosell, D., Honorato, B., Lopez, J., Arocena, J., Sanz, G., Androgen receptor mutations are associated with Gleason score in localized prostate cancer (2006) BJU Int., 98 (6), pp. 1320-1325Santos, F.C.A., Carvalho, H.F., Góes, R.M., Taboga, S.R., Ultrastructural characterization of secretory cell in the gerbil female prostate (2003) Tissue Cell, 34, pp. 273-282Shulz, W.A., Burchard, T.M., Cronauer, M.V., Molecular biology of prostate cancer (2003) Mol. Hum. Reprod., 9 (8), pp. 437-448Signoretti, S., Waltrengny, D., Dilks, J., Isaac, B., Lin, D., Garraway, L., Yang, A., Loda, M., P63 is a prostate basal cell marker and is required for prostate development (2000) Am. J. Pathol., 157 (6), pp. 1769-1775Singh, P., Uzgare, A., Litvinov, I., Denmeade, S.R., Isaacs, J.T., Combinatorial androgen receptor targeted therapy for prostate cancer (2006) Endocr. Relat. Cancer, 13, pp. 653-666Singh, S.M., Gauthier, S., Labrie, F., Androgen receptor antagonists (antiandrogens): structure-activity relationships (2000) Curr. Med. Chem., 7, pp. 211-247Steers, W.D., 5alpha-reductase activity in the prostate (2001) Urology, 58 (6 SUPPL. 1), pp. 17-24Sugimura, Y., Cunha, G.R., Donjacour, A.A., Morphogenesis of ductal networks in the mouse prostate (1986) Biol. Reprod., 34, pp. 961-971Sweat, S.D., Pacelli, A., Bergstralh, E.J., Slezak, J.M., Bostwick, D., Androgen receptor expression in prostatic intraepithelial neoplasia and cancer (1999) J. Urol., 161, pp. 1229-1232Takeda, H., Chodak, G., Mutchnik, S., Nakamoto, T., Chang, C., Immunohistochemical localization of androgen receptors with mono and polyclonal antibodies to androgen receptor (1990) J. Endrocrinol., 126, pp. 17-25Taplin, M.E., Ho, S.M., The endocrinology of prostate cancer (2001) J. Clin. Endocr. Metab., 86 (8), pp. 3467-3477Thomson, A.A., Foster, B.A., Cunha, G.R., Analyses of growth factor and receptor mRNA levels during development of the rat seminal vesicle and prostate (1997) Development, 124, pp. 2431-2439Tilley, W.D., Horsfall, D.J., Mcgee, M.A., Henderson, D.W., Marshall, V.R., Distribution of oestrogen and androgen receptors between the stroma and epithelium of the guinea-pig prostate (1985) J. Steroid Biochem., 22 (6), pp. 713-719Titus, M.A., Schell, M.J., Lih, F.B., Tomer, K.B., Mohler, J.L., Testosterone and dihydrotestosterone tissue levels in recurrent prostate cancer (2005) Clin. Cancer Res., 11 (13), pp. 4653-4657Van Leenders, G.J.L.H., Schalken, J.A., Epithelial cell differentiation in the human prostate epithelium: implications for the pathogenesis and therapy of prostate cancer (2003) Crit. Rev. Oncol./Hematol., 46 (1), pp. 3-10Vilamaior, P.S.L., Felisbino, S.R., Taboga, S.R., Carvalho, H.F., Collagen fiber reorganization in the rat ventral prostate following androgen deprivation: an possible role for the smooth muscle cells (2000) Prostate, 45, pp. 253-258Vilamaior, P.S.L., Taboga, S.R., Carvalho, H.F., Postnatal growth of the ventral prostate in Wistar rats: a stereological and morphometrical study (2006) Anat. Rec., 288, pp. 885-892Weibel, E.R., Principles and methods for the morphometric study of the lung and other organs (1963) Lab. Invest., 12, pp. 131-155Wernet, N., Gerdes, J., Loy, V., Seitz, G., Scherr, O., Dhom, G., Investigations of the estrogen (ER-ICA-test) and progesterone receptor in the prostate and prostatic carcinoma on immunohistochemical basis (1988) Virchows Arch. Pathol., 412, pp. 387-391Wilson, R.P., Bruchovsky, N., Shnitka, T.K., Rennie, P.S., Comeau, T.L., Stromal 5α-reductase activity is elevated in benign prostatic hyperplasia (1980) Acta Endocrinol., 94, pp. 284-288Zhao, G.Q., Holterhus, P.M., Dammshauser, I., Hoffbauer, G., Aümuller, G., Estrogen-induced morphological and immunohistochemical changes in stroma and epithelium of rat ventral prostate (1994) Prostate, 21, pp. 183-19

    Androgen Therapy Reverses Injuries Caused By Ethanol Consumption In The Prostate: Testosterone As A Possible Target To Ethanol-related Disorders

    No full text
    Aims: Chronic ethanol consumption leads to reproductive damages, since it can act directly in the tissues or indirectly, causing a hormonal imbalance. Prostate is a hormone-dependent gland and, consequently, susceptible to ethanol. The potential of testosterone therapy in the ethanol-related disorders was investigated in the prostate microenvironment. Main methods: UChB rats aged 90 dayswere divided into 2 experimental groups (n = 20): C: drinking water only and EtOH: drinking 10% (v/v) ethanol at > 2 g/kg body weight / day + water. At 150 days old, 10 rats from each group received subcutaneous injections of testosterone cypionate (5 mg/kg body weight) diluted in corn oil every other day for 4 weeks, constituting T and EtOH+T, while the remaining animals received corn oil as vehicle. Animals were euthanized at 180 days old, by decapitation. Blood was collected to obtain hormone concentrations and ventral prostate was dissected and processed for light microscope and molecular analyses. Key findings: Ventral prostateweight, plasma testosterone and DHT and intraprostatic testosterone concentrations were increased after testosterone treatment. Plasma estradiol level was reduced in the EtOH+T. Inflammatory foci, metaplasia and epithelial atrophy were constantly found in the prostate of EtOH and were not observed after hormonal therapy. No differences were found in the expression of AR, ERβ and DACH-1. Additionally, testosterone treatment down-regulated ERα and increased the e-cadherin and α-actinin immunoreactivities. Significance: Testosterone was able to reverse damages caused by ethanol consumption in the prostate microenvironment and becomes a possible target to be investigated to ethanol-related disorders.1202230Adams, J.Y., Leav, I., Lau, K.M., Ho, S.M., Pflueger, S.M., Expression of estrogen receptor beta in the fetal, neonatal, and prepubertal human prostate (2002) Prostate, 52, pp. 69-81Bosland, M.C., Sex steroids and prostate carcinogenesis: Integrated, multifactorial working hypothesis (2006) Ann. N. Y. Acad. Sci., 1089, pp. 168-176Campana, A.O., Burini, R.C., Outa, A.Y., Camargo, J.L., Experimental protein deficiency in adult rats (1975) Rev. Bras. Pesqui. Med. Biol., 8, pp. 221-226Cândido, E.M., Carvalho, C.A.F., Martinez, F.E., Cagnon, V.H.A., Experimental alcoholism and pathogenesis of prostatic diseases in UChB rats (2007) Cell Biol. Int., 31, pp. 459-472Carreau, S., Bourguiba, S., Lambard, S., Galeraud-Denis, I., Genissel, C., Levallet, J., Reproductive system: Aromatase and estrogens (2003) Mol. Cell. Endocrinol., 193, pp. 137-143Cordeiro, R.S., Scarano, W.R., Campos, S.G., Santos, F.C., Vilamaior, P.S., Góes, R.M., Taboga, S.R., Androgen receptor in the Mongolian gerbil ventral prostate: Evaluation during different phases of postnatal development and following and androgen blockage (2008) Micron, 39, pp. 1312-1324Debes, J.D., Tindall, D.J., The role of androgens and the androgen receptor in prostate cancer (2002) Cancer Lett., 187, pp. 1-7Ellem, S.J., Risbrindger, G.P., The dual, opposing roles of estrogen in the prostate (2009) Ann. N. Y. Acad. Sci., 1155, pp. 174-186Fávaro, W.J., Cagnon, V.H.A., Immunolocalization of androgen and oestrogen receptors in the ventral lobe of rat (Rattus norvegicus) prostate after long-term treatment with ethanol and nicotine (2008) Int. J. Androl., 31, pp. 609-618García-Cao, I., Duran, A., Collado, M., Carrascosa, M.J., Martín-Caballero, J., Flores, J.M., Tumor suppression activity of the proapoptotic regulator PAR4 (2005) EMBO Rep., 6, pp. 577-583Gomes, I.C., Cagnon, V.H.A., De Luca, I.M., Stereology and ultrastructure of the seminal vesicle of C57/BL/6J mice following chronic alcohol ingestion (2002) Tissue Cell, 34, pp. 177-186Gonc¸avez, B.F., Campos, S.G.P., Zanetoni, C., Scarano, W.R., Faleiros, L.R., Jr., Amorim, R.L., Góes, R.M., Taboga, S.R., A new proposed rodent model of chemically induced prostate carcinogenesis: Distinct time-course prostate cancer progression in the dorsolateral and ventral lobes (2013) Prostate, 73, pp. 1202-1213Griffts, K., Morton, M.S., Nicholson, R.I., Androgens, androgens receptors, antiandrogens and the treatment of prostate cancer (1997) Eur. Urol., 32, pp. 24-40Guess, H.A., Benign prostatic hyperplasia and prostate cancer (2001) Epidemiol. Rev., 23, pp. 152-158Gurumurthy, S., Rangnekar, V.M., Par-4 inducible apoptosis in prostate cancer cells (2004) J. Cell. Biochem., 91, pp. 504-512Harkonen, P.L., Makela, S.I., Role of estrogens in development of prostate cancer (2004) J. Steroid Biochem. Mol. Biol., 92, pp. 297-305Kawashima, H., Nakatani, T., Involvement of estrogen receptors in prostatic diseases (2012) Int. J. Urol., 19, pp. 512-522Landau, D., Tsakok, T., Aylwin, S., Hughes, S., Should testosterone replacement be offered to hypogonadal men treated previously for prostatic carcinoma? (2012) Clin. Endocrinol., 76, pp. 179-181Lewis, M.I., Horvitz, G.D., Clemmons, D.R., Fournier, M., Role of IGF-I and IGF-binding proteins within diaphragm muscle in modulating the effects of nandrolone (2002) Am. J. Physiol. Endocrinol. Metab., 282, pp. 483-490Li, T.K., Lumeng, L., McBride, W.J., Murphy, J.M., Rodent lines selected for factors affecting alcohol solution (1987) Alcohol Alcohol., 1, pp. 91-96Lieber, C.S., Alcohol and the liver: 1984 update (1984) Hepatology, 4, pp. 1243-1260Lin, E.Y., Pollard, J.W., Role of infiltrated leucocytes in tumour growth and spread (2004) Br. J. Cancer, 90, pp. 2053-2058Mardones, J., Segovia-Riquelme, N., Thirty two years of selection of rats by ethanol preference: UChA and UChB strains (1983) Neurobehav. Toxicol. Teratol., 5, pp. 171-178Morani, A., Warner, M., Gustafsson, J.A., Biological functions and clinical implications of oestrogen receptors alfa and beta in epithelial tissues (2008) J. Intern. Med., 264, pp. 128-142Muthusamy, S., Andersson, S., Kim, H.J., Butler, R., Waage, L., Bergerheim, U., Estrogen receptor beta and 17beta-hydroxysteroid dehydrogenase type 6, a growth regulatory pathway that is lost in prostate cancer (2011) Proc. Natl. Acad. Sci. U. S. A., 108, pp. 20090-20094National Research of Council, (1995) Nutrient Requirements of Laboratory Animals, 4th Ed., , National Academy Press, Washington, DCRittmaster, R.S., 5α-Reductase inhibitors in benign prostatic hyperplasia and prostate cancer risk reduction (2008) Best Pract. Res. Clin. Endocrinol. Metab., 22, pp. 389-402Rochel-Maia, S.S., Santos, F.C.A., Alonso-Magdalena, P., Góes, R.M., Vilamaior, P.S.L., Warner, M., Estrogen receptors alpha and beta in male and female gerbil prostates (2013) Biol. Reprod., 88, pp. 1-7Ronis, M.J.J., Wands, J.R., Badger, T.M., De La Monte, S.M., Lang, C.H., Calissendorff, J., Alcohol-induced disruption of endocrine signaling (2007) Alcohol. Clin. Exp. Res., 31, pp. 1269-1285Roy-Burman, P., Wu, H., Powell, W.C., Hagenkord, J., Cohen, M.B., Genetically defined mouse models that mimic natural aspects of human prostate cancer development (2004) Endocr. Relat. Cancer, 11, pp. 225-254Salomen, I., Huhtaniemi, I., Effects of chronic ethanol diet on pituitary-testicular function of the rat (1990) Biol. Reprod., 42, pp. 55-62Sáttolo, S., Carvalho, C.A.F., Cagnon, V.H.A., Influence of hormonal replacement on the ventral lobe of the prostate of rats (Rattus novergicus albinus) submitted to chronic ethanol treatment (2004) Tissue Cell, 36, pp. 417-430Saxena, S., Meehan, D., Coney, P., Wimalasena, J., Ethanol has direct inhibitory effects on steroidogenesis in human granulosa cells: Specific inhibition of LH action (1990) Alcohol. Clin. Exp. Res., 14, pp. 522-527Scarano, W.R., Vilamaior, P.S.L., Taboga, S.R., Tissue evidence of the testosterone role on the abnormal growth and aging effects reversion in the gerbil (Meriones unguilatus) prostate (2006) Anat. Rec. A Discov. Mol. Cell Biol., 11, pp. 1190-1200Sugiyama, N., Barros, R.P., Warner, M., Gustafsson, J.A., ERβ: Recent understanding of estrogen signaling (2010) Trends Endocrinol. Metab., 21, pp. 545-552Tadic, S.D., Elm, M.S., Subbotin, V.M., Eaggon, P.K., Hypogonadism precedes liver feminization in chronic alcohol-fed male rats (2000) Hepatology, 31, pp. 1135-1140Thomas, C., Gustafsson, J.A., The different roles of ER subtypes in cancer biology and therapy (2011) Nat. Rev., 11, pp. 597-608Weihua, Z., Warner, M., Gustafsson, J.A., Estrogen receptor beta in the prostate (2002) Mol. Cell. Endocrinol., 193, pp. 1-5Weibel, E.R., Principles and methods for the morphometric study of the lung and other organs (1963) Lab. Investig., 12, pp. 131-155Wilson, J., Balkwill, F., The role of cytokines in the epithelial cancer microenvironment (2002) Semin. Cancer Biol., 12, pp. 113-120Wu, K., Katiyar, S., Witkiewicz, A., Li, A., McCue, P., Song, L.N., The cell fate determination factor dachshund inhibits androgen receptor signaling and prostate cancer cellular growth (2009) Cancer Res., pp. 593347-593355Yatkin, E., Bernoulli, J., Talvitie, E., Santti, R., Inflammation and epithelial alterations in the rat prostate: Impact of the androgen to oestrogen ratio (2009) Int. J. Androl., 32, pp. 399-41

    Gestational Protein Restriction Delays Prostate Morphogenesis In Male Rats

    No full text
    Maternal malnutrition due to a low-protein diet is associated with functional disorders in adulthood, which may be related to embryonic development failures. The effects of gestational protein restriction on prostate morphogenesis in male offspring were investigated. Pregnant rat dams were divided into normoprotein (NP; fed a normal diet containing 17% protein) and hypoprotein (LP; fed a diet containing 6% protein) groups. On the day of birth (PND1), anogenital distance and bodyweight were measured in male pups. Seven males per experimental group (one male per litter) were killed, and the pelvic urethra was evaluated. LP offspring showed a significant reduction in bodyweight and anogenital distance on PND1. On three-dimensional reconstruction of the prostate, the number of prostatic buds was lower in LP than in NP males. Mesenchymal cells surrounding the buds were androgen-receptor positive, and the quantity and intensity of nucleus immunoreactivity was decreased in LP. The proliferation index was lower in LP than in NP prostatic buds. Immunoreactivity for α-actin in mesenchymal cells and that for epidermal growth factor receptor in epithelial cells was higher in NP than in LP. Our findings demonstrate that maternal protein restriction delays prostatic morphogenesis, probably because of considerable disruption in the epithelium-mesenchyme interaction. © 2014 CSIRO.267967973Barker, D.J., In utero programming of cardiovascular disease (2000) Theriogenology, 53, pp. 555-574. , doi:10.1016/S0093-691X(99)00258-7Cónsole, G.M., Jurado, S.B., Oyhenart, E., Ferese, C., Pucciarelli, H., Gómez Dumm, C.L., Morphometric and ultrastructural analysis of different pituitary cell populations in undernourished monkeys (2001) Braz. J. Med. Biol. Res., 34, pp. 65-74. , doi:10.1590/S0100- 879X2001000100008Cooke, P.S., Young, P., Cunha, G.R., Androgen receptor expression in developing male reproductive organs (1991) Endocrinology, 128, pp. 2867-2873. , doi:10.1210/ENDO-128-6-2867Cunha, G.R., Alarid, E.T., Turner, T., Donjacour, A.A., Boutin, E.L., Foster, B.A., Normal and abnormal development of the male urogenital tract: Role of androgens, mesenchymal-epithelial interactions and growth factors (1992) J. Androl., 13, pp. 465-475Cunha, G.R., Foster, B., Thomson, A., Sugimura, Y., Tanji, N., Tsuji, M., Terada, N., Donjacour, A.A., Growth factors as mediators of androgen action during the development of the male urogenital tract (1995) World J. Urol., 13, pp. 264-276. , doi:10.1007/ BF00185969Da Silva Faria, T., Da Fonte Ramos, C., Sampaio, F.J., Puberty onset in the female offspring of rats submitted to protein or energy restricted diet during lactation (2004) J. Nutr. Biochem., 15, pp. 123-127. , doi:10.1016/J.JNUTBIO.2003.08.011Desai, M., Crowther, N.J., Lucas, A., Hales, C.N., Organselective growth in the offspring of protein-restricted mothers (1996) Br. J. Nutr., 76, pp. 591-603. , doi:10.1079/BJN19960065Fagundes, A.T., Moura, E.G., Passos, M.C.F., Oliveira, E., Toste, F.P., Bonomo, I.T., Trevenzoli, I.H., Lisboa, P.C., Maternal low-protein diet during lactation programmes body composition and glucose homeostasis in the adult rat offspring (2007) Br. J. Nutr., 98, pp. 922-928. , doi:10.1017/S0007114507750924Gardner, D.S., Ozanne, S.E., Sinclair, K.D., Effect of the earlylife nutritional environment on fecundity and fertility of mammals (2009) Philos. Trans. R. Soc. Lond. B Biol. Sci., 364, pp. 3419-3427. , doi:10.1098/ RSTB.2009.0121Gosby, A.K., Stanton, L.M.L., Maloney, C.A., Thompson, M., Briody, J., Baxter, R.C., Bryson, J.M., Caterson, I.D., Postnatal nutrition alters body composition in adult offspring exposed to maternal protein restriction (2009) Br. J. Nutr., 101 (12), pp. 1878-1884. , doi:10.1017/S0007114508135851Graham, S., Gandelman, R., The expression of anogenital distance data in the mouse (1986) Physiol. Behav., 36, pp. 103-104. , doi:10.1016/ 0031-9384(86)90081-8Hayashi, N., Sugimura, Y., Kawamura, J., Donjacour, A.A., Cunha, G.R., Morphological and functional heterogeneity in the rat prostatic gland (1991) Biol. Reprod., 45, pp. 308-321. , doi:10.1095/BIOLREPROD45.2.308Lucas, A., Programming by early nutrition: An experimental approach (1998) J. Nutr., 128, pp. 401S-406SMarker, P.C., Donjacour, A.A., Dahiya, R., Cunha, G.R., Hormonal, cellular and molecular control of prostatic development (2003) Dev. Biol., 253, pp. 165-174. , doi:10.1016/S0012-1606(02)00031-3McMillen, I.C., Maclaughlin, S.M., Muhlhausler, B.S., Gentili, S., Duffield, J.L., Morrison, J.L., Developmental origins of adult health and disease: The role of periconceptional and foetal nutrition (2008) Basic Clin. Pharmacol. Toxicol., 102, pp. 82-89. , doi:10.1111/J.1742-7843. 2007.00188.XPage, K.C., Sottas, C.M., Hardy, M.P., Prenatal exposure to dexamethasone alters Leydig cell steroidogenic capacity in immature and adult rats (2001) J. Androl., 22, pp. 973-980Prins, G.S., Birch, L., The developmental pattern of androgen receptor expression in rat prostate lobes is altered after neonatal exposure to oestrogen (1995) Endocrinology, 136, pp. 1303-1314. , doi:10.1210/ EN.136.3.1303Prins, G.S., Cooke, P.S., Birch, L., Donjacour, A.A., Yalcinkaya, T.M., Siiteri, P.K., Cunha, G.R., Androgen receptor expression and 5a-reductase activity along the proximal-distal axis of the rat prostatic duct (1992) Endocrinology, 130, pp. 3066-3073. , doi:10.1210/EN.130.5.3066Prins, G.S., Birch, L., Habermann, H., Chang, W.Y., Tebeau, C., Putz, O., Bieberich, C., Influence of neonatal oestrogens on rat prostate development (2001) Reprod. Fertil. Dev., 13, pp. 241-252. , doi:10.1071/RD00107Puchtler, H., Waldrop, F.S., Meloan, S.N., Terry, M.S., Conner, H.M., Methacarn (methanol-carnoy) fixation: Practical and theoretical considerations (1970) Histochemie, 21, pp. 97-116. , doi:10.1007/BF00306176Reeves, P.G., Nielsen, F.H., Fahey, G.C., AIN-93 purified diets for laboratory rodents: Final report of the American Institute of Nutrition Ad Hoc Writing Committee on the reformulation of the AIN-76 rodent diet (1993) J. Nutr., 123, pp. 1939-1951Rinaldi, J.C., Justulin, L.A., Lacorte, L.M., Sarobo, C., Boer, P.A., Scarano, W.R., Felisbino, S.L., Implications of intrauterine protein malnutrition on prostate growth, maturation and aging (2013) Life Sci., 92, pp. 763-774. , doi:10.1016/J.LFS.2013.02.007Santos, F.C.A., Leite, R.P., Custódio, A.M.G., Carvalho, K.P., Monteiro- Leal, L.H., Santos, A.B., Goes, R.M., Taboga, S.R., Testosterone stimulates growth and secretory activity of the female prostate in the adult gerbil (Meriones unguiculatus) (2006) Biol. Reprod., 75, pp. 370-379. , doi:10.1095/BIOLREPROD.106.051789Scarano, W.R., Toledo, F.C., Guerra, M.T., Campos, S.G.P., Justulin Jr., L.A., Felisbino, S.L., Anselmo-Franci, J.A., Kempinas, W.G., Long-term effects of developmental exposure to di-N-butyl-phthalate (DBP) on rat prostate: Proliferative and inflammatory disorders and a possible role of androgens (2009) Toxicology, 262 (3), pp. 215-223. , doi:10.1016/J.TOX.2009.06.011Sugimura, Y., Cunha, G.R., Donjacour, A.A., Morphogenesis of ductal networks in the mouse prostate (1986) Biol. Reprod., 34, pp. 961-971. , doi:10.1095/BIOLREPROD34.5.961Swan, S.H., Main, K.M., Liu, F., Stewart, S.L., Kruse, R.L., Calafat, A.M., Mao, C.S., Teague, J.L., Decrease in ano-genital distance among male infants with prenatal phthalate exposure (2005) Environ. Health Perspect., 113, pp. 1056-1061. , doi:10.1289/EHP.8100Thomson, A.A., Role of androgens and fibroblast growth factors in prostatic development (2001) Reproduction, 121, pp. 187-195. , doi:10.1530/REP. 0.1210187Thomson, A.A., Cunha, G.R., Prostatic growth and development are regulated by FGF10 (1999) Development, 126, pp. 3693-3701Timms, B.G., Prostate development: A historical perspective (2008) Differentiation, 76, pp. 565-577. , doi:10.1111/J.1432-0436.2008.00278.XTimms, B.G., Mohs, T.J., Didio, L.J., Ductal budding and branching patterns in the developing prostate (1994) J. Urol., 151, pp. 1427-1432Vilamaior, P.S.L., Taboga, S.R., Carvalho, H.F., Postnatal growth of the ventral prostate in Wistar rats: A stereological and morphometrical study (2006) Anat. Rec. A Discov. Mol. Cell. Evol. Biol., 288 A, pp. 885-892. , doi:10.1002/AR.A.20363Wang, Y., Hayward, S., Cao, M., Thayer, K., Cunha, G.R., Cell differentiation lineage in the prostate (2001) Differentiation, 68, pp. 270-279. , doi:10.1046/J.1432-0436.2001.680414.XWoodham, C., Birch, L., Prins, G.S., Neonatal oestrogen downregulates prostatic androgen receptor through a proteosome-mediated protein degradation pathway (2003) Endocrinology, 144, pp. 4841-4850. , doi:10.1210/EN.2003-0035Zambrano, E., Rodrigez-González, G.L., Guzmán, C., García-Becerra, R., Boeck, L., Díaz, L., Menjivar, M., Nathanielsz, P.W., A maternal low-protein diet during pregnancy and lactation in the rat impairs male reproductive development (2005) J. Physiol., 563, pp. 275-284. , doi:10.1113/JPHYSIOL.2004.07854

    Briófitas do Parque Nacional da Restinga de Jurubatiba, RJ, Brasil Bryophytes of Restinga de Jurubatiba National Park, Rio de Janeiro State, Brazil

    No full text
    Este trabalho representa uma síntese do conhecimento florístico de briófitas do Parque Nacional da Restinga de Jurubatiba, com resultados de campo, material de herbário e publicações. Foram registrados 61 táxons de briófitas, pertencentes a 15 famílias e 33 gêneros. Para cada táxon foram analisados a distribuição geográfica, o tipo de substrato e a forma de vida. A família predominante foi Lejeuneaceae (25 espécies), seguida de Jubulaceae (sete), Calymperaceae (quatro) e Sphagnaceae (quatro). Entre as espécies, predominaram as de distribuição Neotropical (35%). Com relação ao substrato preferencial, a maior parte dos táxons é corticícola. A forma de vida predominante foi o tapete, seguido pelo tufo e a trama. O Parque possui brioflora bastante representativa no que diz respeito à vegetação de restinga, com 45% dos táxons de briófitas conhecidas para as restingas no Brasil e 82% das conhecidas para as restingas do estado do Rio de Janeiro. É apresentada uma chave para identificação das espécies.<br>We present a synthesis of the bryophyte flora in Restinga de Jurubatiba National Park, based on field work, herbaria material and bibliography. Sixty one taxa were recorded, in 15 families and 33 genera. For each taxa, we analyze geographic distribution, substrata and life form. The most species-rich family was Lejeuneaceae (25 species), followed by Jubulaceae (seven), Calymperaceae (four) and Sphagnaceae (four). Most taxa have a Neotropical (35%) distribution, and most taxa prefer the corticicolous substrate. Mat was the most common life form. The bryoflora of this National Park is an important reservoir of bryophyte diversity in the restingas, with 45% of the taxa known from the sandy coastal plains of Brazil and 82% known from the this habitat in Rio de Janeiro state. A key to identification of the species is given
    corecore