Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell Death Dis ; 7: e2294, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27415424

RESUMEN

GPRC5A is an orphan G-protein coupled receptor with an intriguing dual behavior, acting as an oncogene in some cancers and as a tumor suppressor in other cancers. In the pancreatic cancer context, very little is known about GPRC5A. By analyzing messenger RNA (mRNA) expression data from 675 human cancer cell lines and 10 609 samples from The Cancer Genome Atlas (TCGA) we found that GPRC5A's abundance in pancreatic cancer is highest (cell lines) or second highest (TCGA) among all tissues and cancer types. Further analyses of an independent set of 252 pancreatic normal and cancer samples showed GPRC5A mRNA to be more than twofold upregulated in primary tumor samples compared with normal pancreas (P-value<10(-5)), and even further upregulated in pancreatic cancer metastases to various organs (P-value=0.0021). Immunostaining of 208 cores (103 samples) of a tissue microarray showed generally low expression of GPRC5A protein in normal pancreatic ductal cells; on the other hand, in primary and metastatic samples, GPRC5A protein levels were dramatically increased in pancreatic ductal cells. In vitro studies of multiple pancreatic cancer cell lines showed that an increase in GPRC5A protein levels promoted pancreatic cancer cell growth and migration. Unexpectedly, when we treated pancreatic cancer cell lines with gemcitabine (2',2'-difluorodeoxycytidine), we observed an increase in GPRC5A protein abundance. On the other hand, when we knocked down GPRC5A we sensitized pancreatic cancer cells to gemcitabine. Through further experimentation we showed that the monotonic increase in GPRC5A protein levels that we observe for the first 18 h following gemcitabine treatment results from interactions between GPRC5A's mRNA and the RNA-binding protein HuR, which is an established key mediator of gemcitabine's efficacy in cancer cells. As we discovered, the interaction between GPRC5A and HuR is mediated by at least one HuR-binding site in GPRC5A's mRNA. Our findings indicate that GPRC5A is part of a complex molecular axis that involves gemcitabine and HuR, and, possibly, other genes. Further work is warranted before it can be established unequivocally that GPRC5A is an oncogene in the pancreatic cancer context.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/genética , Desoxicitidina/análogos & derivados , Proteína 1 Similar a ELAV/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/genética , Receptores Acoplados a Proteínas G/genética , Sitios de Unión , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/farmacología , Proteína 1 Similar a ELAV/metabolismo , Humanos , Metástasis de la Neoplasia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Unión Proteica , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Análisis de Matrices Tisulares , Gemcitabina
2.
Oncogene ; 35(19): 2529-41, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26387536

RESUMEN

Previously, it has been shown that pancreatic ductal adenocarcinoma (PDA) tumors exhibit high levels of hypoxia, characterized by low oxygen pressure (pO2) and decreased O2 intracellular perfusion. Chronic hypoxia is strongly associated with resistance to cytotoxic chemotherapy and chemoradiation in an understudied phenomenon known as hypoxia-induced chemoresistance. The hypoxia-inducible, pro-oncogenic, serine-threonine kinase PIM1 (Proviral Integration site for Moloney murine leukemia virus 1) has emerged as a key regulator of hypoxia-induced chemoresistance in PDA and other cancers. Although its role in therapeutic resistance has been described previously, the molecular mechanism behind PIM1 overexpression in PDA is unknown. Here, we demonstrate that cis-acting AU-rich elements (ARE) present within a 38-base pair region of the PIM1 mRNA 3'-untranslated region mediate a regulatory interaction with the mRNA stability factor HuR (Hu antigen R) in the context of tumor hypoxia. Predominantly expressed in the nucleus in PDA cells, HuR translocates to the cytoplasm in response to hypoxic stress and stabilizes the PIM1 mRNA transcript, resulting in PIM1 protein overexpression. A reverse-phase protein array revealed that HuR-mediated regulation of PIM1 protects cells from hypoxic stress through phosphorylation and inactivation of the apoptotic effector BAD and activation of MEK1/2. Importantly, pharmacological inhibition of HuR by MS-444 inhibits HuR homodimerization and its cytoplasmic translocation, abrogates hypoxia-induced PIM1 overexpression and markedly enhances PDA cell sensitivity to oxaliplatin and 5-fluorouracil under physiologic low oxygen conditions. Taken together, these results support the notion that HuR has prosurvival properties in PDA cells by enabling them with growth advantages in stressful tumor microenvironment niches. Accordingly, these studies provide evidence that therapeutic disruption of HuR's regulation of PIM1 may be a key strategy in breaking an elusive chemotherapeutic resistance mechanism acquired by PDA cells that reside in hypoxic PDA microenvironments.


Asunto(s)
Resistencia a Antineoplásicos , Proteína 1 Similar a ELAV/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas c-pim-1/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular , Fluorouracilo/farmacología , Humanos , Compuestos Organoplatinos/farmacología , Oxaliplatino , Oxígeno/metabolismo , Proto-Oncogenes Mas , ARN Mensajero/metabolismo
3.
Oncogene ; 32(48): 5481-91, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23708653

RESUMEN

The cyclin/cyclin-dependent kinase (CDK)/retinoblastoma (RB)-axis is a critical modulator of cell cycle entry and is aberrant in many human cancers. New nodes of therapeutic intervention are needed that can delay or combat the onset of malignancies. The antitumor properties and mechanistic functions of PD-0332991 (PD; a potent and selective CDK4/6 inhibitor) were investigated using human prostate cancer (PCa) models and primary tumors. PD significantly impaired the capacity of PCa cells to proliferate by promoting a robust G1-arrest. Accordingly, key regulators of the G1-S cell cycle transition were modulated including G1 cyclins D, E and A. Subsequent investigation demonstrated the ability of PD to function in the presence of existing hormone-based regimens and to cooperate with ionizing radiation to further suppress cellular growth. Importantly, it was determined that PD is a critical mediator of PD action. The anti-proliferative impact of CDK4/6 inhibition was revealed through reduced proliferation and delayed growth using PCa cell xenografts. Finally, first-in-field effects of PD on proliferation were observed in primary human prostatectomy tumor tissue explants. This study shows that selective CDK4/6 inhibition, using PD either as a single-agent or in combination, hinders key proliferative pathways necessary for disease progression and that RB status is a critical prognostic determinant for therapeutic efficacy. Combined, these pre-clinical findings identify selective targeting of CDK4/6 as a bona fide therapeutic target in both early stage and advanced PCa and underscore the benefit of personalized medicine to enhance treatment response.


Asunto(s)
Antineoplásicos/farmacología , Terapia Molecular Dirigida , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Inhibidores de Proteínas Quinasas/farmacología , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Estudios de Factibilidad , Humanos , Masculino , Ratones , Piperazinas/farmacología , Piperazinas/uso terapéutico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Retinoblastoma/tratamiento farmacológico , Retinoblastoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Breast Cancer Res Treat ; 68(1): 65-73, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11678310

RESUMEN

The pp32 gene family consists of at least three closely related members, pp32, pp32r1 and pp32r2. In spite of a high degree of identity at the nucleotide level, pp32 functionally behaves as a tumor suppressor where as pp32r1 and pp32r2 are pro-oncogenic. The purpose of this pilot study was to determine pp32-related expression and whether alternative gene use among the pp32 family members occurred in human breast cancer. As a first step, in situ hybridization with a riboprobe capable of hybridizing with all the three members showed abundant pp32-related mRNA in benign ducts and acini and in infiltrating ductal carcinomas. A total of 100/102 cases were positive. Further, a detailed molecular analysis by RT-PCR, cloning, and sequencing was performed in five frozen infiltrating breast carcinomas and matched benign breast tissues. Oncogenic pp32r1 (5/5) and pp32r2 (3/5) expression was observed in carcinomas where as benign breast tissues expressed pp32. 4/5 carcinomas continued to express pp32 but one was devoid of pp32 expression. These results suggest that alternative expression of pp32 family members may be common in human breast cancer and the analysis of the profile of pp32-related expression might be helpful in understanding the role of these genes in breast cancer pathogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Regulación Neoplásica de la Expresión Génica , Familia de Multigenes/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Estudios de Casos y Controles , Cartilla de ADN , Femenino , Humanos , Hibridación in Situ , Proyectos Piloto , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Oncogene ; 20(17): 2153-60, 2001 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-11360199

RESUMEN

Alternative use of genes of the closely-related pp32 family is a common occurrence in human prostate cancer. pp32r1 and pp32r2, the oncogenic members of the pp32 family, are expressed in prostatic adenocarcinoma, while adjacent benign prostate continues to express pp32. This study focuses upon the role of pp32 in tumor suppression. We demonstrate that antisense inhibition of pp32 in NIH3T3 cells leads to a variety of phenotypic changes associated with transformation including reduced serum dependence and loss of contact inhibition. NIH3T3 cells with antisense-inhibited pp32 are not tumorigenic, but are markedly more susceptible to oncogenic stimuli such as ras. In contrast, constitutive expression of pp32 abolishes ras mediated transformation in vitro and tumorigenesis in vivo. These data demonstrate, from the functional aspect, that pp32 acts as a tumor suppressor. Furthermore, inactivation of pp32 function through alternative gene use may be a critical event in tumor evolution and progression.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Genes Supresores de Tumor , Proteínas Nucleares/biosíntesis , Fosfoproteínas/biosíntesis , Células 3T3/citología , Células 3T3/metabolismo , Animales , División Celular/fisiología , Transformación Celular Neoplásica/genética , ADN sin Sentido/genética , ADN Complementario/genética , Regulación de la Expresión Génica , Genes ras/fisiología , Humanos , Ratones , Proteínas Nucleares/genética , Fosfoproteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transfección
6.
Am J Pathol ; 155(4): 1293-302, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10514411

RESUMEN

The terminal components of complement C5b-C9 can cause significant injury to cardiac allografts. Using C6-deficient rats, we have found that the rejection of major histocompatibility (MHC) class I-incompatible PVG.R8 (RT1.A(a)B(u)) cardiac allografts by PVG.1U (RT1.A(u)B(u)) recipients is particularly dependent on C6. This model was selected to determine whether tissue injury results from C6 produced by macrophages, which are a conspicuous component of infiltrates in rejecting transplants. We demonstrated that high levels of C6 mRNA are expressed in isolated populations of macrophages. The relevance of macrophage-produced C6 to cardiac allograft injury was investigated by transplanting hearts from PVG. R8 (C6-) donors to PVG.1U (C6-) rats which had been reconstituted with bone marrow from PVG.1U (C6+) rats as the sole source of C6. Hearts grafted to hosts after C6 reconstitution by bone marrow transplantation underwent rejection characterized by deposition of IgG and complement on the vascular endothelium together with extensive intravascular aggregates of P-selectin-positive platelets. At the time of acute rejection, the cardiac allografts contained extensive perivascular and interstitial macrophage infiltrates. RT-PCR and in situ hybridization demonstrated high levels of C6 mRNA in the macrophage-laden transplants. C6 protein levels were also increased in the circulation during rejection. To determine the relative contribution to cardiac allograft rejection of the low levels of circulating C6 produced systemically by macrophages, C6 containing serum was passively transferred to PVG.1U (C6-) recipients of PVG.R8 (C6-) hearts. This reconstituted the C6 levels to about 3 to 6% of normal values, but failed to induce allograft rejection. In control PVG.1U (C6-) recipients that were reconstituted with bone marrow from PVG.1U (C6-) donors, C6 levels remained undetectable and PVG.R8 cardiac allografts were not rejected. These results indicate that C6 produced by macrophages can cause significant tissue damage.


Asunto(s)
Complemento C6/biosíntesis , Complemento C6/inmunología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Macrófagos/metabolismo , Animales , Animales Congénicos , Trasplante de Médula Ósea , Complemento C3/metabolismo , Complemento C3d/metabolismo , Complemento C6/genética , Ensayo de Inmunoadsorción Enzimática , Rechazo de Injerto/sangre , Prueba de Histocompatibilidad , Inmunofenotipificación , Hibridación in Situ , Isoanticuerpos/sangre , Isoanticuerpos/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Masculino , Miocardio/patología , Selectina-P/biosíntesis , ARN Mensajero/biosíntesis , Ratas , Ratas Mutantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Biol Chem ; 274(29): 20053-5, 1999 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-10400610

RESUMEN

Oncogenic potential in prostate cancer is modulated in part by alternative use of genes of the pp32 family. This family includes the tumor suppressor pp32, expressed in normal tissue, and the pro-oncogenic genes pp32r1 and pp32r2 that are found principally in neoplastic cells. At the protein level, pp32, pp32r1, and pp32r2 are approximately 90% identical, yet they subsume opposite functions. In this study, we identify the region of pp32 associated with the ability to inhibit oncogene-mediated transformation in a rat embryo fibroblast system, an in vitro correlate of tumor-suppressive activity. Deletion and truncation analysis define a region spanning pp32 amino acids 150-174 as absolutely required for inhibition of transformed foci elicited by RAS and MYC. Comparison of pp32 with the pp32r1 sequence by moving averages of sequence identity reveals divergence over this region; pp32r2 also differs in this region through truncation after pp32 amino acid 131. The deletion experiments and the experiments of nature therefore converge to demonstrate that tumor-suppressive functions of pp32 reside in amino acids 150-174. Identification of this minimal tumor-suppressive region should help elaborate the pathways and mechanisms through which pp32 family members exert their functions.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas Nucleares/fisiología , Oncogenes , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Proteínas Nucleares/genética , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Ratas , Eliminación de Secuencia
8.
Nat Med ; 5(3): 275-9, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10086381

RESUMEN

Only a small percentage of primary prostate cancers have genetic changes. In contrast, nearly 90% of clinically significant human prostate cancers seems to express high levels of the nuclear phosphoprotein pp32 by in situ hybridization. Because pp32 inhibits oncogene-mediated transformation, we investigated its paradoxical expression in cancer by comparing the sequence and function of pp32 species from paired benign prostate tissue and adjacent prostatic carcinoma from three patients. Here we demonstrate that pp32 is expressed in benign prostatic tissue, but pp32r1 and pp32r2, closely-related genes located on different chromosomes, are expressed in prostate cancer. Although pp32 is a tumor suppressor, pp32r1 and pp32r2 are tumorigenic. Alternative use of the pp32, pp32r1 and pp32r2 genes may modulate the oncogenic potential of human prostate cancer.


Asunto(s)
Empalme Alternativo , Proteínas Nucleares/genética , Hiperplasia Prostática/genética , Neoplasias de la Próstata/genética , Humanos , Masculino , Fosfoproteínas/genética , ARN Mensajero
9.
Endocrinology ; 139(11): 4708-13, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9794483

RESUMEN

The role of epithelial and stromal progesterone (P) receptors (PR) in the regulation of uterine epithelial DNA synthesis by P was investigated by analyzing the four types of tissue recombinants prepared with uterine stroma (S) and epithelium (E) from wild-type (wt) and PR knockout (PRKO) mice: wt-S + wt-E, PRKO-S + PRKO-E, wt-S + PRKO-E, and PRKO-S + wt-E. 17-Beta estradiol (E2) stimulated DNA synthesis in all four types of tissue recombinants. On the other hand, P inhibited E2-induced DNA synthesis only in tissue recombinants prepared with wild-type (PR-positive) stroma (wt-S + wt-E or wt-S + PRKO-E) but not knockout (PR-negative) stroma (PRKO-S + wt-E or PRKO-S + PRKO-E). These results clearly demonstrate that the inhibitory effect of P on uterine epithelial DNA synthesis is mediated by stromal PR. Epithelial PR is neither necessary nor sufficient for P inhibition of E2-induced epithelial DNA synthesis.


Asunto(s)
ADN/biosíntesis , Células Epiteliales/metabolismo , Estrógenos/farmacología , Progesterona/farmacología , Receptores de Progesterona/metabolismo , Células del Estroma/metabolismo , Útero/metabolismo , Animales , Autorradiografía , Células Epiteliales/efectos de los fármacos , Estradiol/biosíntesis , Estradiol/farmacología , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Recombinación Genética , Útero/efectos de los fármacos
10.
Prostate ; 34(3): 231-7, 1998 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9492852

RESUMEN

BACKGROUND: pp32 is a differentiation-regulated nuclear phosphoprotein that is highly expressed in many cancers, but is restricted to self-renewing and long-lived normal cell populations. During murine embryogenesis, pp32 is expressed in primitive cell populations, diminishing as tissues terminally differentiate. Functionally, pp32 confers resistance to programmed cell death and, paradoxically, inhibits transformation mediated in vitro by a broad range of oncogenes, suggesting that pp32 is a multifunctional molecule with potentially complex activities in cancer. METHODS: We studied pp32 expression in prostatic adenocarcinomas and benign prostatic hyperplasia by in situ hybridization. RESULTS: In benign prostatic tissues, moderate pp32 expression occurs only in the basal cells. This study found elevated pp32 expression in 98% (54/55) of prostatic adenocarcinomas of Gleason score > or = 5 (P < 0.0001). CONCLUSIONS: These results suggest that pp32 may be diagnostically useful and may contribute mechanistically to prostate tumor development. In comparison to other molecular alterations, increased pp32 expression is one of the most frequent events in primary prostate cancer.


Asunto(s)
Adenocarcinoma/metabolismo , Proteínas Nucleares/biosíntesis , Hiperplasia Prostática/metabolismo , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/patología , Humanos , Masculino , Fosfoproteínas/biosíntesis , Hiperplasia Prostática/patología , Neoplasia Intraepitelial Prostática/metabolismo , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología
11.
Mol Biol Cell ; 7(12): 2045-56, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8970164

RESUMEN

pp32 is a nuclear protein found highly expressed in normal tissues in those cells capable of self-renewal and in neoplastic cells. We report the cloning of cDNAs encoding human and murine pp32. The clones encode a 28.6-kDa protein; approximately two-thirds of the N-terminal predicts an amphipathic alpha helix containing two possible nuclear localization signals and a potential leucine zipper motif. The C-terminal third is exceptionally acidic, comprised of approximately 70% aspartic and glutamic acid residues; the predicted pI of human pp32 is 3.81. Human and murine pp32 cDNAs are 88% identical; the predicted proteins are 89% identical and 95% similar. Although the structure of pp32 is suggestive of a transcription factor, pp32 did not significantly modulate transcription of a reporter construct when fused to the Gal4 DNA-binding domain. In contrast, in cotransfection experiments, pp32 inhibited the ability of a broad assortment of oncogene pairs to transform rat embryo fibroblasts, including ras + myc, ras + jun, ras + E1a, ras + mutant p53, and E6 + E7. In related experiments, pp32 inhibited the ability of Rat 1a-myc cells to grow in soft agar, whereas it failed to affect ras-induced focus formation in NIH3T3 cells. These results suggest that pp32 may play a key role in self-renewing cell populations where it may act in the nucleus to limit their sensitivity to transformation.


Asunto(s)
Regulación de la Expresión Génica , Genes myc , Genes ras , Proteínas Nucleares/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , ADN Complementario , Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/química , Oncogenes , Fosfoproteínas/química , Fosfoproteínas/genética , ARN Mensajero , Ratas , Transcripción Genética , Transformación Genética , Células Tumorales Cultivadas
12.
Differentiation ; 60(4): 219-27, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8765052

RESUMEN

Wholemount immunocytochemical staining was used to visualize basal and luminal epithelial-cell-specific cytokeratin and smooth muscle alpha-actin expression in the developing and adult rat prostate, in the pregnant rat mammary gland and adult rat salivary gland. The stained samples were examined using an Edge R400 3D microscope. Images were collected in both single-image and stereo-pair formats. Prostatic basal epithelial cells were found to have a cell body covering an area of 52-62 microns 2. The mean footprint size of basal cells was not significantly different between prostatic lobes. Basal epithelial cells were most dense in the anterior and most sparse in the ventral prostatic lobes. Basal epithelial cells had a large body with many processes, which spread around the duct and projected between luminal cells towards the lumen. These processes closely approached their counterparts from adjacent basal cells. In the developing prostate the differentiation of the basal cells from undifferentiated epithelial cords was observed at the region of ductal widening associated with canalization. Following castration prostatic basal epithelial cells became more closely packed, though the size of individual cells was not significantly changed. There was a two-to four-fold increase in basal cell density by 7 days after surgery. Most prostatic luminal cells were found to have hexagonal bases though some were pentagonal in shape. Luminal cells had a mean basal area of 50 microns 2. In the prostate immunocytochemical staining against smooth muscle alpha-actin revealed discrete bands of muscle surrounding individual prostatic ducts. In the mammary and salivary glands the epithelium was organized into an alveolar arrangement. In the salivary gland a single basal epithelial cell covered the top of each alveolus with processes arranged down the side of the structure. In the mammary gland several basal cells were draped over each alveolus. The mammary and salivary gland basal cells expressed smooth muscle alpha-actin, indicating their myoepithelial phenotype. The organization of the mammary and salivary gland basal cells placed them in an ideal position to squeeze the alveolar structures.


Asunto(s)
Metabolismo Basal , Procesamiento de Imagen Asistido por Computador , Glándulas Mamarias Animales/citología , Próstata/citología , Glándulas Salivales/citología , Actinas/análisis , Animales , Tamaño de la Célula , Células Epiteliales , Femenino , Inmunohistoquímica , Queratinas/análisis , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley
13.
Development ; 106(2): 219-34, 1989 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2591312

RESUMEN

Functional cytodifferentiation of seminal vesicle epithelium was investigated in tissue recombinants. Neonatal rat and mouse seminal vesicles were separated into epithelium and mesenchyme using trypsin. Epithelium and mesenchyme were then recombined in vitro to form interspecific rat/mouse homotypic recombinants. Growth as renal grafts in adult male athymic mice resulted in seminal vesicle morphogenesis in 70% of the recombinants (the remaining 30% failed to grow). Functional cytodifferentiation was judged by the expression of the major androgen-dependent secretory proteins characteristic of the seminal vesicles of adult rats and mice. Antibodies specific for each of these proteins were used to screen tissue sections by immunocytochemistry and to probe protein extracts by immunoblotting techniques. The heterospecific recombinants synthesized the full range of seminal vesicle secretory proteins that typifies the species providing the epithelium of the recombinant, not the mesenchyme. There was little functional variation between individual recombinants. The time course of development corresponded to that of intact neonatal seminal vesicles grown under the same conditions. Morphogenesis and functional cytodifferentiation were not evident after one week, but were well advanced after two weeks. Seminal vesicle recombinants grown for three weeks were indistinguishable morphologically and functionally from normal adult seminal vesicles. In addition, the ability of adult seminal vesicle epithelium to be induced to proliferate was examined. In association with neonatal seminal vesicle mesenchyme, the epithelium of the adult seminal vesicle proliferated and retained its normal functional activity. Thus, seminal vesicle functional cytodifferentiation can be faithfully reproduced in homotypic tissue recombinants. The methods used in this study will be used to investigate seminal vesicle development in instructive inductions of heterotypic epithelia.


Asunto(s)
Vesículas Seminales/citología , Animales , Animales Recién Nacidos , Diferenciación Celular , Electroforesis en Gel de Poliacrilamida , Células Epiteliales , Immunoblotting , Inmunohistoquímica , Masculino , Mesodermo/citología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Morfogénesis , Ratas , Ratas Endogámicas , Vesículas Seminales/crecimiento & desarrollo , Vesículas Seminales/trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA