Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
In Vitro Cell Dev Biol Anim ; 55(10): 861-871, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31529417

RESUMO

The mammary gland develops from the placode at ectodermal invagination. The rudimentary parenchyma (mammary bud) develops mammary trees and alveolar structures, suggesting that the mammary bud consists of stem/progenitor cells. Here, we established a clonal stem cell line from a mammary bud of a p53 null female embryo at day 14.5. FP5-3-1 line was a homogeneous cell population with polygonal epithelial morphology and spontaneously became heterogeneous during passages. Recloning gave rise to four sublines; three sublines have basal epithelial property and one subline has luminal epithelial property. The former sublines generate functional mammary glands when injected into cleared fat pads and the latter subline does not. The cell lines also express many stemness-related genes. The clonal cell lines established in the present study are shown to be mammary stem cells and not tumorigenic. They provide useful models for normal and tumor biology of the mammary gland in vivo and in vitro.


Assuntos
Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/embriologia , Tecido Adiposo/citologia , Animais , Linhagem Celular , Células Cultivadas , Feminino , Expressão Gênica , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Células-Tronco/citologia , Células-Tronco/fisiologia , Proteína Supressora de Tumor p53/genética
3.
J Mammary Gland Biol Neoplasia ; 18(2): 189-97, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23604977

RESUMO

Mammary glands of adult human females are secretory organs comprised of interdependent epithelial and mesenchymal cells. These cells constitute an assemblage of collecting ducts that end in terminal duct lobular units with hollow alveolar ductules that can differentiate to produce and expel milk. Systemic and maternal hormones, autocrine and paracrine growth factors, and cytokines regulate virtually all phases of mammary gland development. During organogenesis, epithelial and mesenchymal cells interact to form precursors of the parenchyma and stroma in the mature gland. Organogenesis precedes five stages of postnatal development: puberty, pregnancy, lactation, involution, and menopause. Each stage requires a specific set of morphogenetic changes in glandular structure and function. Cycles of cell proliferation, differentiation, and involution may recur until menopause. In addition, physiological responses such as inflammation and pathological events such as tumorigenesis are remarkable for their similarities to embryonic morphogenesis. Here we take a succinct look at the ever-improving understanding of stroma-epithelial interactions and mesenchyme function in mammary gland biology.


Assuntos
Carcinogênese/patologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/patologia , Animais , Feminino , Humanos , Células Estromais/patologia
4.
Gynecol Endocrinol ; 21(2): 111-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16109598

RESUMO

During the peri-implantation period, the endometrium undergoes tissue remodeling and cellular rearrangement. To clarify the involvement of matrix metalloproteinases (MMPs) in endometrial remodeling, we isolated total RNAs from the endometrium of non-pregnant and pregnant mice on days 3 to 5 and evaluated mRNA expression of MMP-2, -3, -9, -11 and -13 using reverse transcription-polymerase chain reaction (PCR). Prompt increases in MMP-3 and -13 mRNA were found on day 4 of pregnancy. Quantitative real-time PCR showed that expression of MMP-3 and -13 increased significantly on day 4, up to 8.4 +/- 2.7 times and 3.4 +/- 1.5 times, respectively, the level in non-pregnant endometrium (p < 0.05). On day 4, immunohistochemistry demonstrated MMP-3-positive endometrial stromal cells. At the same time, tenascin-C (TN-C) mRNA increased 11.1 +/- 4.0 times from the level in non-pregnant endometrium (p < 0.004). To clarify regulation of MMP-3 expression, we examined the effects of interleukin-1alpha (IL-1alpha) and TN-C on MMP-3 mRNA in cultured mouse endometrial stromal cells. Both substances resulted in a dose-dependent increase in MMP-3 mRNA (6.1 +/- 1.8-fold at 1 ng/ml of IL-1alpha and 3.9 +/- 1.8-fold at 10 mug/ml of TN-C). This study shows that MMP-3 expression is upregulated in endometrial stromal cells of the peri-implantation period and may be controlled by IL-1alpha and TN-C.


Assuntos
Colagenases/biossíntese , Colagenases/efeitos dos fármacos , Interleucina-1/farmacologia , Tenascina/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Implantação do Embrião/fisiologia , Endométrio/citologia , Endométrio/metabolismo , Feminino , Imuno-Histoquímica , Metaloproteinase 13 da Matriz , Metaloproteinase 3 da Matriz , Camundongos , Camundongos Endogâmicos C3H , Gravidez , RNA Mensageiro/metabolismo , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Regulação para Cima
5.
Am J Pathol ; 167(1): 71-80, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15972953

RESUMO

Tenascin-C (TN-C) is an extracellular matrix molecule that is expressed during wound healing in various tissues. Although not detectable in the normal adult heart, it is expressed under pathological conditions. Previously, using a rat model, we found that TN-C was expressed during the acute stage after myocardial infarction and that alpha-smooth muscle actin (alpha-SMA)-positive myofibroblasts appeared in TN-C-positive areas. In the present study, we examined whether TN-C controls the dynamics of myofibroblast recruitment and wound healing after electrical injury to the myocardium of TN-C knockout (TNKO) mice compared with wild-type (WT) mice. In TNKO mice, myocardial repair seemed to proceed normally, but the appearance of myofibroblasts was delayed. With cultured cardiac fibroblasts, TN-C significantly accelerated cell migration, alpha-SMA expression, and collagen gel contraction but did not affect proliferation. Using recombinant fragments of murine TN-C, the functional domain responsible for promoting migration of cardiac fibroblasts was mapped to the conserved fibronectin type III (FNIII)-like repeats and the fibrinogen (Fbg)-like domain. Furthermore, alternatively spliced FNIII and Fbg-like domains proved responsible for the up-regulation of alpha-SMA expression. These results indicate that TN-C promotes recruitment of myofibroblasts in the early stages of myocardial repair by stimulating cell migration and differentiation.


Assuntos
Movimento Celular/fisiologia , Traumatismos Cardíacos/patologia , Miocárdio/citologia , Tenascina/metabolismo , Cicatrização/fisiologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Fibroblastos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tenascina/genética
6.
Int J Exp Pathol ; 85(6): 373-9, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15566434

RESUMO

Transforming growth factor-beta (TGF-beta), tenascin-C (TN-C) and matrix metalloproteinases (MMPs) have been demonstrated independently to be associated with disease progression and poor prognosis in patients with breast cancer. The present study explored effects of TGF-beta and TN-C on MMP-9 expression and cancer invasion. An experimental study was designed to analyse MDA-MB-231 breast cancer cells, known for their high invasiveness, after stimulation with TGF-beta1 and/or TN-C. TGF-beta1 stimulated TN-C expression in the cells. Co-stimulation of MDA-MB-231 cells with TN-C and TGF-beta increased MMP-9 expression at both the gene (28-fold) and the protein levels. The in vitro invasion also increased (4-fold). GM6001 inhibited the invasion induced by the co-stimulation. The combined effect of TN-C and TGF-beta resulted in enhanced MMP-9 expression and cancer invasion in vitro.


Assuntos
Neoplasias da Mama/enzimologia , Carcinógenos/farmacologia , Metaloproteinase 9 da Matriz/análise , Proteínas de Neoplasias/farmacologia , Tenascina/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Invasividade Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Regulação para Cima/fisiologia
7.
Breast Cancer Res Treat ; 83(1): 43-56, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14997054

RESUMO

A rat strain carrying the human c-Ha-ras protooncogene, established by our laboratory, is highly susceptible to mammary chemical carcinogens. The transgenic rats exhibit increased number of terminal endbuds (TEBs) at the tips of developing ducts in the mammary gland compared to non-transgenic littermates. Confocal microscopy revealed the level of active mitogen-activated protein kinase to be elevated in these TEBs, and a close correlation between their numbers and tumorigenic response initiated by 7,12-dimethylbenz[a]anthracene was confirmed. Single injections of N-methyl-N-nitrosourea into the transgenic rats caused mutations in codon 12 of human c-Ha-ras transgene in TEBs before tumor development, supporting the conclusion that these structures are the major targets of chemical carcinogens. In contrast, with spontaneous development of lesions, alveolar hyperplasia with elevated expression levels of rat and human c-Ha-ras protooncogenes is the first morphological alteration which becomes apparent. Some but not all hyperplastic alveolar nodules were found to harbor mutations in the transgene. The results indicate that elevated expression of c-Ha-ras protooncogene is sufficient in itself to cause a highly proliferative phenotype of mammary alveoli. Our data suggest that TEBs and acini are the major targets for chemical and sporadic carcinogenesis, respectively, in the mammary glands of human c-Ha-ras protooncogene transgenic rats.


Assuntos
Genes ras , Neoplasias Mamárias Experimentais/genética , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Animais Geneticamente Modificados , Carcinógenos/toxicidade , Divisão Celular , Transformação Celular Neoplásica , Primers do DNA , Feminino , Humanos , Hibridização in Situ Fluorescente , Polimorfismo Genético , Polimorfismo de Fragmento de Restrição , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Differentiation ; 71(4-5): 291-8, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12823230

RESUMO

One of a family of extracellular matrix proteins, tenascin-C (TNC) is expressed in a spatiotemporally restricted pattern associated with tissue remodeling during embryonic development, wound healing, cancer invasion and tissue regeneration. Another form, tenascin-X (TNX), is found in most tissues but most predominantly in heart and muscle, often complementarily to TNC. The present analysis demonstrated their expression during early heart development, using mouse lines containing the lacZ gene targeted to the TNC locus, by RT-PCR, immunohistochemistry, and in situ hybridization. TNC was transiently expressed at important steps during heart development: (1) precardiac mesodermal cells differentiating to cardiomyocytes and endocardial cells at E 7.5 - 8.5; (2) cardiomyocytes in the outflow tract at E 8.5 - 12; (3) endocardial cells forming cushion tissue at E 9.5 - 13; and (4) mesenchymal cells in the proepicardial organ (PEO), the precursors of coronary vessels, at E 9.5. When PEO cells were transferred onto the heart surface, the expression of TNC was downregulated, while TNX was upregulated at E 11. Initially, epicardial cells around the AV groove and atrium started to express TNX. TNX-positive cells then gradually spread all over the entire surface of the heart and invaded and formed primitive vascular channels in the myocardium. Despite restricted expression at important sites and steps during cardiogenesis, the hearts of TNC deficient mice developed normally. No difference in the expression pattern of TNX were observed in TNC knockout and wild mice. These results suggest; (1) TNC could play important roles in the differentiation of cardiomyocytes and the early morphogenesis of the heart; (2) TNX could be involved in coronary vasculogenesis; (3) TNX does not compensate for the loss of TNC.


Assuntos
Coração/embriologia , Miocárdio/metabolismo , Tenascina/genética , Animais , Diferenciação Celular/fisiologia , Endocárdio/embriologia , Endocárdio/metabolismo , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica/fisiologia , Tenascina/biossíntese
9.
Am J Pathol ; 162(6): 1857-67, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12759243

RESUMO

Alternative splicing of fibronectin-like type III (FNIII) repeats of tenascin-C (Tn-C) generates a number of splice variants. The distribution of large variants, typical components of provisional extracellular matrices that are up-regulated during tumor stroma remodeling, was here studied by immunoblotting and immunohistochemistry using a monoclonal antibody against the FNIII B domain (named 4C8MS) in a series of human breast cancers. Large Tn-C variants were found at only low levels in normal breast tissues, but were highly expressed at invading sites of intraductal cancers and in the stroma of invasive ductal cancers, especially at invasion fronts. There was a positive correlation between the expression of large Tn-C variants and the cell proliferation rate determined by immunolabeling of the Ki-67 antigen. Of the Tn-C recombinant fragments (all FNIII repeats or mFNIII FL, the conserved FNIII domain only, the epidermal growth factor-like domain, and the fibrinogen-like domain) which were expressed by CHO-K1 cells transfected with mouse Tn-C cDNAs, only the mFNIII FL enhanced in vitro migration and mitotic activity of mammary cancer cells derived from a Tn-C-null mouse. Addition of 4C8MS blocked the function of mFNIII FL. These findings provide strong evidence that the FNIII alternatively spliced region has important roles in tumor progression of breast cancer.


Assuntos
Processamento Alternativo , Neoplasias da Mama/patologia , Tenascina/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Progressão da Doença , Relação Dose-Resposta a Droga , Feminino , Fibronectinas/genética , Fibronectinas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sequências Repetitivas de Ácido Nucleico/genética , Sequências Repetitivas de Ácido Nucleico/imunologia , Tenascina/imunologia , Tenascina/metabolismo , Células Tumorais Cultivadas
10.
Int J Cancer ; 105(1): 53-60, 2003 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12672030

RESUMO

Tenascin-C (TN-C) and matrix metalloproteinases (MMPs) are highly expressed in cancer tissues and probably promote cell migration during cancer progression. TN-C and MMPs are often co-localized in areas of active tissue remodeling in pathologic conditions, suggesting reciprocal regulation. To investigate whether TN-C regulates MMPs expression in cancer cells, we first exposed mammary cancer cells derived from TN-C-deficient mice to TN-C and examined MMPs expression. TN-C was then compared with fibronectin (FN), laminin (LN), basic fibroblast growth factor (b-FGF) and transforming growth factor-beta1 (TGF-beta1). Results of endpoint RT-PCR, quantitative real-time RT-PCR and gelatin zymography demonstrated that TN-C, strongly and dose dependently, upregulates MMP-9 expression in murine mammary cancer cells. TN-C weakly induced MMP-2, MMP-3 and MMP-13. FN and LN induced MMP-9 to lesser extents compared with TN-C. b-FGF had no effect on MMP-9 expression. TGF-beta1 induced MMP-9 expression in a dose-dependent manner, and this induction was significantly enhanced by addition of TN-C. TN-C and TGF-beta1 also upregulated MMP-9 expression in the human breast cancer cell line MDA-MB-231. Neutralization with specific anti-TGF-beta1 antibody showed decreased expression of MMP-9, indicating that TGF-beta controls the baseline MMP-9 expression by a direct autocrine mechanism. Under neutralization of TGF-beta, addition of TN-C still upregulated MMP-9. Conversely, neutralization of endogenous TN-C (in a TN-C-positive mammary cancer cell line) downregulated TGF-beta-induced MMP-9 expression. Thus, TN-C induces MMP-9 expression directly and by collaboration with TGF-beta. These findings reveal a novel role of TN-C in breast cancer progression.


Assuntos
Neoplasias Mamárias Animais/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Tenascina/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , Animais , Meios de Cultivo Condicionados/farmacologia , Meios de Cultura Livres de Soro/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibronectinas/metabolismo , Laminina/metabolismo , Neoplasias Mamárias Animais/metabolismo , Camundongos , Fosforilação , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta1 , Células Tumorais Cultivadas
11.
J Pathol ; 197(3): 388-94, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12115886

RESUMO

Tenascin-C (TNC) is an extracellular matrix protein which appears at active sites of tissue remodelling during embryogenesis or cancer invasion. In normal heart, TNC is only present during the early stages of development but reappears in pathological states. This study examined the diagnostic value of TNC for assessing disease activity of myocarditis. Expression of TNC was examined in myosin-induced autoimmune myocarditis mouse models. Sequential changes in amount, localization and the producing cells were analysed by reverse transcriptase-polymerase chain reaction, western blotting, immunohistochemistry and in situ hybridization and compared with the histological picture. The expression of TNC was upregulated at a very early stage of myocarditis. Immunostaining was detectable before cell infiltration and myocytolysis became histologically apparent, remained during the active stage while cell infiltration and necrosis continued, and disappeared in scar tissue with healing. TNC immunostaining was always observed at the periphery of necrotic or degenerating cardiomyocytes in foci of inflammation, the expression level correlating with histological evidence of inflammatory activity. Interstitial fibroblasts were the major source of TNC, expressing the large isoform containing alternative splicing sites. These data demonstrate that TNC is a useful marker for evaluation of disease activity in myocarditis.


Assuntos
Doenças Autoimunes/diagnóstico , Miocardite/diagnóstico , Miocárdio/química , Tenascina/análise , Doença Aguda , Animais , Biomarcadores/análise , Western Blotting , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos , Modelos Animais , Miosinas , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tenascina/genética
12.
Dev Growth Differ ; 35(3): 271-281, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37281282

RESUMO

In this study the cellular mechanisms of male sterility in F1 hybrids (BNF1) between BALB/c and wild-derived M.MUS-NJL (NJL) was investigated. Cell proliferation and differentiation in the sterile testis were examined by bromodeoxyuridine-labeling and use of germ cell stage-specific antibodies. In BNF1 testes, spermatogonia actively proliferated with a seminiferous epithelial cycle, and were retained in the basal layer of the tubules. However, preleptotene, leptotene and zygotene spermatocytes moved to the adluminal region. Immunohistological data with germ cell stage-specific antibodies indicated the presence of few, if any, pachytene spermatocytes in BNF1 testes. Thus, spermatogenesis seemed to be blocked at the zygotene stage. For examination of germ cell-Sertoli cell interactions, testes of aggregation chimeras between BNF1 and C3H/HeN were analyzed immunohistologically with C3H-specific antibody. Results showed that spermatogenesis of C3H-germ cells was normal, even when these cells in contact with BNF1-Sertoli cells. Differentiation of BNF1-germ cells progressed from zygotene to pachytene stage spermatocytes when these cells were surrounded by C3H-Sertoli cells, but never proceeded beyond the pachytene stage. These observations suggest that at least two different cellular factors may be involved in spermatogenesis, one acting in the germ cells and the other mediated by Sertoli cells. Furthermore, mating experiments revealed that the degree of spermatogenesis varied in different F1 hybrids, and that the major sterility factor was closely linked to the T-locus on chromosome 17.

13.
Dev Growth Differ ; 35(2): 209-220, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37282238

RESUMO

In order to investigate the role of the c-kit oncogene in the melanoblast development, a rat monoclonal antibody (ACK2) against the mouse c-kit protein was used to localize cells expressing c-kit during fetal development. ACK2 was also injected directly into the amniotic cavity of mouse fetuses at successive developmental stages. After birth, the offspring were examined to determine the resulting coat color patterns. c-kit positive melanoblasts first appeared in dermis of fetuses at 11.5 days postcoitum (dpc). Subsequently, these cells increased in number and migrated dorsolaterally to the ventral region, and by 12.5 dpc some of them began to invade the epidermis. Treatment of fetuses by ACK2 microinjection appeared to affect the pigmentation in the coat, inducing a variety of spotting patterns in offspring, and the location of the spots was closely correlated with gestational stage. ACK2 injection of early fetuses produced major changes in coat color even though few c-kit positive cells were detectable in the dermal mesenchyme at the time of injection. Large spots were also induced when mid-stage fetuses with a only few c-kit positive cells in the dorsal region were injected. By contrast, except for spot formation in the center of ventral region, ACK2 injection did not appear to affect melanogenesis in late stage fetuses that had many c-kit positive cells.

14.
Dev Growth Differ ; 30(3): 229-240, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37281156

RESUMO

Insoluble "biomatrix" of mesenchyme is a stimulator of mammary cell differentiation in vitro, but its effect in the morphogenesis is unknown. Fetal salivary mesenchyme induces intense local duct formation when implanted into adult mammary gland. We have therefore tested whether biomatrix prepared from fetal salivary mesenchyme retains this abillity to stimulate duct formation in vivo. Salivary mesenchyme isolated from mouse fetuses at 13.5-14.0 days of gestation, extracted sequentially with water and with 1 M NaCl, then digested with DNAse and RNAse was implanted into mammary glands of female mice and left for periods of 1-35 days. In approximately 40% of recipients, the local epithelium either formed cyst like structures, or else "spikes" of mammary epithelium penetrated the matrix forming a simplified ductwork inside it. Similar responses were elicited by salivary mesenchyme killed by freezing and also by biomatrix prepared from fetal mammary fat pad precursor tissue, mesenchyme of fetal lung, and fetal heart, liver, and brain. However when mesenchyme was either fixed with glutaraldehyde or sonicated and embedded in polymer blocks before implantation, no epithelial response was noted. These observations suggest that the biomatrix provides a passive scaffolding that contributes to morphogenesis of mammary ducts, is insufficient to support normal morphogenesis.

15.
Dev Growth Differ ; 29(4): 351-362, 1987 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37281783

RESUMO

Fourteen-day fetal mammary fat pad precursor tissue (FP) has the capacity to support various fetal epithelia allowing them to accomplish their characteristic development in vivo, without their own mesenchyme (1). This capacity decreases with age of fetal fat pad and is lost postnatally. To analyse the molecular mechanism of such interaction, a method for in vitro duplication of organogenesis is necessary. In the present paper, a co-culture system of fetal epithelium with prospective mammary fat pad is described. The explanted mammary epithelium started budding, then grew out forming branched mammary ducts with end buds. Ultrastructurally, the developing ductal structures exhibited the typical mammary gland morphogenesis. 3 H-Thymidine incorportion assessed by autoradiography showed that the mammary gland morphogenesis in vitro was due to the proliferation of epithelial cells, not merely to a change of the shape of the epithelium. This supportive capacity of 14-day FP also decreased with aging; explanted mammary epithelium did not grow into 17-day FP. When insoluble, non-living biomatrix was used in place of living FP the epithelium grew into the matrix but the resulting structures lacked characteristic morphology of epithelium on living fetal FP. The difference of capacity between 14-day and 17-day tissues was also lost.

16.
Dev Growth Differ ; 26(3): 263-271, 1984.
Artigo em Inglês | MEDLINE | ID: mdl-37281611

RESUMO

Epithelial-mesenchymal interaction in the early development of the anterior pituitary gland was examined by chronological observations on fetal pituitary epithelium grafted in vivo with and without its own mesenchyme. At 8.5 days of gestation, the RATHKE'S pouch began to evaginate toward the diencephalon. The mesenchymal tissue around the pouch was at first very sparsely scattered, but then condensed, on day 10 becoming visible under a dissecting microscope. When RATHKE'S pouch epithelia from 10- and 12-day fetuses were transplanted alone under the kidney capsule, they proliferated slightly to form cysts, the cells of which differentiated into ACTH-producing cells, but not into prolactin-producing cells. Pituitary morphogenesis did not occur. When these epithelia were recombined with homotypic mesenchyme and transplanted, the epithelia proliferated remarkably on one side of the wall of the pouch, resulting in formation of a pars distalis that contained both ACTH-producing cells and prolactin-producing cells. Heterotypic mesenchyme, such as lung, dermis and mammary gland mesenchyme, could induce 12-day epithelium, but not 10-day epithelium to develop into pars distalis. Thus, fetal pituitary epithelium has the capacity of autodifferentiation into ACTH-producing cells, not into prolactin-producing cells, and requires mesenchymal support for development of the pars distalis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...