Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 418
Filtrar
1.
J Invest Dermatol ; 144(6): 1223-1237.e10, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38159590

RESUMO

The Wnt/ß-catenin pathway plays a critical role in cell fate specification, morphogenesis, and stem cell activation across diverse tissues, including the skin. In mammals, the embryonic surface epithelium gives rise to the epidermis as well as the associated appendages including hair follicles and mammary glands, both of which depend on epithelial Wnt/ß-catenin activity for initiation of their development. Later on, Wnts are thought to enhance mammary gland growth and branching, whereas in hair follicles, they are essential for hair shaft formation. In this study, we report a strong downregulation of epithelial Wnt/ß-catenin activity as the mammary bud progresses to branching. We show that forced activation of epithelial ß-catenin severely compromises embryonic mammary gland branching. However, the phenotype of conditional Lef1-deficient embryos implies that a low level of Wnt/ß-catenin activity is necessary for mammary cell survival. Transcriptomic profiling suggests that sustained high ß-catenin activity leads to maintenance of mammary bud gene signature at the expense of outgrowth/branching gene signature. In addition, it leads to upregulation of epidermal differentiation genes. Strikingly, we find a partial switch to hair follicle fate early on upon stabilization of ß-catenin, suggesting that the level of epithelial Wnt/ß-catenin signaling activity may contribute to the choice between skin appendage identities.


Assuntos
Diferenciação Celular , Glândulas Mamárias Animais , Morfogênese , Via de Sinalização Wnt , beta Catenina , Animais , beta Catenina/metabolismo , beta Catenina/genética , Camundongos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Feminino , Via de Sinalização Wnt/fisiologia , Folículo Piloso/embriologia , Folículo Piloso/metabolismo , Folículo Piloso/citologia , Folículo Piloso/crescimento & desenvolvimento , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
2.
J Cell Biol ; 222(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37367826

RESUMO

Branching morphogenesis is an evolutionary solution to maximize epithelial function in a compact organ. It involves successive rounds of branch elongation and branch point formation to generate a tubular network. In all organs, branch points can form by tip splitting, but it is unclear how tip cells coordinate elongation and branching. Here, we addressed these questions in the embryonic mammary gland. Live imaging revealed that tips advance by directional cell migration and elongation relies upon differential cell motility that feeds a retrograde flow of lagging cells into the trailing duct, supported by tip proliferation. Tip bifurcation involved localized repression of cell cycle and cell motility at the branch point. Cells in the nascent daughter tips remained proliferative but changed their direction to elongate new branches. We also report the fundamental importance of epithelial cell contractility for mammary branching morphogenesis. The co-localization of cell motility, non-muscle myosin II, and ERK activities at the tip front suggests coordination/cooperation between these functions.


Assuntos
Células Epiteliais , Glândulas Mamárias Animais , Morfogênese , Divisão Celular , Movimento Celular , Glândulas Mamárias Animais/embriologia , Morfogênese/fisiologia , Mamíferos , Miosina Tipo II/fisiologia
3.
Methods Mol Biol ; 2471: 19-48, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35175590

RESUMO

Multidimensional fluorescence imaging represents a powerful approach for studying the dynamic cellular processes underpinning the development, function, and maintenance of the mammary gland. Here, we describe key multidimensional imaging strategies that enable visualization of mammary branching morphogenesis and epithelial cell fate dynamics during postnatal and embryonic mammary gland development. These include 4-dimensional intravital microscopy and ex vivo imaging of embryonic mammary cultures, in addition to methods that facilitate 3-dimensional imaging of the ductal epithelium at single-cell resolution within its native stroma. Collectively, these approaches provide a window into mammary developmental dynamics, and the perturbations underlying tissue dysfunction and disease.


Assuntos
Células Epiteliais , Glândulas Mamárias Animais , Animais , Epitélio , Microscopia Intravital/métodos , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Morfogênese , Imagem Óptica
4.
Development ; 148(12)2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34128985

RESUMO

Epithelial attachment to the basement membrane (BM) is essential for mammary gland development, yet the exact roles of specific BM components remain unclear. Here, we show that Laminin α5 (Lama5) expression specifically in the luminal epithelial cells is necessary for normal mammary gland growth during puberty, and for alveologenesis during pregnancy. Lama5 loss in the keratin 8-expressing cells results in reduced frequency and differentiation of hormone receptor expressing (HR+) luminal cells. Consequently, Wnt4-mediated crosstalk between HR+ luminal cells and basal epithelial cells is compromised during gland remodeling, and results in defective epithelial growth. The effects of Lama5 deletion on gland growth and branching can be rescued by Wnt4 supplementation in the in vitro model of branching morphogenesis. Our results reveal a surprising role for BM-protein expression in the luminal mammary epithelial cells, and highlight the function of Lama5 in mammary gland remodeling and luminal differentiation.


Assuntos
Diferenciação Celular/genética , Epitélio/metabolismo , Laminina/genética , Glândulas Mamárias Animais/metabolismo , Transdução de Sinais , Proteína Wnt4/genética , Animais , Biomarcadores , Células Epiteliais , Feminino , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Laminina/metabolismo , Glândulas Mamárias Animais/embriologia , Camundongos , Modelos Biológicos , Morfogênese/genética , Organogênese/genética , Proteína Wnt4/metabolismo
5.
J Cell Biol ; 220(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34042944

RESUMO

The mammary gland develops from the surface ectoderm during embryogenesis and proceeds through morphological phases defined as placode, hillock, bud, and bulb stages followed by branching morphogenesis. During this early morphogenesis, the mammary bud undergoes an invagination process where the thickened bud initially protrudes above the surface epithelium and then transforms to a bulb and sinks into the underlying mesenchyme. The signaling pathways regulating the early morphogenetic steps have been identified to some extent, but the underlying cellular mechanisms remain ill defined. Here, we use 3D and 4D confocal microscopy to show that the early growth of the mammary rudiment is accomplished by migration-driven cell influx, with minor contributions of cell hypertrophy and proliferation. We delineate a hitherto undescribed invagination mechanism driven by thin, elongated keratinocytes-ring cells-that form a contractile rim around the mammary bud and likely exert force via the actomyosin network. Furthermore, we show that conditional deletion of nonmuscle myosin IIA (NMIIA) impairs invagination, resulting in abnormal mammary bud shape.


Assuntos
Actomiosina/metabolismo , Movimento Celular , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Mecanotransdução Celular , Animais , Proliferação de Células , Células Epiteliais/ultraestrutura , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Hipertrofia , Queratinócitos/metabolismo , Queratinócitos/ultraestrutura , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/ultraestrutura , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Morfogênese
6.
Dis Model Mech ; 14(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33969421

RESUMO

Little is known about the role of Sox11 in the regulation of mammary progenitor cells. Sox11 is expressed by mammary bud epithelial cells during embryonic mammary gland development and is not detected in mammary epithelial cells after birth. As Sox11 is an oncofetal gene, we investigated the effects of reducing Sox11 levels in embryonic mammary progenitor cells and found that Sox11 regulates proliferative state, stem cell activity and lineage marker expression. We also investigated the effect of reducing Sox11 levels in two transplantable Brca1-deficient oestrogen receptor-negative mouse mammary tumour cell lines, to assess whether Sox11 regulates similar functions in tumour progenitor cells. When Sox11 levels were reduced in one Brca1-deficient mammary tumour cell line that expressed both epithelial and mesenchymal markers, similar effects on proliferation, stem cell activity and expression of lineage markers to those seen in the embryonic mammary progenitor cells were observed. Orthotopic grafting of mammary tumour cells with reduced Sox11 levels led to alterations in tumour-initiating capacity, latency, expression of lineage markers and metastatic burden. Our results support a model in which tumours expressing higher levels of Sox11 have more stem and tumour-initiating cells, and are less proliferative, whereas tumours expressing lower levels of Sox11 become more proliferative and capable of morphogenetic/metastatic growth, similar to what occurs during embryonic mammary developmental progression.


Assuntos
Proteína BRCA1/deficiência , Carcinogênese/metabolismo , Carcinogênese/patologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Fatores de Transcrição SOXC/metabolismo , Animais , Proteína BRCA1/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Linhagem da Célula , Proliferação de Células , Sobrevivência Celular , Células-Tronco Embrionárias/metabolismo , Feminino , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/embriologia , Camundongos , Metástase Neoplásica
7.
Semin Cell Dev Biol ; 114: 83-92, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33472760

RESUMO

Embryonic mammary gland development involves the formation of mammary placodes, invagination of flask-shaped mammary buds and development of miniature bi-layered ductal trees. Currently there is a good understanding of the factors that contribute to ectodermal cell movements to create these appendages and of pathways that lead to mammary specification and commitment. Gene expression profiles of early bipotent mammary stem cells populations as well as cell surface proteins and transcription factors that promote the emergence of unipotent progenitors have been identified. Analyses of these populations has illuminated not only embryonic mammary development, but highlighted parallel processes in breast cancer. Here we provide an overview of the highly conserved pathways that shape the embryonic mammary gland. Understanding the dynamic signaling events that occur during normal mammary development holds considerable promise to advance attempts to eliminate cancer by restoring differentiative signals.


Assuntos
Glândulas Mamárias Animais , Glândulas Mamárias Humanas/embriologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Animais , Modelos Animais de Doenças , Feminino , Humanos , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos
8.
Development ; 147(22)2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-33191272

RESUMO

The mammary gland is a unique tissue and the defining feature of the class Mammalia. It is a late-evolving epidermal appendage that has the primary function of providing nutrition for the young, although recent studies have highlighted additional benefits of milk including the provision of passive immunity and a microbiome and, in humans, the psychosocial benefits of breastfeeding. In this Review, we outline the various stages of mammary gland development in the mouse, with a particular focus on lineage specification and the new insights that have been gained by the application of recent technological advances in imaging in both real-time and three-dimensions, and in single cell RNA sequencing. These studies have revealed the complexity of subpopulations of cells that contribute to the mammary stem and progenitor cell hierarchy and we suggest a new terminology to distinguish these cells.


Assuntos
Desenvolvimento Embrionário/fisiologia , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Humanas/embriologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Organogênese/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Embrião de Mamíferos , Células Epiteliais/fisiologia , Feminino , Humanos , Glândulas Mamárias Animais/citologia , Camundongos , Células-Tronco/fisiologia
9.
Dev Biol ; 463(2): 101-109, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32422143

RESUMO

Loss of expression of the transcription regulator DC-SCRIPT (Zfp366) is a prominent prognostic event in estrogen receptor-positive breast cancer patients. Studying the inherent link between breast morphogenesis and tumorigenesis, we recently reported that DC-SCRIPT affects normal mammary branching morphogenesis and mammary epithelium homeostasis. Here we investigated the molecular mechanism involved in DC-SCRIPT mediated regulation of FGF2 induced mammary branching morphogenesis in a 3D organoid culture system. Our data show that the delayed mammary organoid branching observed in DC-SCRIPT-/- organoids cannot be compensated for by increasing FGF2 levels. Interestingly, FGFR1, the dominant FGF2 receptor, was expressed at a significantly lower level in basal epithelial cells of DC-SCRIPT deficient organoids relative to wildtype organoids. A potential link between DC-SCRIPT and FGFR1 was further supported by the predicted locations of the DC-SCRIPT DNA binding motif at the Fgfr1 gene. Moreover, ERK1/2 phosphorylation downstream of the FGFR1 pathway was decreased in basal epithelial cells of DC-SCRIPT deficient organoids. Altogether, this study shows a relationship between DC-SCRIPT and FGFR1 related pERK signaling in modulating the branching morphogenesis of mammary organoids in vitro.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glândulas Mamárias Animais/embriologia , Proteínas Nucleares/metabolismo , Organogênese , Organoides/embriologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Feminino , Sistema de Sinalização das MAP Quinases , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Organoides/citologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Fatores de Transcrição/genética
10.
Development ; 147(10)2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32444428

RESUMO

Over the past 5 years, several studies have begun to uncover the links between the classical signal transduction pathways and the physical mechanisms that are used to sculpt branched tissues. These advances have been made, in part, thanks to innovations in live imaging and reporter animals. With modern research tools, our conceptual models of branching morphogenesis are rapidly evolving, and the differences in branching mechanisms between each organ are becoming increasingly apparent. Here, we highlight four branched epithelia that develop at different spatial scales, within different surrounding tissues and via divergent physical mechanisms. Each of these organs has evolved to employ unique branching strategies to achieve a specialized final architecture.


Assuntos
Epitélio/metabolismo , Morfogênese/fisiologia , Transdução de Sinais/fisiologia , Animais , Feminino , Humanos , Rim/embriologia , Rim/crescimento & desenvolvimento , Rim/metabolismo , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/embriologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Glândulas Salivares/embriologia , Glândulas Salivares/crescimento & desenvolvimento , Glândulas Salivares/metabolismo
11.
Development ; 147(5)2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32098763

RESUMO

17ß-Estradiol induces the postnatal development of mammary gland and influences breast carcinogenesis by binding to the estrogen receptor ERα. ERα acts as a transcription factor but also elicits rapid signaling through a fraction of ERα expressed at the membrane. Here, we have used the C451A-ERα mouse model mutated for the palmitoylation site to understand how ERα membrane signaling affects mammary gland development. Although the overall structure of physiological mammary gland development is slightly affected, both epithelial fragments and basal cells isolated from C451A-ERα mammary glands failed to grow when engrafted into cleared wild-type fat pads, even in pregnant hosts. Similarly, basal cells purified from hormone-stimulated ovariectomized C451A-ERα mice did not produce normal outgrowths. Ex vivo, C451A-ERα basal cells displayed reduced matrix degradation capacities, suggesting altered migration properties. More importantly, C451A-ERα basal cells recovered in vivo repopulating ability when co-transplanted with wild-type luminal cells and specifically with ERα-positive luminal cells. Transcriptional profiling identified crucial paracrine luminal-to-basal signals. Altogether, our findings uncover an important role for membrane ERα expression in promoting intercellular communications that are essential for mammary gland development.


Assuntos
Epitélio/metabolismo , Receptor alfa de Estrogênio/biossíntese , Glândulas Mamárias Animais/embriologia , Comunicação Parácrina/fisiologia , Animais , Células Epiteliais/metabolismo , Células Epiteliais/transplante , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Lipoilação/fisiologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais
13.
Ecotoxicol Environ Saf ; 188: 109918, 2020 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-31753310

RESUMO

Hormonal regulation controls mammary gland (MG) development. Therefore some hormone-related factors can disrupt the early phases of MGs development, making the gland more susceptible to long term modifications in its response to circulating hormones. Endocrine disruptors, such as bisphenol A (BPA), are able to cause alterations in hormone receptor expression, leading to changes in the cell proliferation index, which may expose the tissue to neoplastic alterations. Thus, we evaluated the variations in hormone receptor expression in the MG of 6-month old Mongolian gerbils exposed to BPA and 17ß estradiol during the perinatal period. Receptors for estrogen alpha (ERα), beta (ERß), progesterone (PGR), prolactin (PRL-R), and co-localization of connexin 43 (Cx43) and ERα in gerbils were analyzed, and serum concentrations of estradiol and progesterone were assessed. No alterations in body, liver, and ovary-uterus complex weights were observed. However, there was an increase in epithelial ERα expression in the 17ß estradiol (E2) group and in PGR in the BPA group. Although immunohistochemistry did not show alterations in ERß expression, western blotting revealed a decrease in this protein in the BPA group. PRL-R was more present in epithelial cells in the vehicle control (VC), E2, and BPA groups in comparison to the intact control group. Cx43 was more frequent in E2 and BPA groups, suggesting a protective response from the gland against possible malignancy. Serum concentration of estradiol reduced in VC, E2, and BPA groups, confirming that alterations also impacts steroid levels. Consequently, perinatal exposure to BPA and the reference endogenous estrogen, 17ß estradiol, are able to increase the tendency of endocrine disruption in MG in a long term manner, since repercussions are observed even 6 months after exposure.


Assuntos
Compostos Benzidrílicos/toxicidade , Disruptores Endócrinos/toxicidade , Estradiol/toxicidade , Glândulas Mamárias Animais/efeitos dos fármacos , Fenóis/toxicidade , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Animais , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Gerbillinae , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente
14.
Development ; 146(23)2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31699800

RESUMO

Fibroblast growth factor (FGF) signaling is crucial for mammary gland development. Although multiple roles for FGF signaling in the epithelium have been described, the function of FGF signaling in mammary stroma has not been elucidated. In this study, we investigated FGF signaling in mammary fibroblasts. We found that murine mammary fibroblasts express FGF receptors FGFR1 and FGFR2 and respond to FGF ligands. In particular, FGF2 and FGF9 induce sustained ERK1/2 signaling and promote fibroblast proliferation and migration in 2D cultures. Intriguingly, only FGF2 induces fibroblast migration in 3D extracellular matrix (ECM) through regulation of actomyosin cytoskeleton and promotes force-mediated collagen remodeling by mammary fibroblasts. Moreover, FGF2 regulates production of ECM proteins by mammary fibroblasts, including collagens, fibronectin, osteopontin and matrix metalloproteinases. Finally, using organotypic 3D co-cultures we show that FGF2 and FGF9 signaling in mammary fibroblasts enhances fibroblast-induced branching of mammary epithelium by modulating paracrine signaling, and that knockdown of Fgfr1 and Fgfr2 in mammary fibroblasts reduces branching of mammary epithelium. Our results demonstrate a pleiotropic role for FGF signaling in mammary fibroblasts, with implications for regulation of mammary stromal functions and epithelial branching morphogenesis.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 9 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/metabolismo , Sistema de Sinalização das MAP Quinases , Glândulas Mamárias Animais/embriologia , Comunicação Parácrina , Animais , Feminino , Fibroblastos/citologia , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Endogâmicos ICR , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo
15.
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
16.
Curr Opin Cell Biol ; 61: 72-78, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31387017

RESUMO

Branching morphogenesis is a fundamental developmental program that generates large epithelial surfaces in a limited three-dimensional space. It is regulated by inductive tissue interactions whose effects are mediated by soluble signaling molecules, and cell-cell and cell-extracellular matrix interactions. Here, we will review recent studies on inductive signaling interactions governing branching morphogenesis in light of phenotypes of mouse mutants and ex vivo organ culture studies with emphasis on developing mammary and salivary glands. We will highlight advances in understanding how cell fate decisions are intimately linked with branching morphogenesis. We will also discuss novel insights into the molecular control of cellular mechanisms driving the formation of these arborized ductal structures and reflect upon how distinct spatial patterns are generated.


Assuntos
Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/metabolismo , Morfogênese/fisiologia , Glândulas Salivares/embriologia , Glândulas Salivares/metabolismo , Animais , Mama/embriologia , Diferenciação Celular , Células Epiteliais/citologia , Matriz Extracelular , Feminino , Camundongos , Técnicas de Cultura de Órgãos , Transdução de Sinais
17.
Animal ; 13(S1): s11-s19, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31280748

RESUMO

Milk production by the sow is a major factor limiting the growth and survival of her litter. Understanding the process of morphogenesis of the sow's mammary gland and the factors that regulate mammary development are important for designing successful management tools that may enhance milk production. Primordia of the mammary glands are first observable in the porcine embryo at approximately 23 days of gestation. The glands then progress through a series of morphologically distinct developmental stages such that, at birth, each mammary gland is composed of the teat, an organized fat pad and two separate lactiferous ducts each with a few ducts branching into the fat pad. The glands continue to grow slowly until about 90 days of age when the rate of growth increases significantly. The increased rate of mammary gland growth coincides with the appearance of large ovarian follicles and an increase in circulating estrogen. After puberty, the continued growth of the gland and elongation and branching of the duct system into the fat pad takes place in response to the elevated levels of estrogen occurring as part of the estrous cycles. After conception, parenchymal mass of each gland increases slowly during early pregnancy and then grows increasingly rapidly during the final trimester. This growth is in response to estrogen, progesterone, prolactin and relaxin. Lobuloalveolar development occurs primarily during late pregnancy. By parturition, the fat pad of the mammary gland has been replaced by colostrum-secreting epithelial cells that line the lumen of the alveoli, lobules and small ducts. All mammary glands develop during pregnancy, however, the extent of development is dependent on the location of the mammary gland on the sow's underline. The mammary glands undergo significant functional differentiation immediately before and after farrowing with the formation of colostrum and the transition through the stages of lactogenesis. Further growth of the glands during lactation is stimulated by milk removal. Individual glands may grow or transiently regress in response to the intensity of suckling during the initial days postpartum. Attempts to enhance milk production by manipulation of mammary development at stages before lactation generally have met with limited success. A more in depth understanding of the processes regulating porcine mammary gland morphogenesis at all stages of development is needed to make further progress.


Assuntos
Colostro/metabolismo , Hormônios Esteroides Gonadais/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Leite/metabolismo , Suínos/crescimento & desenvolvimento , Animais , Células Epiteliais/metabolismo , Estrogênios/metabolismo , Ciclo Estral , Feminino , Desenvolvimento Fetal , Lactação , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/fisiologia , Parto , Gravidez , Progesterona/metabolismo , Prolactina/metabolismo , Suínos/embriologia , Suínos/fisiologia
18.
Toxicology ; 424: 152234, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31201878

RESUMO

Humans are exposed to estrogenic chemicals in food and food packaging, personal care products, and other industrial and consumer goods. Bisphenol A (BPA), a well-studied xenoestrogen, is known to alter development of estrogen-sensitive organs including the brain, reproductive tract, and mammary gland. Bisphenol S (BPS; 4,4'-sulfonyldiphenol), which has a similar chemical structure to BPA, is also used in many consumer products, but its effects on estrogen-sensitive organs in mammals has not been thoroughly examined. Here, we quantified the effects of perinatal exposures to BPS on the male mouse mammary gland. In our first study, pregnant CD-1 mice were orally exposed to BPS (2 or 200 µg/kg/day) starting on pregnancy day 9 through lactation day 20, and male mammary glands were evaluated on embryonic day 16, prior to puberty, and in early adulthood. We observed modest changes in tissue organization in the fetal gland, and significant increases in growth of the gland induced by developmental BPS exposure in adulthood. In our second study, pregnant CD-1 mice were orally exposed to BPS (2, 200 or 2000 µg/kg/day) starting on pregnancy day 9 through lactational day 2. After weaning, the male pups were administered either oil (vehicle) or an estrogen challenge (1 µg ethinyl estradiol/kg/day) for ten days starting prior to puberty. After the 10-day estrogen challenge, we examined hormone-sensitive outcomes including anogenital index (AGI), weight of the seminal vesicles, and morphological parameters of the mammary gland. Although AGI and seminal vesicle weight were not affected by BPS, we observed dose-specific effects on the response of male mammary glands to the peripubertal estrogen challenge. Because male mammary glands are structurally less developed compared to females, they may provide a simple model tissue to evaluate the effects of putative xenoestrogens.


Assuntos
Disruptores Endócrinos/toxicidade , Estrogênios/toxicidade , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Fenóis/toxicidade , Sulfonas/toxicidade , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Feminino , Genitália/efeitos dos fármacos , Genitália/crescimento & desenvolvimento , Masculino , Glândulas Mamárias Animais/embriologia , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Estrogênio/efeitos dos fármacos , Glândulas Seminais/efeitos dos fármacos , Glândulas Seminais/crescimento & desenvolvimento , Maturidade Sexual
19.
Yi Chuan ; 41(5): 384-390, 2019 May 20.
Artigo em Chinês | MEDLINE | ID: mdl-31106774

RESUMO

The pig teat traits are important indices of genetic improvement in pig breeding, which belong to reproductive traits and can directly affect the sows lactation rate and piglet survival rate. Understanding the genetic mechanism underlying the variation of teat traits is of immense value for the improvement of pig reproductive performance. However, the genetic mechanism underlying teat traits (including teat number, type, location distribution, and fluctuating asymmetry) remains elusive. In this review, we summarize the studies on physiology and genetics of teat traits in pigs, including the development process of the mammary gland, the QTL mapping, and candidate gene researches. This review aims to provide a new perspective for the identification of causal mutations and major genes affecting the teat traits and revealing the complex genetic mechanism of the differences in teat number, type and location distribution during embryonic development in pigs.


Assuntos
Cruzamento , Glândulas Mamárias Animais/embriologia , Suínos/genética , Animais , Mapeamento Cromossômico , Feminino , Fenótipo , Gravidez , Locos de Características Quantitativas , Reprodução
20.
Environ Sci Pollut Res Int ; 26(5): 5054-5064, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30607847

RESUMO

Commercial artificial sweeteners present in the market are usually made of combination of nutritive and artificial sweeteners such as sorbitol and aspartame. The aim of this research was to study the effect of in utero exposure to commercial artificial sweeteners on the mouse development and on mammary gland in different stages (18-day embryos and 4-week-old mice). Pregnant mice of treated groups were given 50 mg/kg body weight of commercial artificial sweetener. The dose was given on day 1 of pregnancy until 3-week nursing, while the controls were given distilled water. Congenital malformations were seen in treated 18-day fetus and 4-week-old mice, such as a significant decrease in the diameter of the placenta and the weight of the fetuses, while in 4-week-old mice, a significant decrease in the length of the body, limbs, and tail was seen compared to the controls. The result of this study showed that in 18-day fetuses, clusters of mammary gland in the treated mice seemed to be more differentiated than the controls. In 4-week-old mice, the number of mammary gland ducts in the treated group was significantly more than the control group, and the lumen of the ducts in the treated sections seemed to be narrower than the controls, also many regressing terminal end buds (TEBs) were seen in the treated group. A significant increase in the mammary gland area of treated group was seen compared to the controls.


Assuntos
Glândulas Mamárias Animais/ultraestrutura , Organogênese/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Edulcorantes/toxicidade , Animais , Peso Corporal/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Feminino , Idade Gestacional , Glândulas Mamárias Animais/embriologia , Exposição Materna , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA