Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 248
Filtrar
1.
J Mammary Gland Biol Neoplasia ; 29(1): 16, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39177859

RESUMO

Postpartum breast cancer (PPBC) is a unique subset of breast cancer, accounting for nearly half of the women diagnosed during their postpartum years. Mammary gland involution is widely regarded as being a key orchestrator in the initiation and progression of PPBC due to its unique wound-healing inflammatory signature. Here, we provide dialogue suggestive that lactation may also facilitate neoplastic development as a result of sterile inflammation. Immune cells are involved in all stages of postnatal mammary development. It has been proposed that the functions of these immune cells are partially directed by mammary epithelial cells (MECs) and the cytokines they produce. This suggests that a more niche area of exploration aimed at assessing activation of innate immune pathways within MECs could provide insight into immune cell contributions to the developing mammary gland. Immune cell contribution to pubertal development and mammary gland involution has been extensively studied; however, investigations into pregnancy and lactation remain limited. During pregnancy, the mammary gland undergoes dramatic expansion to prepare for lactation. As a result, MECs are susceptible to replicative stress. During lactation, mitochondria are pushed to capacity to fulfill the high energetic demands of producing milk. This replicative and metabolic stress, if unresolved, can elicit activation of innate immune pathways within differentiating MECs. In this review, we broadly discuss postnatal mammary development and current knowledge of immune cell contribution to each developmental stage, while also emphasizing a more unique area of study that will be beneficial in the discovery of novel therapeutic biomarkers of PPBC.


Assuntos
Lactação , Glândulas Mamárias Animais , Glândulas Mamárias Humanas , Feminino , Humanos , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/imunologia , Glândulas Mamárias Humanas/patologia , Animais , Lactação/imunologia , Gravidez , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Células Epiteliais/imunologia , Imunidade Inata
2.
Stem Cells Dev ; 33(17-18): 449-467, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38943275

RESUMO

The origin of breast cancer (BC) has traditionally been a focus of medical research. It is widely acknowledged that BC originates from immortal mammary stem cells and that these stem cells participate in two division modes: symmetric cell division (SCD) and asymmetrical cell division (ACD). Although both of these modes are key to the process of breast development and their imbalance is closely associated with the onset of BC, the molecular mechanisms underlying these phenomena deserve in-depth exploration. In this review, we first outline the molecular mechanisms governing ACD/SCD and analyze the role of ACD/SCD in various stages of breast development. We describe that the changes in telomerase activity, the role of polar proteins, and the stimulation of ovarian hormones subsequently lead to two distinct consequences: breast development or carcinogenesis. Finally, gene mutations, abnormalities in polar proteins, modulation of signal-transduction pathways, and alterations in the microenvironment disrupt the balance of BC stem cell division modes and cause BC. Important regulatory factors such as mammalian Inscuteable mInsc, Numb, Eya1, PKCα, PKCθ, p53, and IL-6 also play significant roles in regulating pathways of ACD/SCD and may constitute key targets for future research on stem cell division, breast development, and tumor therapy.


Assuntos
Neoplasias da Mama , Carcinogênese , Glândulas Mamárias Humanas , Humanos , Feminino , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/genética , Animais , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/patologia , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Carcinogênese/patologia , Carcinogênese/metabolismo , Carcinogênese/genética , Células-Tronco/metabolismo , Células-Tronco/citologia , Divisão Celular , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/metabolismo , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Transdução de Sinais
3.
J Mammary Gland Biol Neoplasia ; 28(1): 10, 2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37219601

RESUMO

The ERBB tyrosine kinase receptors and their ligands belong to a complex family that has diverse biological effects and expression profiles in the developing mammary glands, where its members play an essential role in translating hormone signals into local effects. While our understanding of these processes stems mostly from mouse models, there is the potential for differences in how this family functions in the mammary glands of other species, particularly in light of their unique histomorphological features. Herein we review the postnatal distribution and function of ERBB receptors and their ligands in the mammary glands of rodents and humans, as well as for livestock and companion animals. Our analysis highlights the diverse biology for this family and its members across species, the regulation of their expression, and how their roles and functions might be modulated by varying stromal composition and hormone interactions. Given that ERBB receptors and their ligands have the potential to influence processes ranging from normal mammary development to diseased states such as cancer and/or mastitis, both in human and veterinary medicine, a more complete understanding of their biological functions should help to direct future research and the identification of new therapeutic targets.


Assuntos
Receptores ErbB , Glândulas Mamárias Animais , Glândulas Mamárias Humanas , Animais , Feminino , Humanos , Camundongos , Modelos Animais de Doenças , Ligantes , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Animais/crescimento & desenvolvimento
4.
Cells ; 10(10)2021 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-34685621

RESUMO

Breast cancers display dynamic reprogrammed metabolic activities as cancers develop from premalignant lesions to primary tumors, and then metastasize. Numerous advances focus on how tumors develop pro-proliferative metabolic signaling that differs them from adjacent, non-transformed epithelial tissues. This leads to targetable oncogene-driven liabilities among breast cancer subtypes. Other advances demonstrate how microenvironments trigger stress-response at single-cell resolution. Microenvironmental heterogeneities give rise to cell regulatory states in cancer cell spheroids in three-dimensional cultures and at stratified terminal end buds during mammary gland morphogenesis, where stress and survival signaling juxtapose. The cell-state specificity in stress signaling networks recapture metabolic evolution during cancer progression. Understanding lineage-specific metabolic phenotypes in experimental models is useful for gaining a deeper understanding of subtype-selective breast cancer metabolism.


Assuntos
Adaptação Fisiológica , Neoplasias da Mama/patologia , Progressão da Doença , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/patologia , Morfogênese , Estresse Fisiológico , Feminino , Humanos
5.
J Mammary Gland Biol Neoplasia ; 26(3): 221-226, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34448098

RESUMO

The twelfth annual workshop of the European Network for Breast Development and Cancer focused on methods in mammary gland biology and breast cancer, was scheduled to take place on March 26-28, 2020, in Weggis, Switzerland. Due to the COVID-19 pandemic, the meeting was rescheduled twice and eventually happened as a virtual meeting on April 22 and 23, 2021. The main topics of the meeting were branching and development of the mammary gland, tumor microenvironment, circulating tumor cells, tumor dormancy and breast cancer metastasis. Novel and unpublished findings related to these topics were presented, with a particular focus on the methods used to obtain them. Virtual poster sessions were a success, with many constructive and fruitful interactions between researchers and covered many areas of mammary gland biology and breast cancer.


Assuntos
Pesquisa Biomédica/métodos , Neoplasias da Mama/patologia , Glândulas Mamárias Humanas/patologia , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Terapia Combinada , Europa (Continente) , Feminino , Humanos , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Metástase Neoplásica , Estadiamento de Neoplasias , Células Neoplásicas Circulantes , Prognóstico , Microambiente Tumoral
6.
J Mammary Gland Biol Neoplasia ; 26(3): 309-320, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34374886

RESUMO

Mammary gland development primarily occurs postnatally, and this unique process is complex and regulated by systemic hormones and local growth factors. The mammary gland is also a highly dynamic organ that undergoes profound changes at puberty and during the reproductive cycle. These changes are driven by mammary stem cells (MaSCs). Breast cancer is one of the most common causes of cancer-related death in women. Cancer stem cells (CSCs) play prominent roles in tumor initiation, drug resistance, tumor recurrence, and metastasis. The highly conserved Notch signaling pathway functions as a key regulator of the niche mediating mammary organogenesis and breast neoplasia. In this review, we discuss mechanisms by which Notch contributes to breast carcinoma pathology and suggest potentials for therapeutic targeting of Notch in breast cancer. In summary, we provide a comprehensive overview of Notch functions in regulating MaSCs, mammary development, and breast cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Carcinogênese/metabolismo , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Receptores Notch/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Carcinogênese/patologia , Feminino , Humanos , Glândulas Mamárias Humanas/patologia , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/patologia
7.
Dev Cell ; 56(13): 1875-1883, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34256927

RESUMO

Cell fate decisions are critical for adequate tissue development, maintenance and regeneration. In the mammary gland, epithelial cell fates are tightly controlled by the microenvironment. Here, we review how cell fate decisions are regulated by components of the microenvironment during mammary gland development and how pathological changes in the microenvironment can alter cell fates, leading to malignancy. Specifically, we describe the current understanding of how mammary cell fate is controlled and directed by three elements: the extracellular matrix, the immune microenvironment, and hormones-and how these elements can converge to create microenvironments that promote a fourth element: DNA damage.


Assuntos
Microambiente Celular/genética , Matriz Extracelular/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Animais , Mama/crescimento & desenvolvimento , Mama/patologia , Diferenciação Celular/genética , Linhagem da Célula/genética , Feminino , Humanos , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Neoplasias/genética , Neoplasias/patologia , Microambiente Tumoral
8.
Cell Mol Life Sci ; 78(15): 5681-5705, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34156490

RESUMO

17ß-estradiol controls post-natal mammary gland development and exerts its effects through Estrogen Receptor ERα, a member of the nuclear receptor family. ERα is also critical for breast cancer progression and remains a central therapeutic target for hormone-dependent breast cancers. In this review, we summarize the current understanding of the complex ERα signaling pathways that involve either classical nuclear "genomic" or membrane "non-genomic" actions and regulate in concert with other hormones the different stages of mammary development. We describe the cellular and molecular features of the luminal cell lineage expressing ERα and provide an overview of the transgenic mouse models impacting ERα signaling, highlighting the pivotal role of ERα in mammary gland morphogenesis and function and its implication in the tumorigenic processes. Finally, we describe the main features of the ERα-positive luminal breast cancers and their modeling in mice.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Transdução de Sinais/fisiologia , Animais , Carcinogênese/metabolismo , Feminino , Humanos
9.
Nat Commun ; 12(1): 2759, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980857

RESUMO

Epithelial branch elongation is a central developmental process during branching morphogenesis in diverse organs. This fundamental growth process into large arborized epithelial networks is accompanied by structural reorganization of the surrounding extracellular matrix (ECM), well beyond its mechanical linear response regime. Here, we report that epithelial ductal elongation within human mammary organoid branches relies on the non-linear and plastic mechanical response of the surrounding collagen. Specifically, we demonstrate that collective back-and-forth motion of cells within the branches generates tension that is strong enough to induce a plastic reorganization of the surrounding collagen network which results in the formation of mechanically stable collagen cages. Such matrix encasing in turn directs further tension generation, branch outgrowth and plastic deformation of the matrix. The identified mechanical tension equilibrium sets a framework to understand how mechanical cues can direct ductal branch elongation.


Assuntos
Colágeno/fisiologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Organoides/crescimento & desenvolvimento , Fenômenos Biofísicos , Movimento Celular , Células Epiteliais/citologia , Matriz Extracelular/fisiologia , Humanos , Glândulas Mamárias Humanas/citologia , Morfogênese , Organoides/citologia
10.
Semin Cell Dev Biol ; 114: 171-185, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33810979

RESUMO

Understanding the complexity and heterogeneity of mammary cell subpopulations is vital to delineate the mechanisms behind breast cancer development, progression and prevention. Increasingly sophisticated tools for investigating these cell subtypes has led to the development of a greater understanding of these cell subtypes, complex interplay of certain subtypes and their developmental potential. Of note, increasing accessibility and affordability of single cell technologies has led to a plethora of studies being published containing data from mammary cell subtypes and their differentiation potential in both mice and human data sets. Here, we review the different types of single cell technologies and how they have been used to improve our understanding of mammary gland development.


Assuntos
Glândulas Mamárias Humanas/crescimento & desenvolvimento , Análise de Célula Única/métodos , Feminino , Humanos
11.
FEBS J ; 288(19): 5629-5649, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33811729

RESUMO

Many metabolic phenotypes in cancer cells are also characteristic of proliferating nontransformed mammalian cells, and attempts to distinguish between phenotypes resulting from oncogenic perturbation from those associated with increased proliferation are limited. Here, we examined the extent to which metabolic changes corresponding to oncogenic KRAS expression differed from those corresponding to epidermal growth factor (EGF)-driven proliferation in human mammary epithelial cells (HMECs). Removal of EGF from culture medium reduced growth rates and glucose/glutamine consumption in control HMECs despite limited changes in respiration and fatty acid synthesis, while the relative contribution of branched-chain amino acids to the TCA cycle and lipogenesis increased in the near-quiescent conditions. Most metabolic phenotypes measured in HMECs expressing mutant KRAS were similar to those observed in EGF-stimulated control HMECs that were growing at comparable rates. However, glucose and glutamine consumption as well as lactate and glutamate production were lower in KRAS-expressing cells cultured in media without added EGF, and these changes correlated with reduced sensitivity to GLUT1 inhibitor and phenformin treatment. Our results demonstrate the strong dependence of metabolic behavior on growth rate and provide a model to distinguish the metabolic influences of oncogenic mutations and nononcogenic growth.


Assuntos
Neoplasias da Mama/genética , Carcinogênese/genética , Fator de Crescimento Epidérmico/genética , Transportador de Glucose Tipo 1/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Mama/crescimento & desenvolvimento , Mama/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Glucose/metabolismo , Transportador de Glucose Tipo 1/antagonistas & inibidores , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Humanos , Ácido Láctico/metabolismo , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/patologia , Células Tumorais Cultivadas
12.
Breast Cancer Res ; 23(1): 39, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33761981

RESUMO

BACKGROUND: Transforming growth factor beta1 (TGFB1) is a multi-functional cytokine that regulates mammary gland development and cancer progression through endocrine, paracrine and autocrine mechanisms. TGFB1 also plays roles in tumour development and progression, and its increased expression is associated with an increased breast cancer risk. Macrophages are key target cells for TGFB1 action, also playing crucial roles in tumourigenesis. However, the precise role of TGFB-regulated macrophages in the mammary gland is unclear. This study investigated the effect of attenuated TGFB signalling in macrophages on mammary gland development and mammary cancer susceptibility in mice. METHODS: A transgenic mouse model was generated, wherein a dominant negative TGFB receptor is activated in macrophages, in turn attenuating the TGFB signalling pathway specifically in the macrophage population. The mammary glands were assessed for morphological changes through wholemount and H&E analysis, and the abundance and phenotype of macrophages were analysed through immunohistochemistry. Another cohort of mice received carcinogen 7,12-dimethylbenz(a)anthracene (DMBA), and tumour development was monitored weekly. Human non-neoplastic breast tissue was also immunohistochemically assessed for latent TGFB1 and macrophage marker CD68. RESULTS: Attenuation of TGFB signalling resulted in an increase in the percentage of alveolar epithelium in the mammary gland at dioestrus and an increase in macrophage abundance. The phenotype of macrophages was also altered, with inflammatory macrophage markers iNOS and CCR7 increased by 110% and 40%, respectively. A significant decrease in DMBA-induced mammary tumour incidence and prolonged tumour-free survival in mice with attenuated TGFB signalling were observed. In human non-neoplastic breast tissue, there was a significant inverse relationship between latent TGFB1 protein and CD68-positive macrophages. CONCLUSIONS: TGFB acts on macrophage populations in the mammary gland to reduce their abundance and dampen the inflammatory phenotype. TGFB signalling in macrophages increases mammary cancer susceptibility potentially through suppression of immune surveillance activities of macrophages.


Assuntos
Macrófagos/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , 9,10-Dimetil-1,2-benzantraceno/efeitos adversos , Animais , Suscetibilidade a Doenças , Intervalo Livre de Doença , Células Epiteliais/metabolismo , Ciclo Estral , Feminino , Humanos , Inflamação , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Proteína Smad2/metabolismo
13.
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
14.
Semin Cell Dev Biol ; 114: 159-170, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33109441

RESUMO

Prolactin has a rich mechanistic set of actions and signaling in order to elicit developmental effects in mammals. Historically, prolactin has been appreciated as an endocrine peptide hormone that is responsible for final, functional mammary gland development and lactation. Multiple signaling pathways impacted upon by the microenvironment contribute to cell function and differentiation. Endocrine, autocrine and paracrine signaling are now apparent in not only mammary development, but also in cancer, and involve multiple cell types including those of the immune system. Multiple ligands agonists are capable of binding to the prolactin receptor, potentially expanding receptor function. Prolactin has an important role not only in tumorigenesis of the breast, but also in a number of hormonally responsive cancers such as prostate, ovarian and endometrial cancer, as well as pancreatic and lung cancer. Although pituitary and extra-pituitary sources of prolactin such as the epithelium are important, stromal sourced prolactin is now also being recognized as an important factor in tumor progression, all of which potentially signal to multiple cell types in the tumor microenvironment. While prolactin has important roles in milk production including calcium and bone homeostasis, in the disease state it can also affect bone homeostasis. Prolactin also impacts metastatic cancer of the breast to modulate the bone microenvironment and promote bone damage. Prolactin has a fascinating contribution in both physiologic and pathologic settings of mammals.


Assuntos
Glândulas Mamárias Animais , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Neoplasias/etiologia , Prolactina/efeitos adversos , Prolactina/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Metástase Neoplásica
15.
Semin Cell Dev Biol ; 114: 93-112, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33082117

RESUMO

Our understanding of the molecular events underpinning the development of mammalian organ systems has been increasing rapidly in recent years. With the advent of new and improved next-generation sequencing methods, we are now able to dig deeper than ever before into the genomic and epigenomic events that play critical roles in determining the fates of stem and progenitor cells during the development of an embryo into an adult. In this review, we detail and discuss the genes and pathways that are involved in mammary gland development, from embryogenesis, through maturation into an adult gland, to the role of pregnancy signals in directing the terminal maturation of the mammary gland into a milk producing organ that can nurture the offspring. We also provide an overview of the latest research in the single-cell genomics of mammary gland development, which may help us to understand the lineage commitment of mammary stem cells (MaSCs) into luminal or basal epithelial cells that constitute the mammary gland. Finally, we summarize the use of 3D organoid cultures as a model system to study the molecular events during mammary gland development. Our increased investigation of the molecular requirements for normal mammary gland development will advance the discovery of targets to predict breast cancer risk and the development of new breast cancer therapies.


Assuntos
Células Epiteliais/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Animais , Diferenciação Celular , Feminino , Humanos
16.
Semin Cell Dev Biol ; 114: 143-158, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33309487

RESUMO

Mammographic density refers to the radiological appearance of fibroglandular and adipose tissue on a mammogram of the breast. Women with relatively high mammographic density for their age and body mass index are at significantly higher risk for breast cancer. The association between mammographic density and breast cancer risk is well-established, however the molecular and cellular events that lead to the development of high mammographic density are yet to be elucidated. Puberty is a critical time for breast development, where endocrine and paracrine signalling drive development of the mammary gland epithelium, stroma, and adipose tissue. As the relative abundance of these cell types determines the radiological appearance of the adult breast, puberty should be considered as a key developmental stage in the establishment of mammographic density. Epidemiological studies have pointed to the significance of pubertal adipose tissue deposition, as well as timing of menarche and thelarche, on adult mammographic density and breast cancer risk. Activation of hypothalamic-pituitary axes during puberty combined with genetic and epigenetic molecular determinants, together with stromal fibroblasts, extracellular matrix, and immune signalling factors in the mammary gland, act in concert to drive breast development and the relative abundance of different cell types in the adult breast. Here, we discuss the key cellular and molecular mechanisms through which pubertal mammary gland development may affect adult mammographic density and cancer risk.


Assuntos
Densidade da Mama/fisiologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Adulto , Idoso , Feminino , Humanos , Pessoa de Meia-Idade
17.
Semin Cell Dev Biol ; 114: 134-142, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33158729

RESUMO

The essential role of mammary gland stroma in the regulation of mammary epithelial development, function, and cancer has long been recognized. Only recently, though, the functions of individual stromal cell populations have begun to become more clarified. Mammary fibroblasts have emerged as master regulators and modulators of epithelial cell behavior through paracrine signaling, extracellular matrix production and remodeling, and through regulation of other stromal cell types. In this review article, we summarize the crucial studies that helped to untangle the roles of fibroblasts in mammary gland development. Furthermore, we discuss the origin, heterogeneity, and plasticity of mammary fibroblasts during mammary development and cancer progression.


Assuntos
Fibroblastos/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos
18.
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
19.
Growth Factors ; 38(1): 16-24, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-32646254

RESUMO

During the period of lactation, there is extensive growth and development of the mammary gland in order to fulfil the increased demands of milk for the growing infant. Angiogenesis plays a key role in alveolar development and facilitates optimal milk production. Vascular endothelial growth factor (VEGF) is one of the key growth factors regulating angiogenesis in mammary gland. Apart from VEGF, neurotrophins are also known to regulate angiogenesis through direct or indirect mechanisms. Few studies have demonstrated mRNA levels of neurotrophins and their receptors in mammary gland both in humans and rodents. A cross talk between VEGF and neurotrophins has been described in placental development. The enteric and central nervous system are not fully developed at birth, making it imperative to have appropriate levels of angiogenic factors and neurotrophins during postnatal period. The current review summarises studies which describe the role of neurotrophins and angiogenic factors in the mammary gland development.


Assuntos
Glândulas Mamárias Humanas/metabolismo , Fatores de Crescimento Neural/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Humanos , Glândulas Mamárias Humanas/irrigação sanguínea , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Neovascularização Fisiológica , Fatores de Crescimento Neural/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética
20.
Prostate ; 80(13): 1145-1156, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32659025

RESUMO

BACKGROUND: Epithelial stem cells (ESCs) demonstrate a capacity to maintain normal tissues homeostasis and ESCs with a deregulated behavior can contribute to cancer development. The ability to reprogram normal tissue epithelial cells into prostate or mammary stem-like cells holds great promise to help understand cell of origin and lineage plasticity in prostate and breast cancers in addition to understanding normal gland development. We previously showed that an intracellular chemokine, CXCL12γ induced cancer stem cells and neuroendocrine characteristics in both prostate and breast adenocarcinoma cell lines. However, its role in normal prostate or mammary epithelial cell fate and development remains unknown. Therefore, we sought to elucidate the functional role of CXCL12γ in the regulation of ESCs and tissue development. METHODS: Prostate epithelial cells (PNT2) or mammary epithelial cells (MCF10A) with overexpressed CXCL12γ was characterized by quantitative real-time polymerase chain reaction, Western blots, and immunofluorescence for lineage marker expression, and fluorescence activated cell sorting analyses and sphere formation assays to examine stem cell surface phenotype and function. Xenotransplantation animal models were used to evaluate gland or acini formation in vivo. RESULTS: Overexpression of CXCL12γ promotes the reprogramming of cells with a differentiated luminal phenotype to a nonluminal phenotype in both prostate (PNT2) and mammary (MCF10A) epithelial cells. The CXCL12γ-mediated nonluminal type cells results in an increase of epithelial stem-like phenotype including the subpopulation of EPCAMLo /CD49fHi /CD24Lo /CD44Hi cells capable of sphere formation. Critically, overexpression of CXCL12γ promotes the generation of robust gland-like structures from both prostate and mammary epithelial cells in in vivo xenograft animal models. CONCLUSIONS: CXCL12γ supports the reprogramming of epithelial cells into nonluminal cell-derived stem cells, which facilitates gland development.


Assuntos
Quimiocina CXCL12/biossíntese , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Próstata/crescimento & desenvolvimento , Animais , Reprogramação Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Xenoenxertos , Humanos , Masculino , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Camundongos , Próstata/citologia , Próstata/metabolismo , Isoformas de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA