Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.638
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
EMBO J ; 43(12): 2308-2336, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38760574

RESUMO

How cells coordinate morphogenetic cues and fate specification during development remains a fundamental question in organogenesis. The mammary gland arises from multipotent stem cells (MaSCs), which are progressively replaced by unipotent progenitors by birth. However, the lack of specific markers for early fate specification has prevented the delineation of the features and spatial localization of MaSC-derived lineage-committed progenitors. Here, using single-cell RNA sequencing from E13.5 to birth, we produced an atlas of matched mouse mammary epithelium and mesenchyme and reconstructed the differentiation trajectories of MaSCs toward basal and luminal fate. We show that murine MaSCs exhibit lineage commitment just prior to the first sprouting events of mammary branching morphogenesis at E15.5. We identify early molecular markers for committed and multipotent MaSCs and define their spatial distribution within the developing tissue. Furthermore, we show that the mammary embryonic mesenchyme is composed of two spatially restricted cell populations, and that dermal mesenchyme-produced FGF10 is essential for embryonic mammary branching morphogenesis. Altogether, our data elucidate the spatiotemporal signals underlying lineage specification of multipotent MaSCs, and uncover the signals from mesenchymal cells that guide mammary branching morphogenesis.


Assuntos
Linhagem da Célula , Células Epiteliais , Glândulas Mamárias Animais , Células-Tronco Mesenquimais , Animais , Camundongos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/metabolismo , Feminino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Diferenciação Celular , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 10 de Crescimento de Fibroblastos/genética , Morfogênese , Análise de Célula Única , Mesoderma/citologia , Mesoderma/metabolismo , Mesoderma/embriologia
2.
Physiol Rev ; 100(2): 489-523, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31539305

RESUMO

The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.


Assuntos
Diferenciação Celular , Linhagem da Célula , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Humanas/patologia , Morfogênese , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Transdução de Sinais , Células-Tronco/patologia , Fatores de Transcrição/genética , Microambiente Tumoral
3.
Development ; 151(2)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38276965

RESUMO

The varying pathways of mammary gland development across species and evolutionary history are underexplored, largely due to a lack of model systems. Recent progress in organoid technology holds the promise of enabling in-depth studies of the developmental adaptations that have occurred throughout the evolution of different species, fostering beneficial phenotypes. The practical application of this technology for mammary glands has been mostly confined to rodents and humans. In the current study, we have successfully created next-generation 3D mammary gland organoids from eight eutherian mammals and the first branched organoid of a marsupial mammary gland. Using mammary organoids, we identified a role for ROCK protein in regulating branching morphogenesis, a role that manifests differently in organoids from different mammals. This finding demonstrates the utility of the 3D organoid model for understanding the evolution and adaptations of signaling pathways. These achievements highlight the potential for organoid models to expand our understanding of mammary gland biology and evolution, and their potential utility in studies of lactation or breast cancer.


Assuntos
Glândulas Mamárias Humanas , Marsupiais , Humanos , Feminino , Animais , Marsupiais/genética , Organoides/metabolismo , Lactação , Eutérios , Glândulas Mamárias Animais/metabolismo
4.
Development ; 151(6)2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38391249

RESUMO

Lactation is an essential process for mammals. In sheep, the R96C mutation in suppressor of cytokine signaling 2 (SOCS2) protein is associated with greater milk production and increased mastitis sensitivity. To shed light on the involvement of R96C mutation in mammary gland development and lactation, we developed a mouse model carrying this mutation (SOCS2KI/KI). Mammary glands from virgin adult SOCS2KI/KI mice presented a branching defect and less epithelial tissue, which were not compensated for in later stages of mammary development. Mammary epithelial cell (MEC) subpopulations were modified, with mutated mice having three times as many basal cells, accompanied by a decrease in luminal cells. The SOCS2KI/KI mammary gland remained functional; however, MECs contained more lipid droplets versus fat globules, and milk lipid composition was modified. Moreover, the gene expression dynamic from virgin to pregnancy state resulted in the identification of about 3000 differentially expressed genes specific to SOCS2KI/KI or control mice. Our results show that SOCS2 is important for mammary gland development and milk production. In the long term, this finding raises the possibility of ensuring adequate milk production without compromising animal health and welfare.


Assuntos
Lactação , Glândulas Mamárias Animais , Animais , Feminino , Camundongos , Gravidez , Células Epiteliais/metabolismo , Lactação/genética , Glândulas Mamárias Animais/metabolismo , Leite/metabolismo , Mutação/genética
5.
Nat Immunol ; 16(8): 880-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26147688

RESUMO

Secretory immunoglobulin A (SIgA) shields the gut epithelium from luminal antigens and contributes to host-microbe symbiosis. However, how antibody responses are regulated to achieve sustained host-microbe interactions is unknown. We found that mice and humans exhibited longitudinal persistence of clonally related B cells in the IgA repertoire despite major changes in the microbiota during antibiotic treatment or infection. Memory B cells recirculated between inductive compartments and were clonally related to plasma cells in gut and mammary glands. Our findings suggest that continuous diversification of memory B cells constitutes a central process for establishing symbiotic host-microbe interactions and offer an explanation of how maternal antibodies are optimized throughout life to protect the newborn.


Assuntos
Adaptação Fisiológica/imunologia , Anticorpos/imunologia , Linfócitos B/imunologia , Trato Gastrointestinal/imunologia , Imunoglobulina A Secretora/imunologia , Microbiota/imunologia , Animais , Antibacterianos/farmacologia , Anticorpos/genética , Anticorpos/metabolismo , Linfócitos B/metabolismo , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoglobulina A/genética , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Memória Imunológica/imunologia , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microbiota/genética , Microbiota/fisiologia , Mutação , Plasmócitos/imunologia , Plasmócitos/metabolismo , RNA Ribossômico 16S/genética , Simbiose/efeitos dos fármacos , Simbiose/imunologia , Adulto Jovem
6.
PLoS Biol ; 22(1): e3002093, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38198514

RESUMO

Epithelial branching morphogenesis is an essential process in living organisms, through which organ-specific epithelial shapes are created. Interactions between epithelial cells and their stromal microenvironment instruct branching morphogenesis but remain incompletely understood. Here, we employed fibroblast-organoid or fibroblast-spheroid co-culture systems and time-lapse imaging to reveal that physical contact between fibroblasts and epithelial cells and fibroblast contractility are required to induce mammary epithelial branching. Pharmacological inhibition of ROCK or non-muscle myosin II, or fibroblast-specific knock-out of Myh9 abrogate fibroblast-induced epithelial branching. The process of fibroblast-induced branching requires epithelial proliferation and is associated with distinctive epithelial patterning of yes associated protein (YAP) activity along organoid branches, which is dependent on fibroblast contractility. Moreover, we provide evidence for the in vivo existence of contractile fibroblasts specifically surrounding terminal end buds (TEBs) of pubertal murine mammary glands, advocating for an important role of fibroblast contractility in branching in vivo. Together, we identify fibroblast contractility as a novel stromal factor driving mammary epithelial morphogenesis. Our study contributes to comprehensive understanding of overlapping but divergent employment of mechanically active fibroblasts in developmental versus tumorigenic programs.


Assuntos
Células Epiteliais , Glândulas Mamárias Animais , Camundongos , Animais , Glândulas Mamárias Animais/metabolismo , Células Epiteliais/metabolismo , Morfogênese/fisiologia , Técnicas de Cocultura , Fibroblastos/metabolismo
7.
Nat Methods ; 20(12): 2021-2033, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37919421

RESUMO

Organoid culture has been extensively exploited for normal tissue reconstruction and disease modeling. However, it is still challenging to establish organoids that mimic in vivo-like architecture, size and function under homeostatic conditions. Here we describe the development of a long-term adult stem cell-derived mammary mini gland culture system that supports robust three-dimensional outgrowths recapitulating the morphology, scale, cellular context and transcriptional heterogeneity of the normal mammary gland. The self-organization ability of stem cells and the stability of the outgrowths were determined by a coordinated combination of extracellular matrix, environmental signals and dynamic physiological cycles. We show that these mini glands were hormone responsive and could recapitulate the entire postnatal mammary development including puberty, estrus cycle, lactation and involution. We also observed that these mini glands maintained the presence of mammary stem cells and could also recapitulate the fate transition from embryonic bipotency to postnatal unipotency in lineage tracing assays. In addition, upon induction of oncogene expression in the mini glands, we observed tumor initiation in vitro and in vivo in a mouse model. Together, this study provides an experimental system that can support a dynamic miniature mammary gland for the study of physiologically relevant, complex biological processes.


Assuntos
Glândulas Mamárias Animais , Células-Tronco , Camundongos , Feminino , Animais , Glândulas Mamárias Animais/metabolismo , Carcinogênese , Células Epiteliais
8.
PLoS Biol ; 21(1): e3001583, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36656812

RESUMO

Cell turnover in adult tissues is essential for maintaining tissue homeostasis over a life span and for inducing the morphological changes associated with the reproductive cycle. However, the underlying mechanisms that coordinate the balance of cell death and proliferation remain unsolved. Using the mammary gland, we have discovered that Rac1 acts as a nexus to control cell turnover. Postlactational tissue regression is characterised by the death of milk secreting alveoli, but the process is reversible within the first 48 h if feeding recommences. In mice lacking epithelial Rac1, alveolar regression was delayed. This defect did not result from failed cell death but rather increased cell turnover. Fitter progenitor cells inappropriately divided, regenerating the alveoli, but cell death also concomitantly accelerated. We discovered that progenitor cell hyperproliferation was linked to nonautonomous effects of Rac1 deletion on the macrophageal niche with heightened inflammation. Moreover, loss of Rac1 impaired cell death with autophagy but switched the cell death route to apoptosis. Finally, mammary gland reversibility failed in the absence of Rac1 as the alveoli failed to recommence lactation upon resuckling.


Assuntos
Células Epiteliais , Período Pós-Parto , Proteínas rac1 de Ligação ao GTP , Animais , Feminino , Camundongos , Apoptose/fisiologia , Morte Celular , Células Epiteliais/metabolismo , Lactação , Glândulas Mamárias Animais/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
9.
FASEB J ; 38(7): e23587, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38568835

RESUMO

Mastitis is a disease characterized by congestion, swelling, and inflammation of the mammary gland and usually caused by infection with pathogenic microorganisms. Furthermore, the development of mastitis is closely linked to the exogenous pathway of the gastrointestinal tract. However, the regulatory mechanisms governing the gut-metabolism-mammary axis remain incompletely understood. The present study revealed alterations in the gut microbiota of mastitis rats characterized by an increased abundance of the Proteobacteria phylum. Plasma analysis revealed significantly higher levels of L-isoleucine and cholic acid along with 7-ketodeoxycholic acid. Mammary tissue showed elevated levels of arachidonic acid metabolites and norlithocholic acid. Proteomic analysis showed increased levels of IFIH1, Tnfaip8l2, IRGM, and IRF5 in mastitis rats, which suggests that mastitis triggers an inflammatory response and immune stress. Follistatin (Fst) and progesterone receptor (Pgr) were significantly downregulated, raising the risk of breast cancer. Extracellular matrix (ECM) receptors and focal adhesion signaling pathways were downregulated, while blood-milk barrier integrity was disrupted. Analysis of protein-metabolic network regulation revealed that necroptosis, protein digestion and absorption, and arachidonic acid metabolism were the principal regulatory pathways involved in the development of mastitis. In short, the onset of mastitis leads to changes in the microbiota and alterations in the metabolic profiles of various biological samples, including colonic contents, plasma, and mammary tissue. Key manifestations include disturbances in bile acid metabolism, amino acid metabolism, and arachidonic acid metabolism. At the same time, the integrity of the blood-milk barrier is compromised while inflammation is promoted, thereby reducing cell adhesion in the mammary glands. These findings contribute to a more comprehensive understanding of the metabolic status of mastitis and provide new insights into its impact on the immune system.


Assuntos
Mastite , Infecções Estafilocócicas , Feminino , Humanos , Ratos , Animais , Staphylococcus aureus/fisiologia , Proteômica , Ácido Araquidônico/metabolismo , Mastite/microbiologia , Mastite/patologia , Mastite/veterinária , Inflamação/metabolismo , Redes e Vias Metabólicas , Glândulas Mamárias Animais/metabolismo , Infecções Estafilocócicas/metabolismo
10.
PLoS Biol ; 20(1): e3001518, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35041644

RESUMO

Lipid droplets (LDs) have increasingly been recognized as an essential organelle for eukaryotes. Although the biochemistry of lipid synthesis and degradation is well characterized, the regulation of LD dynamics, including its formation, maintenance, and secretion, is poorly understood. Here, we report that mice lacking Occludin (Ocln) show defective lipid metabolism. We show that LDs were larger than normal along its biogenesis and secretion pathway in Ocln null mammary cells. This defect in LD size control did not result from abnormal lipid synthesis or degradation; rather, it was because of secretion failure during the lactation stage. We found that OCLN was located on the LD membrane and was bound to essential regulators of lipid secretion, including BTN1a1 and XOR, in a C-terminus-dependent manner. Finally, OCLN was a phosphorylation target of Src kinase, whose loss causes lactation failure. Together, we demonstrate that Ocln is a downstream target of Src kinase and promotes LD secretion by binding to BTN1a1 and XOR.


Assuntos
Gotículas Lipídicas/fisiologia , Metabolismo dos Lipídeos , Glândulas Mamárias Animais/metabolismo , Ocludina/metabolismo , Animais , Butirofilinas/metabolismo , Feminino , Lactação/metabolismo , Camundongos , Leite/metabolismo , Ocludina/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
11.
Crit Rev Immunol ; 44(6): 37-47, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38848292

RESUMO

BACKGROUND: Estrogen receptor (ER) signaling plays an important role in the development and functional differentiation of the breast and participates in the process of breast cancer. Activated ER can affect various aspects of the cell's behavior, including proliferation, via modulating the expression of many downstream target genes. Phosphorylation is one of the activation pathways of ER. However, the relationship between estrogen receptor phosphorylation sites and breast development and carcinogenesis is not clear. METHODS: Using Crisper-Cas9 gene editing technology, we constructed ER S309A mutant mice. Using carmine staining of the mammary gland of mice at different developmental stages, we examined the breast development of ER S309A mice. Using hematoxylin-eosin (HE) staining of vaginal smears of mice at the same time for 5 consecutive days, we measured the vaginal epithelial keratinocytes. RESULTS: We established ER S309A mutant mice and observed breast defects in ER S309A mice. In addition, we observed decreased reproductive ability, and estrous cycle disorder in ER S309A mice. The number of vaginal epithelial keratino-cytes in the estrous cycle of ER S309A mice was decreased. CONCLUSION: These results suggest that the phosphorylation site of ER at Serine 309 is important for ER function and breast development.


Assuntos
Serina , Animais , Feminino , Camundongos , Fosforilação , Serina/metabolismo , Humanos , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/genética , Mama/crescimento & desenvolvimento , Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Mutação
12.
Exp Cell Res ; 439(1): 114090, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38740167

RESUMO

Dopamine D2 receptors (D2Rs) play crucial roles in regulating diverse physiological functions of the central nervous system and peripheral organs. D2Rs are also expressed in mammary glands. However, which cell types express D2Rs and whether they are involved in milk production remains unclear. The present findings revealed that D2Rs are expressed in the apical regions of the lateral membranes of mammary epithelial cells (MECs) in lactating mice. We also investigated the effects of the D2R agonist bromocriptine and/or antagonist domperidone on intracellular cAMP levels, milk protein production, and apoptosis in a lactation culture model of MECs that produce major milk components like lactating MECs in vivo. We found that bromocriptine decreased intracellular cAMP levels, whereas domperidone dose-dependently neutralized this effect. Bromocriptine also inhibited casein and lactoferrin production and suppressed activities of STAT5 and glucocorticoid receptors (GRs). Domperidone neutralized the inhibition of casein production as well as STAT5 and GR inactivation induced by bromocriptine. Furthermore, D2R activation by bromocriptine induced apoptosis and inactivated ERK, a signaling molecule responsible for promoting cell proliferation and survival. Domperidone attenuated ERK inactivation and apoptosis induced by bromocriptine. These findings suggest that D2Rs play regulatory roles in milk protein production and apoptosis in MECs.


Assuntos
Apoptose , Bromocriptina , Domperidona , Células Epiteliais , Lactação , Glândulas Mamárias Animais , Proteínas do Leite , Receptores de Dopamina D2 , Animais , Feminino , Camundongos , Apoptose/efeitos dos fármacos , Bromocriptina/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Domperidona/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Lactação/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Proteínas do Leite/metabolismo , Proteínas do Leite/genética , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D2/genética , Fator de Transcrição STAT5/metabolismo
13.
Cell Mol Life Sci ; 81(1): 29, 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38212474

RESUMO

Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 h of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6, and TGFß3, all of which appear to be upregulated by increased intracellular calcium. We further demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis through a process involving inhibition of CDK4/6 and cell cycle progression. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.


Assuntos
Cálcio , Leite , Feminino , Animais , Leite/metabolismo , Cálcio/metabolismo , Morte Celular , Lactação , Lisossomos/metabolismo , Glândulas Mamárias Animais/metabolismo , Fator de Transcrição STAT3/metabolismo
14.
J Mammary Gland Biol Neoplasia ; 29(1): 13, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38916673

RESUMO

Conflicting data exist as to how mammary epithelial cell proliferation changes during the reproductive cycle. To study the effect of endogenous hormone fluctuations on gene expression in the mouse mammary gland, we performed bulk RNAseq analyses of epithelial and stromal cell populations that were isolated either during puberty or at different stages of the adult virgin estrous cycle. Our data confirm prior findings that proliferative changes do not occur in every mouse in every cycle. We also show that during the estrous cycle the main gene expression changes occur in adipocytes and fibroblasts. Finally, we present a comprehensive overview of the Wnt gene expression landscape in different mammary gland cell types in pubertal and adult mice. This work contributes to understanding the effects of physiological hormone fluctuations and locally produced signaling molecules on gene expression changes in the mammary gland during the reproductive cycle and should be a useful resource for future studies investigating gene expression patterns in different cell types across different developmental timepoints.


Assuntos
Células Epiteliais , Perfilação da Expressão Gênica , Glândulas Mamárias Animais , Maturidade Sexual , Células Estromais , Transcriptoma , Animais , Feminino , Camundongos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Células Estromais/metabolismo , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica/métodos , Maturidade Sexual/fisiologia , Proliferação de Células , Ciclo Estral/genética
15.
J Mammary Gland Biol Neoplasia ; 29(1): 10, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38722417

RESUMO

Signal transducers and activators of transcription (STAT) proteins regulate mammary development. Here we investigate the expression of phosphorylated STAT3 (pSTAT3) in the mouse and cow around the day of birth. We present localised colocation analysis, applicable to other mammary studies requiring identification of spatially congregated events. We demonstrate that pSTAT3-positive events are multifocally clustered in a non-random and statistically significant fashion. Arginase-1 expressing cells, consistent with macrophages, exhibit distinct clustering within the periparturient mammary gland. These findings represent a new facet of mammary STAT3 biology, and point to the presence of mammary sub-microenvironments.


Assuntos
Células Epiteliais , Glândulas Mamárias Animais , Fator de Transcrição STAT3 , Animais , Feminino , Bovinos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Células Epiteliais/metabolismo , Fator de Transcrição STAT3/metabolismo , Fosforilação , Gravidez , Parto/fisiologia , Parto/metabolismo , Transdução de Sinais
16.
J Mammary Gland Biol Neoplasia ; 29(1): 11, 2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38761238

RESUMO

The transcription factor STAT3 is activated by multiple cytokines and other extrinsic factors. It plays a key role in immune and inflammatory responses and, when dysregulated, in tumourigenesis. STAT3 is also an indispensable mediator of the cell death process that occurs during post-lactational regression of the mammary gland, one of the most dramatic examples of physiological cell death in adult mammals. During this involution of the gland, STAT3 powerfully enhances the lysosomal system to efficiently remove superfluous milk-producing mammary epithelial cells via a lysosomal-mediated programmed cell death pathway. The lysosome is a membrane-enclosed  cytoplasmic organelle that digests and recycles cellular waste, with an important role as a signalling centre that monitors cellular metabolism. Here, we describe key strategies for investigating the role of STAT3 in regulating lysosomal function using a mammary epithelial cell culture model system. These include protocols for lysosome enrichment and enzyme activity assays, in addition to microscopic analyses of the vesicular compartment in cell lines. Collectively, these approaches provide the tools to investigate multiple aspects of lysosome biogenesis and function, and to define both direct and indirect roles for STAT3.


Assuntos
Células Epiteliais , Lisossomos , Glândulas Mamárias Animais , Fator de Transcrição STAT3 , Lisossomos/metabolismo , Fator de Transcrição STAT3/metabolismo , Feminino , Animais , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/citologia , Humanos , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/citologia , Camundongos , Transdução de Sinais
17.
Dev Biol ; 501: 104-110, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37182733

RESUMO

A healthy mammary gland is a necessity for milk production of dairy goats. The role of chi-miR-3880 in goat lactation is illustrated in our previous study. Among the differentially expressed genes regulated by chi-miR-3880, one seventh were interferon stimulated genes, including MX1, MX2, IFIT3, IFI44L, and DDX58. As the inflammatory cytokine interferon gamma (IFNγ) has been identified as a potential marker of caseous lymphadenitis in lactating sheep, the interaction between IFNγ and immune-related microRNAs was explored in this study. Chi-miR-3880 was found to be one of the microRNAs downregulated by IFNγ in goat mammary epithelial cells (GMECs). The study illustrated that IFNγ/chi-miR-3880/DDX58 axis modulates GMEC proliferation and lipid formation through PI3K/AKT/mTOR pathway, and regulates apoptosis through Caspase-3 and Bcl-2/Bax pathways. The role of the axis in mammary involution was reflected by the expression of p53 and NF-κB. In conclusion, IFNγ/chi-miR-3880/DDX58 axis plays an important part in lactation.


Assuntos
Lactação , MicroRNAs , Feminino , Animais , Ovinos/genética , Lactação/genética , Interferon gama/farmacologia , Interferon gama/metabolismo , Cabras/genética , Cabras/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células Epiteliais/metabolismo , MicroRNAs/metabolismo , Glândulas Mamárias Animais/metabolismo
18.
Physiol Genomics ; 56(2): 128-135, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37955336

RESUMO

The mammary glands are dynamic tissues affected by pregnancy-related hormones during the pregnancy-lactation cycle. Collagen production and its dynamics are essential to the remodeling of the mammary glands. Alterations of the mammary microenvironment and stromal cells during the pregnancy-lactation cycle are important for understanding the physiology of the mammary glands and the development of breast tumors. In this study, we performed an evaluation of collagen dynamics in the mammary fat pad during the pregnancy-lactation cycle. Reanalysis of single-cell RNA-sequencing (scRNA-Seq) data showed the ectopic collagen expression in the immune cells and cell-cell interactions for collagens with single-cell resolution. The scRNA-Seq data showed that type I and type III collagen were produced not only by stromal fibroblasts but also by lymphoid and myeloid cell types in the pregnancy phase. Furthermore, the total cell-cell interaction score for collagen interactions was dramatically increased in the pregnancy tissue. The data presented in this study provide evidence that immune cells contribute, at least in part, to mammary collagen dynamics. Our findings suggest that immune cells, including lymphoid and myeloid cells, might be supportive members of the extracellular matrix orchestration in the pregnancy-lactation cycle of the mammary glands.NEW & NOTEWORTHY Our study evaluated mammary gland collagen dynamics during the pregnancy-lactation cycle using single-cell RNA-sequencing data. We found ectopic collagen expression in immune cells and an increase in collagen interactions during pregnancy. Type I and type III collagen were produced by lymphoid, myeloid, and stromal fibroblast cells during pregnancy. These findings suggest that immune cells, including lymphoid and myeloid cells, play a crucial role in supporting the extracellular matrix in mammary glands during pregnancy-lactation cycles.


Assuntos
Colágeno Tipo III , Colágeno , Gravidez , Feminino , Animais , Colágeno Tipo III/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Lactação/metabolismo , Hormônios/metabolismo , RNA/metabolismo , Glândulas Mamárias Animais/metabolismo
19.
BMC Genomics ; 25(1): 494, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38764031

RESUMO

BACKGROUND: Mammary gland development is a critical process in mammals, crucial for their reproductive success and offspring nourishment. However, the functional roles of key candidate genes associated with teat number, including ABCD4, VRTN, PROX2, and DLST, in this developmental process remain elusive. To address this gap in knowledge, we conducted an in-depth investigation into the dynamic expression patterns, functional implications, and regulatory networks of these candidate genes during mouse mammary gland development. RESULTS: In this study, the spatial and temporal patterns of key genes were characterized in mammary gland development. Using time-series single-cell data, we uncovered differences in the expression of A bcd4, Vrtn, Prox2, and Dlst in cell population of the mammary gland during embryonic and adult stages, while Vrtn was not detected in any cells. We found that only overexpression and knockdown of Abcd4 could inhibit proliferation and promote apoptosis of HC11 mammary epithelial cells, whereas Prox2 and Dlst had no significant effect on these cells. Using RNA-seq and qPCR, further analysis revealed that Abcd4 can induce widespread changes in the expression levels of genes involved in mammary gland development, such as Igfbp3, Ccl5, Tlr2, and Prlr, which were primarily associated with the MAPK, JAK-STAT, and PI3K-AKT pathways by functional enrichment. CONCLUSIONS: These findings revealed ABCD4 as a candidate gene pivotal for regulating mammary gland development and lactation during pregnancy by influencing PRLR expression.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Glândulas Mamárias Animais , Animais , Feminino , Camundongos , Apoptose/genética , Proliferação de Células , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Transdução de Sinais , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo
20.
Breast Cancer Res ; 26(1): 74, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702730

RESUMO

The transcription factor TRPS1 is a context-dependent oncogene in breast cancer. In the mammary gland, TRPS1 activity is restricted to the luminal population and is critical during puberty and pregnancy. Its function in the resting state remains however unclear. To evaluate whether it could be a target for cancer therapy, we investigated TRPS1 function in the healthy adult mammary gland using a conditional ubiquitous depletion mouse model where long-term depletion does not affect fitness. Using transcriptomic approaches, flow cytometry and functional assays, we show that TRPS1 activity is essential to maintain a functional luminal progenitor compartment. This requires the repression of both YAP/TAZ and SRF/MRTF activities. TRPS1 represses SRF/MRTF activity indirectly by modulating RhoA activity. Our work uncovers a hitherto undisclosed function of TRPS1 in luminal progenitors intrinsically linked to mechanotransduction in the mammary gland. It may also provide new insights into the oncogenic functions of TRPS1 as luminal progenitors are likely the cells of origin of many breast cancers.


Assuntos
Glândulas Mamárias Animais , Proteínas Repressoras , Fator de Resposta Sérica , Células-Tronco , Fatores de Transcrição , Animais , Feminino , Camundongos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/citologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Células-Tronco/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Fator de Resposta Sérica/metabolismo , Fator de Resposta Sérica/genética , Humanos , Transativadores/metabolismo , Transativadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA