Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
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
2.
Res Sq ; 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37398309

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 hours 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 also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis. This is the result of increased TGFß signaling and inhibition of cell cycle progression. Finally, we demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3, a process which also appears to be mediated by TGFß signaling. 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.

3.
Calcif Tissue Int ; 113(2): 246-253, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37358786

RESUMO

The treatment of parathyroid hormone-related protein (PTHrP)-mediated hypercalcemia of malignancy includes treating the malignancy, intravenous fluids, and anti-resorptive therapies such as zoledronic acid or denosumab. PTHrP-mediated hypercalcemia has been reported in benign conditions such as systemic lupus erythematous (SLE) and sarcoidosis and appears to be responsive to glucocorticoids. We report a case of PTHrP-induced hypercalcemia due to a malignancy-low grade fibromyxoid sarcoma-that responded to glucocorticoid treatment. This is the first report of glucocorticoids controlling PTHrP-mediated hypercalcemia of malignancy. Immunohistochemistry of the surgical pathology localized PTHrP staining to the vascular endothelial cells within the tumor. Further studies are needed to elucidate the mechanism of glucocorticoid action in the treatment of PTHrP-mediated hypercalcemia of malignancy.


Assuntos
Hipercalcemia , Sarcoma , Humanos , Proteína Relacionada ao Hormônio Paratireóideo , Hipercalcemia/tratamento farmacológico , Hipercalcemia/etiologia , Hipercalcemia/metabolismo , Glucocorticoides/uso terapêutico , Células Endoteliais
4.
Breast Cancer Res ; 24(1): 30, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35440032

RESUMO

BACKGROUND: Parathyroid hormone-related protein (PTHrP) is required for embryonic breast development and has important functions during lactation, when it is produced by alveolar epithelial cells and secreted into the maternal circulation to mobilize skeletal calcium used for milk production. PTHrP is also produced by breast cancers, and GWAS studies suggest that it influences breast cancer risk. However, the exact functions of PTHrP in breast cancer biology remain unsettled. METHODS: We developed a tetracycline-regulated, MMTV (mouse mammary tumor virus)-driven model of PTHrP overexpression in mammary epithelial cells (Tet-PTHrP mice) and bred these mice with the MMTV-PyMT (polyoma middle tumor-antigen) breast cancer model to analyze the impact of PTHrP overexpression on normal mammary gland biology and in breast cancer progression. RESULTS: Overexpression of PTHrP in luminal epithelial cells caused alveolar hyperplasia and secretory differentiation of the mammary epithelium with milk production. This was accompanied by activation of Stat5 and increased expression of E74-like factor-5 (Elf5) as well as a delay in post-lactation involution. In MMTV-PyMT mice, overexpression of PTHrP (Tet-PTHrP;PyMT mice) shortened tumor latency and accelerated tumor growth, ultimately reducing overall survival. Tumors overproducing PTHrP also displayed increased expression of nuclear pSTAT5 and Elf5, increased expression of markers of secretory differentiation and milk constituents, and histologically resembled secretory carcinomas of the breast. Overexpression of PTHrP within cells isolated from tumors, but not PTHrP exogenously added to cell culture media, led to activation of STAT5 and milk protein gene expression. In addition, neither ablating the Type 1 PTH/PTHrP receptor (PTH1R) in epithelial cells nor treating Tet-PTHrP;PyMT mice with an anti-PTH1R antibody prevented secretory differentiation or altered tumor latency. These data suggest that PTHrP acts in a cell-autonomous, intracrine manner. Finally, expression of PTHrP in human breast cancers is associated with expression of genes involved in milk production and STAT5 signaling. CONCLUSIONS: Our study suggests that PTHrP promotes pathways leading to secretory differentiation and proliferation in both normal mammary epithelial cells and in breast tumor cells.


Assuntos
Neoplasias da Mama , Neoplasias Mamárias Animais , Proteína Relacionada ao Hormônio Paratireóideo , Fator de Transcrição STAT5 , Animais , Neoplasias da Mama/patologia , Feminino , Humanos , Lactação/genética , Glândulas Mamárias Animais , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Camundongos , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo
5.
Nat Metab ; 3(3): 378-393, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33686286

RESUMO

TUG tethering proteins bind and sequester GLUT4 glucose transporters intracellularly, and insulin stimulates TUG cleavage to translocate GLUT4 to the cell surface and increase glucose uptake. This effect of insulin is independent of phosphatidylinositol 3-kinase, and its physiological relevance remains uncertain. Here we show that this TUG cleavage pathway regulates both insulin-stimulated glucose uptake in muscle and organism-level energy expenditure. Using mice with muscle-specific Tug (Aspscr1)-knockout and muscle-specific constitutive TUG cleavage, we show that, after GLUT4 release, the TUG C-terminal cleavage product enters the nucleus, binds peroxisome proliferator-activated receptor (PPAR)γ and its coactivator PGC-1α and regulates gene expression to promote lipid oxidation and thermogenesis. This pathway acts in muscle and adipose cells to upregulate sarcolipin and uncoupling protein 1 (UCP1), respectively. The PPARγ2 Pro12Ala polymorphism, which reduces diabetes risk, enhances TUG binding. The ATE1 arginyltransferase, which mediates a specific protein degradation pathway and controls thermogenesis, regulates the stability of the TUG product. We conclude that insulin-stimulated TUG cleavage coordinates whole-body energy expenditure with glucose uptake, that this mechanism might contribute to the thermic effect of food and that its attenuation could promote obesity.


Assuntos
Metabolismo Energético , Glucose/metabolismo , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células 3T3-L1 , Aminoaciltransferases/metabolismo , Animais , Camundongos , Camundongos Knockout , Oxirredução , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteólise , Termogênese
6.
Endocrinology ; 160(8): 1797-1810, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31087002

RESUMO

Prior studies have demonstrated that the calcium pump, plasma membrane calcium ATPase 2 (PMCA2), mediates calcium transport into milk and prevents mammary epithelial cell death during lactation. PMCA2 also regulates cell proliferation and cell death in breast cancer cells, in part by maintaining the receptor tyrosine kinase ErbB2/HER2 within specialized plasma membrane domains. Furthermore, the regulation of PMCA2 membrane localization and activity in breast cancer cells requires its interaction with the PDZ domain-containing scaffolding molecule sodium-hydrogen exchanger regulatory factor (NHERF) 1. In this study, we asked whether NHERF1 also interacts with PMCA2 in normal mammary epithelial cells during lactation. Our results demonstrate that NHERF1 expression is upregulated during lactation and that it interacts with PMCA2 at the apical membrane of secretory luminal epithelial cells. Similar to PMCA2, NHERF1 expression is rapidly reduced by milk stasis after weaning. Examining lactating NHERF1 knockout (KO) mice showed that NHERF1 contributes to the proper apical location of PMCA2, for proper apical-basal polarity in luminal epithelial cells, and that it participates in the suppression of Stat3 activation and the prevention of premature mammary gland involution. Additionally, we found that PMCA2 also interacts with the closely related scaffolding molecule, NHERF2, at the apical membrane, which likely maintains PMCA2 at the plasma membrane of mammary epithelial cells in lactating NHERF1KO mice. Based on these data, we conclude that, during lactation, NHERF1 is required for the proper expression and apical localization of PMCA2, which, in turn, contributes to preventing the premature activation of Stat3 and the lysosome-mediated cell death pathway that usually occur only early in mammary involution.


Assuntos
Glândulas Mamárias Animais/fisiologia , Fosfoproteínas/fisiologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/análise , Trocadores de Sódio-Hidrogênio/fisiologia , Animais , Polaridade Celular , Feminino , Lactação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas/análise , Trocadores de Sódio-Hidrogênio/análise
7.
J Clin Invest ; 129(8): 3058-3071, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31112135

RESUMO

Lactation induces bone loss to provide sufficient calcium in the milk, a process that involves osteoclastic bone resorption but also osteocytes and perilacunar resorption. The exact mechanisms by which osteocytes contribute to bone loss remain elusive. Osteocytes express genes required in osteoclasts for bone resorption, including cathepsin K (Ctsk), and lactation elevates their expression. We show that Ctsk deletion in osteocytes prevented the increase in osteocyte lacunar area seen during lactation, as well as the effects of lactation to increase osteoclast numbers and decrease trabecular bone volume, cortical thickness and mechanical properties. In addition, Ctsk deletion in osteocytes increased bone Parathyroid Hormone related Peptide (PTHrP), prevented the decrease in serum Parathyroid Hormone (PTH) induced by lactation, but amplified the increase in serum 1,25(OH)2D. The net result of these changes is to maintain serum and milk calcium levels in the normal range, ensuring normal offspring skeletal development. Our studies confirm the fundamental role of osteocytic perilacunar remodeling in physiological states of lactation and provides genetic evidence that osteocyte-derived Ctsk contributes not only to osteocyte perilacunar remodeling, but also to the regulation of PTH, PTHrP, 1,25-Dyhydroxyvitamin D (1,25(OH)2D), osteoclastogenesis and bone loss in response to the high calcium demand associated with lactation.


Assuntos
Catepsina K/fisiologia , Lactação/fisiologia , Osteócitos/fisiologia , Osteoporose/etiologia , Hormônio Paratireóideo/sangue , Animais , Remodelação Óssea/fisiologia , Reabsorção Óssea/etiologia , Reabsorção Óssea/prevenção & controle , Cálcio/análise , Catepsina K/deficiência , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Osteogênese , Osteoporose/prevenção & controle
8.
J Biol Chem ; 294(3): 887-901, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30463939

RESUMO

Unlike other ErbB family members, HER2 levels are maintained on the cell surface when the receptor is activated, allowing prolonged signaling and contributing to its transforming ability. Interactions between HER2, HSP90, PMCA2, and NHERF1 within specialized plasma membrane domains contribute to the membrane retention of HER2. We hypothesized that the scaffolding protein ezrin, which has been shown to interact with NHERF1, might also help stabilize the HER2-PMCA2-NHERF1 complex at the plasma membrane. Therefore, we examined ezrin expression and its relationship with HER2, NHERF1, and PMCA2 levels in murine and human breast cancers. We also used genetic knockdown and/or pharmacologic inhibition of ezrin, HSP90, NHERF1, PMCA2, and HER2 to examine the functional relationships between these factors and membrane retention of HER2. We found ezrin to be expressed at low levels at the apical surface of normal mammary epithelial cells, but its expression is up-regulated and correlates with HER2 expression in hyperplasia and tumors in murine mammary tumor virus-Neu mice, in human HER2-positive breast cancer cell lines, and in ductal carcinoma in situ and invasive breast cancers from human patients. In breast cancer cells, ezrin co-localizes and interacts with HER2, NHERF1, PMCA2, and HSP90 in specialized membrane domains, and inhibiting ezrin disrupts interactions between HER2, PMCA2, NHERF1, and HSP90, inhibiting HER2 signaling and causing PKCα-mediated internalization and degradation of HER2. Inhibition of ezrin synergizes with lapatinib in a PKCα-dependent fashion to inhibit proliferation and promote apoptosis in HER2-positive breast cancer cells. We conclude that ezrin stabilizes a multiprotein complex that maintains active HER2 at the cell surface.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas do Citoesqueleto/antagonistas & inibidores , Lapatinib/farmacologia , Proteína Quinase C-alfa/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Humanos , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Proteína Quinase C-alfa/genética , Receptor ErbB-2/genética , Transdução de Sinais/genética
9.
J Biol Chem ; 292(16): 6555-6568, 2017 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-28235801

RESUMO

We examined whether the scaffolding protein sodium-hydrogen exchanger regulatory factor 1 (NHERF1) interacts with the calcium pump PMCA2 and the tyrosine kinase receptor ErbB2/HER2 in normal mammary epithelial cells and breast cancer cells. NHERF1 interacts with the PDZ-binding motif in PMCA2 in both normal and malignant breast cells. NHERF1 expression is increased in HER2-positive breast cancers and correlates with HER2-positive status in human ductal carcinoma in situ (DCIS) lesions and invasive breast cancers as well as with increased mortality in patients. NHERF1 is part of a multiprotein complex that includes PMCA2, HSP90, and HER2 within specific actin-rich and lipid raft-rich membrane signaling domains. Knocking down NHERF1 reduces PMCA2 and HER2 expression, inhibits HER2 signaling, dissociates HER2 from HSP90, and causes the internalization, ubiquitination, and degradation of HER2. These results demonstrate that NHERF1 acts with PMCA2 to regulate HER2 signaling and membrane retention in breast cancers.


Assuntos
Neoplasias da Mama/metabolismo , Fosfoproteínas/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Receptor ErbB-2/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Motivos de Aminoácidos , Animais , Apoptose , Neoplasias da Mama/genética , Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , RNA Mensageiro/metabolismo , Transdução de Sinais
10.
Cancer Res ; 76(18): 5348-60, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27450451

RESUMO

Parathyroid hormone-related protein (PTHrP) contributes to the development and metastatic progression of breast cancer by promoting hypercalcemia, tumor growth, and osteolytic bone metastases, but it is not known how PTHrP is upregulated in breast tumors. Here we report a central role in this process for the calcium-sensing receptor, CaSR, which enables cellular responses to changes in extracellular calcium, through studies of CaSR-PTHrP interactions in the MMTV-PymT transgenic mouse model of breast cancer and in human breast cancer cells. CaSR activation stimulated PTHrP production by breast cancer cells in vitro and in vivo Tissue-specific disruption of the casr gene in mammary epithelial cells in MMTV-PymT mice reduced tumor PTHrP expression and inhibited tumor cell proliferation and tumor outgrowth. CaSR signaling promoted the proliferation of human breast cancer cell lines and tumor cells cultured from MMTV-PyMT mice. Further, CaSR activation inhibited cell death triggered by high extracellular concentrations of calcium. The actions of the CaSR appeared to be mediated by nuclear actions of PTHrP that decreased p27(kip1) levels and prevented nuclear accumulation of the proapoptotic factor apoptosis inducing factor. Taken together, our findings suggest that CaSR-PTHrP interactions might be a promising target for the development of therapeutic agents to limit tumor cell growth in bone metastases and in other microenvironments in which elevated calcium and/or PTHrP levels contribute to breast cancer progression. Cancer Res; 76(18); 5348-60. ©2016 AACR.


Assuntos
Neoplasias da Mama/patologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Animais , Neoplasias da Mama/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Humanos , Immunoblotting , Camundongos , Camundongos Knockout , Análise Serial de Tecidos
11.
Proc Natl Acad Sci U S A ; 113(3): E282-90, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26729871

RESUMO

In the lactating mammary gland, the plasma membrane calcium ATPase2 (PMCA2) transports milk calcium. Its expression is activated in breast cancers, where high tumor levels predict increased mortality. We find that PMCA2 expression correlates with HER2 levels in breast cancers and that PMCA2 interacts with HER2 in specific actin-rich membrane domains. Knocking down PMCA2 increases intracellular calcium, disrupts interactions between HER2 and HSP-90, inhibits HER2 signaling, and results in internalization and degradation of HER2. Manipulating PMCA2 levels regulates the growth of breast cancer cells, and knocking out PMCA2 inhibits the formation of tumors in mouse mammary tumor virus (MMTV)-Neu mice. These data reveal previously unappreciated molecular interactions regulating HER2 localization, membrane retention, and signaling, as well as the ability of HER2 to generate breast tumors, suggesting that interactions between PMCA2 and HER2 may represent therapeutic targets for breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/patologia , Cálcio/farmacologia , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Endocitose/efeitos dos fármacos , Feminino , Imunofluorescência , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Immunoblotting , Espaço Intracelular/metabolismo , Neoplasias Mamárias Animais , Camundongos , Ligação Proteica , Transporte Proteico , Análise de Sobrevida
12.
Endocrinology ; 156(8): 2762-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25961842

RESUMO

Lactation is associated with increased bone turnover and rapid bone loss, which liberates skeletal calcium used for milk production. Previous studies suggested that an increase in the skeletal expression of receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells ligand (RANKL) coupled with a decrease in osteoprotegerin (OPG) levels likely triggered bone loss during lactation. In this study, we treated lactating mice with recombinant OPG to determine whether bone loss during lactation was dependent on RANKL signaling and whether resorption of the maternal skeleton was required to support milk production. OPG treatment lowered bone resorption rates and completely prevented bone loss during lactation but, surprisingly, did not decrease osteoclast numbers. In contrast, OPG was quite effective at lowering osteoblast numbers and inhibiting bone formation in lactating mice. Furthermore, treatment with OPG during lactation prevented the usual anabolic response associated with reversal of lactational bone loss after weaning. Preventing bone loss had no appreciable effect on milk production, milk calcium levels, or maternal calcium homeostasis when mice were on a standard diet. However, when dietary calcium was restricted, treatment with OPG caused maternal hypocalcemia, maternal death, and decreased milk production. These studies demonstrate that RANKL signaling is a requirement for bone loss during lactation, and suggest that osteoclast activity may be required to increase osteoblast numbers during lactation in preparation for the recovery of bone mass after weaning. These data also demonstrate that maternal bone loss is not absolutely required to supply calcium for milk production unless dietary calcium intake is inadequate.


Assuntos
Reabsorção Óssea/prevenção & controle , Cálcio/metabolismo , Lactação/efeitos dos fármacos , Leite/efeitos dos fármacos , Leite/metabolismo , Osteoprotegerina/uso terapêutico , Animais , Animais Lactentes , Densidade Óssea/efeitos dos fármacos , Cálcio da Dieta/farmacologia , Feminino , Lactação/fisiologia , Camundongos , Mães , Osteoprotegerina/farmacologia , Desmame
13.
Breast Cancer Res ; 16(6): 487, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25467960

RESUMO

INTRODUCTION: Studies have identified multi-potent stem cells in the adult mammary gland. More recent studies have suggested that the embryonic mammary gland may also contain stem/progenitor cells that contribute to initial ductal development. We were interested in determining whether embryonic cells might also directly contribute to long-lived stem cells that support homeostasis and development in the adult mammary gland. METHODS: We used DNA-label retention to detect long label-retaining cells in the mammary gland. Mouse embryos were labeled with 5-ethynl-2'-deoxyuridine (EdU) between embryonic day 14.5 and embryonic day 18.5 and were subsequently sacrificed and examined for EdU retention at various intervals after birth. EdU retaining cells were co-stained for various lineage markers and identified after fluorescence activated cell sorting analysis of specific epithelial subsets. EdU-labeled mice were subjected to subsequent 5-bromo-2'-deoxyuridine administration to determine whether EdU-labeled cells could re-enter the cell cycle. Finally, EdU-labeled cells were grown under non-adherent conditions to assess their ability to form mammospheres. RESULTS: We demonstrate embryonically-derived, long label-retaining cells (eLLRCs) in the adult mammary gland. eLLRCs stain for basal markers and are enriched within the mammary stem cell population identified by cell sorting. eLLRCs are restricted to the primary ducts near the nipple region. Interestingly, long label retaining cells (labeled during puberty) are found just in front of the eLLRCs, near where the ends of the ducts had been at the time of DNA labeling in early puberty. A subset of eLLRCs becomes mitotically active during periods of mammary growth and in response to ovarian hormones. Finally, we show that eLLRCs are contained within primary and secondary mammospheres. CONCLUSIONS: Our findings suggest that a subset of proliferating embryonic cells subsequently becomes quiescent and contributes to the pool of long-lived mammary stem cells in the adult. eLLRCs can re-enter the cell cycle, produce both mammary lineages and self-renew. Thus, our studies have identified a putative stem/progenitor cell population of embryonic origin. Further study of these cells will contribute to an understanding of how quiescent stem cells are generated during development and how fetal exposures may alter future breast cancer risk in adults.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Embrionárias/citologia , Glândulas Mamárias Animais/citologia , Células-Tronco Multipotentes/citologia , Células-Tronco Adultas/metabolismo , Animais , Células-Tronco Embrionárias/metabolismo , Feminino , Glândulas Mamárias Animais/metabolismo , Camundongos , Células-Tronco Multipotentes/metabolismo
14.
PLoS One ; 9(5): e90418, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24785493

RESUMO

Parathyroid hormone-related protein (PTHrP) can be secreted from cells and interact with its receptor, the Type 1 PTH/PTHrP Receptor (PTHR1) in an autocrine, paracrine or endocrine fashion. PTHrP can also remain inside cells and be transported into the nucleus, where its functions are unclear, although recent experiments suggest that it may broadly regulate cell survival and senescence. Disruption of either the PTHrP or PTHR1 gene results in many abnormalities including a failure of embryonic mammary gland development in mice and in humans. In order to examine the potential functions of nuclear PTHrP in the breast, we examined mammary gland development in PTHrP (1-84) knock-in mice, which express a mutant form of PTHrP that lacks the C-terminus and nuclear localization signals and which can be secreted but cannot enter the nucleus. Interestingly, we found that PTHrP (1-84) knock-in mice had defects in mammary mesenchyme differentiation and mammary duct outgrowth that were nearly identical to those previously described in PTHrP-/- and PTHR1-/- mice. However, the mammary buds in PTHrP (1-84) knock-in mice had severe reductions in mutant PTHrP mRNA levels, suggesting that the developmental defects were due to insufficient production of PTHrP by mammary epithelial cells and not loss of PTHrP nuclear function. Examination of the effects of nuclear PTHrP in the mammary gland in vivo will require the development of alternative animal models.


Assuntos
Deleção de Genes , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/metabolismo , Sinais de Localização Nuclear/genética , Proteína Relacionada ao Hormônio Paratireóideo/biossíntese , Proteína Relacionada ao Hormônio Paratireóideo/genética , Domínios e Motivos de Interação entre Proteínas , Animais , Biomarcadores/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Camundongos Transgênicos , Proteína Relacionada ao Hormônio Paratireóideo/química , Fragmentos de Peptídeos/genética , Caracteres Sexuais
15.
Endocrinology ; 154(9): 3031-42, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23782944

RESUMO

To meet the demands for milk calcium, the lactating mother adjusts systemic calcium and bone metabolism by increasing dietary calcium intake, increasing bone resorption, and reducing renal calcium excretion. As part of this adaptation, the lactating mammary gland secretes PTHrP into the maternal circulation to increase bone turnover and mobilize skeletal calcium stores. Previous data have suggested that, during lactation, the breast relies on the calcium-sensing receptor (CaSR) to coordinate PTHrP secretion and milk calcium transport with calcium availability. To test this idea genetically, we bred BLG-Cre mice with CaSR-floxed mice to ablate the CaSR specifically from mammary epithelial cells only at the onset of lactation (CaSR-cKO mice). Loss of the CaSR in the lactating mammary gland did not disrupt alveolar differentiation or milk production. However, it did increase the secretion of PTHrP into milk and decreased the transport of calcium from the circulation into milk. CaSR-cKO mice did not show accelerated bone resorption, but they did have a decrease in bone formation. Loss of the mammary gland CaSR resulted in hypercalcemia, decreased PTH secretion, and increased renal calcium excretion in lactating mothers. Finally, loss of the mammary gland CaSR resulted in decreased calcium accrual by suckling neonates, likely due to the combination of increased milk PTHrP and decreased milk calcium. These results demonstrate that the mammary gland CaSR coordinates maternal bone and calcium metabolism, calcium transport into milk, and neonatal calcium accrual during lactation.


Assuntos
Desenvolvimento Ósseo , Cálcio/metabolismo , Lactação/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Animais , Animais Recém-Nascidos , Transporte Biológico , Reabsorção Óssea/metabolismo , Cálcio/sangue , Cálcio/urina , Cruzamentos Genéticos , Feminino , Regulação da Expressão Gênica , Lactação/sangue , Lactação/urina , Lactoglobulinas/genética , Lactoglobulinas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Leite/metabolismo , Hormônio Paratireóideo/sangue , Hormônio Paratireóideo/genética , Hormônio Paratireóideo/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/sangue , Proteína Relacionada ao Hormônio Paratireóideo/genética , RNA Mensageiro/metabolismo , Receptores de Detecção de Cálcio/genética
16.
Development ; 139(22): 4239-49, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23034629

RESUMO

Parathyroid hormone-related protein (PTHrP) regulates cell fate and specifies the mammary mesenchyme during embryonic development. Loss of PTHrP or its receptor (Pthr1) abolishes the expression of mammary mesenchyme markers and allows mammary bud cells to revert to an epidermal fate. By contrast, overexpression of PTHrP in basal keratinocytes induces inappropriate differentiation of the ventral epidermis into nipple-like skin and is accompanied by ectopic expression of Lef1, ß-catenin and other markers of the mammary mesenchyme. In this study, we document that PTHrP modulates Wnt/ß-catenin signaling in the mammary mesenchyme using a Wnt signaling reporter, TOPGAL-C. Reporter expression is completely abolished by loss of PTHrP signaling and ectopic reporter activity is induced by overexpression of PTHrP. We also demonstrate that loss of Lef1, a key component of the Wnt pathway, attenuates the PTHrP-induced abnormal differentiation of the ventral skin. To characterize further the contribution of canonical Wnt signaling to embryonic mammary development, we deleted ß-catenin specifically in the mammary mesenchyme. Loss of mesenchymal ß-catenin abolished expression of the TOPGAL-C reporter and resulted in mammary buds with reduced expression of mammary mesenchyme markers and impaired sexual dimorphism. It also prevented the ectopic, ventral expression of mammary mesenchyme markers caused by overexpression of PTHrP in basal keratinocytes. Therefore, we conclude that a mesenchymal, canonical Wnt pathway mediates the PTHrP-dependent specification of the mammary mesenchyme.


Assuntos
Glândulas Mamárias Animais/embriologia , Mesoderma/embriologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Via de Sinalização Wnt , Animais , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Queratinócitos/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/biossíntese , Glândulas Mamárias Animais/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Proteína Relacionada ao Hormônio Paratireóideo/deficiência , Proteína Relacionada ao Hormônio Paratireóideo/genética , Receptores de Hormônios Paratireóideos/biossíntese , Receptores de Hormônios Paratireóideos/deficiência , Receptores de Hormônios Paratireóideos/genética , Trombospondinas/metabolismo , Proteínas Wnt/biossíntese , Proteínas Wnt/metabolismo , beta Catenina/biossíntese , beta Catenina/metabolismo
17.
Endocrinology ; 151(12): 5591-601, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21047946

RESUMO

During lactation, calcium is mobilized from the maternal skeleton to supply the breast for milk production. This results in rapid but fully reversible bone loss. Prior studies have suggested that PTHrP, secreted from the breast, and estrogen deficiency, due to suckling-induced central hypogonadism, combine to trigger bone resorption. To determine whether this combination was sufficient to explain bone loss during lactation, we raised PTHrP levels and decreased levels of estrogens in nulliparous mice. PTHrP was infused via osmotic minipumps and estrogens were decreased either by using leuprolide, a long-acting GnRH agonist, or by surgical ovariectomy (OVX). Bone mineral density declined by 23.2 ± 1.3% in the spine and 16.8 ± 1.9% in the femur over 10 d of lactation. This was accompanied by changes in trabecular architecture and an increase in both osteoblast and osteoclast numbers. OVX and PTHrP infusion both induced a modest decline in bone mineral density over 10 d, but leuprolide treatment did not. The combination of OVX and PTHrP was more effective than either treatment alone, but there was no interaction between PTHrP and leuprolide. None of the treatments reproduced the same degree of bone loss caused by lactation. However, both forms of estrogen deficiency led to an increase in osteoclasts, whereas infusion of PTHrP increased both osteoblasts and osteoclasts. Therefore, although the combination of PTHrP and estrogen deficiency contributes to bone loss, it is insufficient to reproduce the full response of the skeleton to lactation, suggesting that other factors also regulate bone metabolism during this period.


Assuntos
Densidade Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Estrogênios/metabolismo , Lactação/fisiologia , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Animais , Densidade Óssea/fisiologia , Osso e Ossos/fisiologia , Feminino , Fármacos para a Fertilidade Feminina/administração & dosagem , Fármacos para a Fertilidade Feminina/farmacologia , Leuprolida/administração & dosagem , Leuprolida/farmacologia , Camundongos , Ovariectomia , Proteína Relacionada ao Hormônio Paratireóideo/administração & dosagem
18.
Dev Dyn ; 238(11): 2713-24, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19795511

RESUMO

Parathyroid hormone-related protein (PTHrP) acts on the mammary mesenchyme and is required for proper embryonic mammary development. In order to understand PTHrP's effects on mesenchymal cells, we profiled gene expression in WT and PTHrP(-/-) mammary buds, and in WT and K14-PTHrP ventral skin at E15.5. By cross-referencing the differences in gene expression between these groups, we identified 35 genes potentially regulated by PTHrP in the mammary mesenchyme, including 6 genes known to be involved in BMP signaling. One of these genes was MMP2. We demonstrated that PTHrP and BMP4 regulate MMP2 gene expression and MMP2 activity in mesenchymal cells. Using mammary bud cultures, we demonstrated that MMP2 acts downstream of PTHrP to stimulate ductal outgrowth. Future studies on the functional role of other genes on this list should expand our knowledge of how PTHrP signaling triggers the onset of ductal outgrowth from the embryonic mammary buds.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glândulas Mamárias Animais/embriologia , Metaloproteinase 2 da Matriz/metabolismo , Morfogênese/genética , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Animais , Células Cultivadas , Dipeptídeos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Ácidos Hidroxâmicos/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Queratinas/metabolismo , Glândulas Mamárias Animais/metabolismo , Inibidores de Metaloproteinases de Matriz , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Relacionada ao Hormônio Paratireóideo/genética , Inibidores de Proteases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Regulação para Cima/fisiologia
19.
Endocrinology ; 148(8): 3875-86, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17495007

RESUMO

A significant portion of milk calcium comes from the mother's skeleton, and lactation is characterized by rapid bone loss. The most remarkable aspect of this bone loss is its complete reversibility, and the time after weaning is the most rapid period of skeletal anabolism in adults. Despite this, little is known of the mechanisms by which the skeleton repairs itself after lactation. We examined changes in bone and calcium metabolism defining the transition from bone loss to bone recovery at weaning in mice. Bone mass decreases during lactation and recovers rapidly after weaning. Lactation causes changes in bone microarchitecture, including thinning and perforation of trabecular plates that are quickly repaired after weaning. Weaning causes a rapid decline in urinary C-telopeptide levels and stimulates an increase in circulating levels of osteocalcin. Bone histomorphometry documented a significant reduction in the numbers of osteoclasts on d 3 after weaning caused by a coordinated wave of osteoclast apoptosis beginning 48 h after pup removal. In contrast, osteoblast numbers and bone formation rates, which are elevated during lactation, remain so 3 d after weaning. The cessation of lactation stimulates an increase in circulating calcium levels and a reciprocal decrease in PTH levels. Finally, weaning is associated with a decrease in levels of receptor activator of nuclear factor-kappaB ligand mRNA in bone. In conclusion, during lactation, bone turnover is elevated, and bone loss is rapid. Weaning causes selective apoptosis of osteoclasts halting bone resorption. The sudden shift in bone turnover favoring bone formation subsequently contributes to the rapid recovery of bone mass.


Assuntos
Apoptose/fisiologia , Lactação/fisiologia , Osteoclastos/citologia , Ligante RANK/genética , Desmame , Absorciometria de Fóton , Animais , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/metabolismo , Feminino , Fêmur/citologia , Fêmur/diagnóstico por imagem , Fêmur/fisiologia , Expressão Gênica/fisiologia , Hipercalcemia/metabolismo , Imageamento Tridimensional , Ligantes , Vértebras Lombares/citologia , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/fisiologia , Camundongos , Camundongos Endogâmicos , NF-kappa B/metabolismo , Osteoclastos/fisiologia , Gravidez , Ligante RANK/metabolismo , Tíbia/citologia , Tíbia/diagnóstico por imagem , Tíbia/fisiologia
20.
Development ; 134(6): 1221-30, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17301089

RESUMO

The mammary glands develop initially as buds arising from the ventral embryonic epidermis. Recent work has shed light on signaling pathways leading to the patterning and formation of the mammary placodes and buds in mouse embryos. Relatively little is known of the signaling pathways that initiate branching morphogenesis and the formation of the ducts from the embryonic buds. Previous studies have shown that parathyroid hormone-related protein (PTHrP; also known as parathyroid hormone-like peptide, Pthlh) is produced by mammary epithelial cells and acts on surrounding mesenchymal cells to promote their differentiation into a mammary-specific dense mesenchyme. As a result of PTHrP signaling, the mammary mesenchyme supports mammary epithelial cell fate, initiates ductal development and patterns the overlying nipple sheath. In this report, we demonstrate that PTHrP acts, in part, by sensitizing mesenchymal cells to BMP signaling. PTHrP upregulates BMP receptor 1A expression in the mammary mesenchyme, enabling it to respond to BMP4, which is expressed within mesenchymal cells underlying the ventral epidermis during mammary bud formation. We demonstrate that BMP signaling is important for outgrowth of normal mammary buds and that BMP4 can rescue outgrowth of PTHrP(-/-) mammary buds. In addition, the combination of PTHrP and BMP signaling is responsible for upregulating Msx2 gene expression within the mammary mesenchyme, and disruption of the Msx2 gene rescues the induction of hair follicles on the ventral surface of mice overexpressing PTHrP in keratinocytes (K14-PTHrP). Our data suggest that PTHrP signaling sensitizes the mammary mesenchyme to the actions of BMP4, triggering outgrowth of the mammary buds and inducing MSX2 expression, which, in turn, leads to lateral inhibition of hair follicle formation within the developing nipple sheath.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Folículo Piloso/embriologia , Proteínas de Homeodomínio/genética , Glândulas Mamárias Animais/embriologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Animais , Proteína Morfogenética Óssea 4 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Proteínas Morfogenéticas Ósseas/genética , Embrião de Mamíferos/metabolismo , Folículo Piloso/metabolismo , Glândulas Mamárias Animais/metabolismo , Mesoderma/química , Mesoderma/metabolismo , Camundongos , Camundongos Mutantes , Proteína Relacionada ao Hormônio Paratireóideo/genética , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Transdução de Sinais , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...