Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Mol Life Sci ; 81(1): 29, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38212474

RESUMEN

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.


Asunto(s)
Calcio , Leche , Femenino , Animales , Leche/metabolismo , Calcio/metabolismo , Muerte Celular , Lactancia , Lisosomas/metabolismo , Glándulas Mamarias Animales/metabolismo , Factor de Transcripción STAT3/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34819374

RESUMEN

Cancer cells can develop an immunosuppressive tumor microenvironment to control tumor-infiltrating lymphocytes. The underlying mechanisms still remain unclear. Here, we report that mouse and human colon cancer cells acquire lymphocyte membrane proteins including cellular markers such as CD4 and CD45. We observed cell populations harboring both a tumor-specific marker and CD4 in the tumor microenvironment. Sorted cells from these populations were capable of forming organoids, identifying them as cancer cells. Live imaging analysis revealed that lymphocyte membrane proteins were transferred to cancer cells via trogocytosis. As a result of the transfer in vivo, cancer cells also acquired immune regulatory surface proteins such as CTLA4 and Tim3, which suppress activation of immune cells [T. L. Walunas et al, Immunity 1, 405-413 (1994) and L. Monney et al., Nature 415, 536-541 (2002)]. RNA sequencing analysis of ex vivo-cocultured splenocytes with trogocytic cancer cells showed reductions in Th1 activation and natural killer cell signaling pathways compared with the nontrogocytic control. Cancer cell trogocytosis was confirmed in the patient-derived xenograft models of colorectal cancer and head and neck cancer. These findings suggest that cancer cells utilize membrane proteins expressed in lymphocytes, which in turn contribute to the development of the immunosuppressive tumor microenvironment.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Antígeno CTLA-4/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Linfocitos Infiltrantes de Tumor/citología , Animales , Células CACO-2 , Membrana Celular/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Células Madre Hematopoyéticas/citología , Humanos , Sistema Inmunológico , Inmunosupresores , Células Jurkat , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Organoides/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Trogocitosis , Microambiente Tumoral
3.
J Biol Chem ; 294(3): 887-901, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30463939

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas del Citoesqueleto/antagonistas & inhibidores , Lapatinib/farmacología , Proteína Quinasa C-alfa/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Femenino , Humanos , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Proteína Quinasa C-alfa/genética , Receptor ErbB-2/genética , Transducción de Señal/genética
4.
Proc Natl Acad Sci U S A ; 113(3): E282-90, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26729871

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/patología , Calcio/farmacología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Endocitosis/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Immunoblotting , Espacio Intracelular/metabolismo , Neoplasias Mamarias Animales , Ratones , Unión Proteica , Transporte de Proteínas , Análisis de Supervivencia
5.
J Biol Chem ; 292(16): 6555-6568, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28235801

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fosfoproteínas/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Receptor ErbB-2/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Secuencias de Aminoácidos , Animales , Apoptosis , Neoplasias de la Mama/genética , Calcio/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones , Microscopía Fluorescente , ARN Mensajero/metabolismo , Transducción de Señal
6.
Traffic ; 14(9): 974-86, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23738536

RESUMEN

Milk lipid is secreted by a unique process, during which triacylglycerol droplets bud from mammary cells coated with an outer bilayer of apical membrane. In all current schemes, the integral protein butyrophilin 1A1 (BTN) is postulated to serve as a transmembrane scaffold, which interacts either with itself or with the peripheral proteins, xanthine oxidoreductase (XOR) and possibly perilipin-2 (PLIN2), to form an immobile bridging complex between the droplet and apical surface. In one such scheme, BTN on the surface of cytoplasmic lipid droplets interacts directly with BTN in the apical membrane without binding to either XOR or PLIN2. We tested these models using both biochemical and morphological approaches. BTN was concentrated in the apical membrane in all species examined and contained mature N-linked glycans. We found no evidence for the association of unprocessed BTN with intracellular lipid droplets. BTN-enhanced green fluorescent protein was highly mobile in areas of mouse milk-lipid droplets that had not undergone post-secretion changes, and endogenous mouse BTN comprised only 0.5-0.7% (w/w) of the total protein, i.e. over 50-fold less than in the milk-lipid droplets of cow and other species. These data are incompatible with models of milk-lipid secretion in which BTN is the major component of an immobile global adhesive complex and suggest that interactions between BTN and other proteins at the time of secretion are more transient than previously predicted. The high mobility of BTN in lipid droplets marks it as a potential mobile signaling molecule in milk.


Asunto(s)
Metabolismo de los Lípidos , Glándulas Mamarias Animales/metabolismo , Leche/metabolismo , Animales , Butirofilinas , Bovinos , Membrana Celular/metabolismo , Femenino , Lactancia , Metabolismo de los Lípidos/fisiología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Polisacáridos/metabolismo
7.
bioRxiv ; 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38659853

RESUMEN

Metastasis is the leading cause of cancer-related mortality. Paneth cells provide stem cell niche factors in homeostatic conditions, but the underlying mechanisms of cancer stem cell niche development are unclear. Here we report that Dickkopf-2 (DKK2) is essential for the generation of cancer cells with Paneth cell properties during colon cancer metastasis. Splenic injection of Dkk2-knockout (KO) cancer organoids into C57BL/6 mice resulted in a significant reduction of liver metastases. Transcriptome analysis showed reduction of Paneth cell markers such as lysozymes in KO organoids. Single cell RNA sequencing analyses of murine metastasized colon cancer cells and patient samples identified the presence of lysozyme positive cells with Paneth cell properties including enhanced glycolysis. Further analyses of transcriptome and chromatin accessibility suggested Hepatocyte nuclear factor 4-alpha (HNF4A) as a downstream target of DKK2. Chromatin immunoprecipitation followed by sequencing analysis revealed that HNF4A binds to the promoter region of Sox9, a well-known transcription factor for Paneth cell differentiation. In the liver metastatic foci, DKK2 knockout rescued HNF4A protein levels followed by reduction of lysozyme positive cancer cells. Taken together, DKK2-mediated reduction of HNF4A protein promotes the generation of lysozyme positive cancer cells with Paneth cell properties in the metastasized colon cancers.

8.
Res Sq ; 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37398309

RESUMEN

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.

9.
Cells ; 10(10)2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34685653

RESUMEN

The survival rates for breast cancer (BC) have improved in recent years, but resistance, metastasis, and recurrence still remain major therapeutic challenges for BC. The acidic tumor microenvironment (TME) has attracted attention because of its association with tumorigenesis, metastasis, drug resistance, and immune surveillance. In this study, we evaluated natural compounds from traditional herbal medicine used to treat cancer that selectively target genes regulated by extracellular acidosis. We integrated four transcriptomic data including BC prognostic data from The Cancer Genome Atlas database, gene expression profiles of MCF-7 cells treated with 102 natural compounds, patterns of gene profiles by acidic condition, and single-cell RNA-sequencing from BC patient samples. Bruceine D (BD) was predicted as having the highest therapeutic potential, having an information gain (IG) score of 0.24, to regulate reprogrammed genes driven by acidosis affecting the survival of BC patients. BD showed the highest IG on EMT (IG score: 0.11) and invasion (IG score: 0.1) compared to the other phenotypes with the CancerSEA database. BD also demonstrated therapeutic potential by interfering with the tumor cell-TME interactions by reducing the amyloid beta precursor protein and CD44 expression. Therefore, BD is a potential candidate to target the acidic TME induced metastatic process in BC.


Asunto(s)
Ácidos/química , Productos Biológicos/farmacología , Neoplasias de la Mama/patología , Simulación por Computador , Espacio Extracelular/metabolismo , Neoplasias de la Mama/genética , Comunicación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Pronóstico , Cuassinas/farmacología , RNA-Seq , Análisis de la Célula Individual
10.
Cell Rep ; 37(13): 110160, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34965434

RESUMEN

The lipid raft-resident protein, MAL2, has been implicated as contributing to the pathogenesis of several malignancies, including breast cancer, but the underlying mechanism for its effects on tumorigenesis is unknown. Here, we show that MAL2-mediated lipid raft formation leads to HER2 plasma membrane retention and enhanced HER2 signaling in breast cancer cells. We demonstrate physical interactions between HER2 and MAL2 in lipid rafts using proximity ligation assays. Super-resolution structured illumination microscopy imaging displays the structural organization of the HER2/Ezrin/NHERF1/PMCA2 protein complex. Formation of this protein complex maintains low intracellular calcium concentrations in the vicinity of the plasma membrane. HER2/MAL2 protein interactions in lipid rafts are enhanced in trastuzumab-resistant breast cancer cells. Our findings suggest that MAL2 is crucial for lipid raft formation, HER2 signaling, and HER2 membrane stability in breast cancer cells, suggesting MAL2 as a potential therapeutic target.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas del Citoesqueleto/metabolismo , Resistencia a Antineoplásicos , Microdominios de Membrana/metabolismo , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/metabolismo , Fosfoproteínas/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Receptor ErbB-2/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Proteínas del Citoesqueleto/genética , Endocitosis , Femenino , Humanos , Microdominios de Membrana/efectos de los fármacos , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/genética , Fosfoproteínas/genética , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Receptor ErbB-2/genética , Intercambiadores de Sodio-Hidrógeno/genética , Trastuzumab/farmacología , Células Tumorales Cultivadas
11.
FASEB Bioadv ; 3(12): 971-997, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34938960

RESUMEN

Butyrophilin 1A1 (BTN1A1) is implicated in the secretion of lipid droplets from mammary epithelial cells as a membrane receptor, which forms a secretion complex with the redox enzyme, xanthine oxidoreductase (XDH). The first evidence that BTN1A1 functions in this process was the generation of Btn1a1 -/- mouse lines, in which lipid secretion was disrupted and large unstable droplets were released into alveolar spaces with fragmented surface membranes. We have revisited one of these mutant mouse lines using RNAseq and proteomic analysis to assess the consequences of ablating the Btn1a1 gene on the expression of other genes and proteins. Disruption of intact Btn1a1 protein expression led to a large build-up of Xdh in the cytoplasm, induction of acute phase response genes and Lif-activation of Stat3 phosphorylation. At peak lactation, approx. 10% of the cells were dying, as assessed by TUNEL-analysis of nuclear DNA. Possible cell death pathways included expression of caspase 8 and activated caspase 3, autophagy, Slc5a8-mediated inactivation of survivin (Birc5), and pStat3-mediated lysosomal lysis, the latter of which is the principal death route in involuting wild type cells. Milk secretion was prolonged by renewal of the secretory epithelium, as evidenced by the upregulation of Ki67 in approx. 10% of cell nuclei and expression of cyclins and Fos/Jun. These data highlight the plasticity of the mammary epithelium and the importance of functional BTN1A1 expression for maintenance of terminally differentiated secretory cells and optimal milk production throughout lactation.

12.
iScience ; 24(5): 102411, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-33997693

RESUMEN

Enhanced stemness in colorectal cancer has been reported and it contributes to aggressive progression, but the underlying mechanisms remain unclear. Here we report a Wnt ligand, Dickkopf-2 (DKK2) is essential for developing colorectal cancer stemness. Genetic depletion of DKK2 in intestinal epithelial or stem cells reduced tumorigenesis and expression of the stem cell marker genes including LGR5 in a model of colitis-associated cancer. Sequential mutations in APC, KRAS, TP53, and SMAD4 genes in colonic organoids revealed a significant increase of DKK2 expression by APC knockout and further increased by additional KRAS and TP53 mutations. Moreover, DKK2 activates proto-oncogene tyrosine-protein kinse Src followed by increased LGR5 expressing cells in colorectal cancer through degradation of HNF4α1 protein. These findings suggest that DKK2 is required for colonic epithelial cells to enhance LGR5 expression during the progression of colorectal cancer.

13.
Curr Opin Endocr Metab Res ; 15: 63-70, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33299957

RESUMEN

Lactation requires a series of adaptations in maternal calcium and bone metabolism to ensure a steady supply of calcium to the lactating mammary gland. The alterations in systemic metabolism are accompanied by alterations in the expression of calcium receptors, channels, binding proteins, pumps and transporters in mammary epithelial cells to increase the uptake of calcium from the extracellular fluid and to transport it into milk. Intracellular calcium regulates signaling pathways that mediate changes in cell proliferation, differentiation and death and many of the molecules involved in supporting and coordinating calcium secretion into milk are re-expressed and redeployed to support malignant behavior in breast cancer cells. In this article, we review adaptations of systemic calcium homeostasis during lactation, as well as the mechanisms of milk calcium transport. We then discuss how reactivation of these pathways contributes to the pathophysiology of breast cancer.

14.
Endocrinology ; 160(8): 1797-1810, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31087002

RESUMEN

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.


Asunto(s)
Glándulas Mamarias Animales/fisiología , Fosfoproteínas/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/análisis , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Polaridad Celular , Femenino , Lactancia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/análisis , Intercambiadores de Sodio-Hidrógeno/análisis
15.
PLoS One ; 12(4): e0174849, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28369073

RESUMEN

ErbB2/HER2/Neu is a receptor tyrosine kinase that is overexpressed in 25-30% of human breast cancers, usually associated with amplification of the ERBB2 gene. HER2 has no recognized ligands and heterodimers between HER2 and EGFR (ErbB1/HER1) or HER2 and ErbB3/HER3 are important in breast cancer. Unlike other ErbB family members, HER2 is resistant to internalization and degradation, and remains at the cell surface to signal for prolonged periods after it is activated. Although the mechanisms underlying retention of HER2 at the cell surface are not fully understood, prior studies have shown that, in order to avoid internalization, HER2 must interact with the chaperone, HSP90, and the calcium pump, PMCA2, within specific plasma membrane domains that protrude from the cell surface. In this report, we demonstrate that HER2 signaling, itself, is important for the formation and maintenance of membrane protrusions, at least in part, by maintaining PMCA2 expression and preventing increased intracellular calcium concentrations. Partial genetic knockdown of HER2 expression or pharmacologic inhibition of HER2 signaling causes the depletion of membrane protrusions and disruption of the interactions between HER2 and HSP90. This is associated with the ubiquitination of HER2, its internalization with EGFR or HER3, and its degradation. These results suggest a model by which some threshold of HER2 signaling is required for the formation and/or maintenance of multi-protein signaling complexes that reinforce and prolong HER2/EGFR or HER2/HER3 signaling by inhibiting HER2 ubiquitination and internalization.


Asunto(s)
Membrana Celular/metabolismo , Receptor ErbB-2/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Calcio/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Receptores ErbB/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Lapatinib , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Quinazolinas/farmacología , ARN Interferente Pequeño , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Receptor ErbB-3/metabolismo , Ubiquitinación
16.
Cancer Res ; 76(18): 5348-60, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27450451

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/patología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Receptores Sensibles al Calcio/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Ratones , Ratones Noqueados , Análisis de Matrices Tisulares
17.
J Biol Chem ; 284(33): 22444-22456, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19531472

RESUMEN

Butyrophilin 1A1 (BTN1A1) and xanthine oxidoreductase (XOR) are highly expressed in the lactating mammary gland and are secreted into milk associated with the milk fat globule membrane (MFGM). Ablation of the genes encoding either protein causes severe defects in the secretion of milk lipid droplets, suggesting that the two proteins may function in the same pathway. Therefore, we determined whether BTN1A1 and XOR directly interact using protein binding assays, surface plasmon resonance analysis, and gel filtration. Bovine XOR bound with high affinity in a pH- and salt-sensitive manner (KD=101+/-31 nM in 10 mM HEPES, 150 mM NaCl, pH 7.4) to the PRY/SPRY/B30.2 domain in the cytoplasmic region of bovine BTN1A1. Binding was stoichiometric, with one XOR dimer binding to either two BTN1A1 monomers or one dimer. XOR bound to BTN1A1 orthologs from mice, humans, or cows but not to the cytoplasmic domains of the closely related human paralogs, BTN2A1 or BTN3A1, or to the B30.2 domain of human RoRet (TRIM 38), a protein in the TRIM family. Analysis of the protein composition of the MFGM of wild type and BTN1A1 null mice showed that most of the XOR in mice lacking BTN1A1 was released from the MFGM in a soluble form when the milk lipid droplets were disrupted to prepare membrane, compared with wild-type mice, in which most of the XOR remained membrane-bound. Thus BTN1A1 functions in vivo to stabilize the association of XOR with the MFGM by direct interactions through the PRY/SPRY/B30.2 domain. The potential significance of BTN1A1/XOR interactions in the mammary gland and other tissues is discussed.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Glicoproteínas de Membrana/química , Xantina Deshidrogenasa/química , Animales , Butirofilinas , Bovinos , Citoplasma/metabolismo , Dimerización , Femenino , Humanos , Concentración de Iones de Hidrógeno , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , Leche , Unión Proteica , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda