Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Carcinog ; 61(3): 359-371, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34856027

RESUMEN

The tumor microenvironment (TME) is an important modulator of response and resistance to endocrine therapy in estrogen receptor alpha (ER) positive breast cancer. Endocrine therapy is highly effective at reducing tumor burden and preventing recurrence in most estrogen receptor alpha (ER) positive breast cancers. Existing drugs work either directly by targeting tumor-cell ER or indirectly by inhibiting estrogen production in stromal cells with aromatase inhibitors (AI). However, many stromal cells also express ER and the direct impact of endocrine therapies on ER + stromal cells remain unclear. In this study, we investigated how neoadjuvant endocrine therapy (NET) directly effects stromal cells by measuring changes in stomal components of the TME that favor tumor progression. We previously defined two major subsets of tumor-associated stromal cells (TASCs): CD146 positive/CDCP1 negative (TASCCD146 ), CD146 negative/CDCP1 positive (TASCCDCP1 ), and generated a differentially expressed genes list associated with each type. Here, we applied the TASC gene list for classification and an algorithm that estimates immune cell abundance (TIMEx) to METABRIC transcriptomic data for ER + breast cancer patients coupled with multiplex imaging and analysis of paired tissue samples pre- and post- NET with the AI exemestane. TASCCDCP1 composition predicted for decreased patient survival in the METABRIC cohort. Exemestane treatment significantly increased expression of TASCCDCP1 and decreased expression of TASCCD146 . The posttreatment shift toward TASCCDCP1 composition correlated with increased macrophage infiltration and increased CD8+ T-cell, B cell, and general stromal components. The effectiveness of NET is currently based solely on the reduction of ER+ breast cancer cells. Here, we show NET displays clear TME effects that promote the expansion of the less favorable TASCCDCP1 population which are correlated with TME remodeling and reshaping immune infiltration supportive of tumor progression. Our findings highlight the need to further understand the role of endocrine therapy on TME remodeling, tumor progression, and patient outcomes.


Asunto(s)
Neoplasias de la Mama , Receptores de Estrógenos , Antígenos de Neoplasias , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Antígeno CD146 , Moléculas de Adhesión Celular , Receptor alfa de Estrógeno , Femenino , Humanos , Terapia Neoadyuvante , Receptores de Estrógenos/metabolismo , Microambiente Tumoral
2.
Curr Osteoporos Rep ; 19(6): 580-591, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34729693

RESUMEN

PURPOSE OF REVIEW: In this review, we provide an overview of what is currently known about the impacts of mechanical stimuli on metastatic tumor-induced bone disease (TIBD). Further, we focus on the role of the osteocyte, the skeleton's primary mechanosensory cell, which is central to the skeleton's mechanoresponse, sensing and integrating local mechanical stimuli, and then controlling the downstream remodeling balance as appropriate. RECENT FINDINGS: Exercise and controlled mechanical loading have anabolic effects on bone tissue in models of bone metastasis. They also have anti-tumorigenic properties, in part due to offsetting the vicious cycle of osteolytic bone loss as well as regulating inflammatory signals. The impacts of metastatic cancer on the mechanosensory function of osteocytes remains unclear. Increased mechanical stimuli are a potential method for mitigating TIBD.


Asunto(s)
Biofisica , Neoplasias Óseas/fisiopatología , Neoplasias Óseas/secundario , Remodelación Ósea , Osteocitos/fisiología , Animales , Fenómenos Biomecánicos , Humanos , Ratones , Estrés Mecánico
3.
Mol Carcinog ; 59(7): 822-829, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32233011

RESUMEN

Prostate cancer (PCa) is the most frequently diagnosed cancer for men in the U.S. but does not impede patient survival until the disease is metastatic. Metastatic lesions most frequently occur in the bone, which exhibits a distinct microenvironment of immune and bone cell populations. Advances in the diagnosis and treatment of primary PCa allow for the use of tailored therapeutic approaches based on biomarkers, protein expression, and histopathology. Understanding the molecular and cellular characteristics of primary tumors has advanced therapeutic development and survival for patients with PCa. Personalized medicine has only recently emerged for the treatment of metastatic bone lesions. Tumor induced bone disease (TIBD) in patients with PCa can be classified into lytic, blastic, or mixed pathologies, with most patients exhibiting the blastic phenotype. Progress has been made in treating TIBD, but metastatic PCa has yet to be cured. Immune checkpoint inhibitors have exhibited limited responses in immunosuppressive PCa tumors, but have yet to be assessed in metastatic sites which may be susceptible to an increased inflammatory response. Recent discoveries have uncovered distinct tumor microenvironments (TMEs) of blastic and lytic bone metastases from patients with PCa, identifying actionable targets for therapeutic applications, including immune checkpoint inhibitors and targeted therapeutics. Enrichment for macrophages and T cells in patient samples suggests metastatic sites may be reappraised as immunologically targetable, despite their immunologically "cold" primary tumors. The practice of performing bone biopsies will help identify unique cellular and protein targets in the bone TME that can guide therapy decisions.


Asunto(s)
Neoplasias Óseas/inmunología , Neoplasias de la Próstata/inmunología , Microambiente Tumoral/inmunología , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Humanos , Masculino , Neoplasias de la Próstata/patología , Linfocitos T/inmunología
4.
FASEB J ; 33(2): 1644-1657, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30161001

RESUMEN

Despite the success of approved systemic therapies for estrogen receptor α (ER)-positive breast cancer, drug resistance remains common. We hypothesized that secreted factors from the human tumor microenvironment could modulate drug resistance. We previously screened a library of 297 recombinant-secreted microenvironmental proteins for the ability to confer resistance to the anti-estrogen fulvestrant in 2 ER+ breast cancer cell lines. Herein, we considered whether factors that enhanced drug sensitivity could be repurposed as therapeutics and provide leads for drug development. Screening data revealed bone morphogenic protein (BMP)4 as a factor that inhibited cell growth and synergized with approved anti-estrogens and cyclin-dependent kinase 4/6 inhibitors (CDK4/6i). BMP4-mediated growth inhibition was dependent on type I receptor activin receptor-like kinase (ALK)3-dependent phosphorylation (P) of mothers against decapentaplegic homolog (SMAD/P-SMAD)1 and 5, which could be reversed by BMP receptor inhibitors and ALK3 knockdown. The primary effect of BMP4 on cell fate was cell-cycle arrest, in which RNA sequencing, immunoblot analysis, and RNA interference revealed to be dependent on p21WAF1/Cip1 upregulation. BMP4 also enhanced sensitivity to approved inhibitors of mammalian target of rapamycin complex 1 and CDK4/6 via ALK3-mediated P-SMAD1/5 and p21 upregulation in anti-estrogen-resistant cells. Patients bearing primary ER+ breast tumors, exhibiting a transcriptomic signature of BMP4 signaling, had improved disease outcome following adjuvant treatment with anti-estrogen therapy, independently of age, tumor grade, and tumor stage. Furthermore, a transcriptomic signature of BMP4 signaling was predictive of an improved biologic response to the CDK4/6i palbociclib, in combination with an aromatase inhibitor in primary tumors. These findings highlight BMP4 and its downstream pathway activation as a therapeutic opportunity in ER+ breast cancer.-Shee, K., Jiang, A., Varn, F. S., Liu, S., Traphagen, N. A., Owens, P., Ma, C. X., Hoog, J., Cheng, C., Golub, T. R., Straussman, R., Miller, T. W. Cytokine sensitivity screening highlights BMP4 pathway signaling as a therapeutic opportunity in ER+ breast cancer.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Neoplasias de la Mama/metabolismo , Citocinas/metabolismo , Transducción de Señal , Antagonistas de Andrógenos/uso terapéutico , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Receptores de Estrógenos/metabolismo , Análisis de Supervivencia , Transcriptoma , Microambiente Tumoral
5.
Breast Cancer Res ; 19(1): 105, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28886748

RESUMEN

BACKGROUND: During pregnancy, as the mammary gland prepares for synthesis and delivery of milk to newborns, a luminal mammary epithelial cell (MEC) subpopulation proliferates rapidly in response to systemic hormonal cues that activate STAT5A. While the receptor tyrosine kinase ErbB4 is required for STAT5A activation in MECs during pregnancy, it is unclear how ErbB3, a heterodimeric partner of ErbB4 and activator of phosphatidyl inositol-3 kinase (PI3K) signaling, contributes to lactogenic expansion of the mammary gland. METHODS: We assessed mRNA expression levels by expression microarray of mouse mammary glands harvested throughout pregnancy and lactation. To study the role of ErbB3 in mammary gland lactogenesis, we used transgenic mice expressing WAP-driven Cre recombinase to generate a mouse model in which conditional ErbB3 ablation occurred specifically in alveolar mammary epithelial cells (aMECs). RESULTS: Profiling of RNA from mouse MECs isolated throughout pregnancy revealed robust Erbb3 induction during mid-to-late pregnancy, a time point when aMECs proliferate rapidly and undergo differentiation to support milk production. Litters nursed by ErbB3 KO dams weighed significantly less when compared to litters nursed by ErbB3 WT dams. Further analysis revealed substantially reduced epithelial content, decreased aMEC proliferation, and increased aMEC cell death during late pregnancy. Consistent with the potent ability of ErbB3 to activate cell survival through the PI3K/Akt pathway, we found impaired Akt phosphorylation in ErbB3 KO samples, as well as impaired expression of STAT5A, a master regulator of lactogenesis. Constitutively active Akt rescued cell survival in ErbB3-depleted aMECs, but failed to restore STAT5A expression or activity. Interestingly, defects in growth and survival of ErbB3 KO aMECs as well as Akt phosphorylation, STAT5A activity, and expression of milk-encoding genes observed in ErbB3 KO MECs progressively improved between late pregnancy and lactation day 5. We found a compensatory upregulation of ErbB4 activity in ErbB3 KO mammary glands. Enforced ErbB4 expression alleviated the consequences of ErbB3 ablation in aMECs, while combined ablation of both ErbB3 and ErbB4 exaggerated the phenotype. CONCLUSIONS: These studies demonstrate that ErbB3, like ErbB4, enhances lactogenic expansion and differentiation of the mammary gland during pregnancy, through activation of Akt and STAT5A, two targets crucial for lactation.


Asunto(s)
Mama/citología , Mama/metabolismo , Diferenciación Celular/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Lactancia/genética , Receptor ErbB-3/genética , Alelos , Animales , Proliferación Celular/genética , Supervivencia Celular/genética , Femenino , Técnicas de Inactivación de Genes , Inmunohistoquímica , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor ErbB-3/metabolismo , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal
6.
BMC Cancer ; 17(1): 88, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28143493

RESUMEN

BACKGROUND: The role of the chemokine CCL2 in breast cancer is controversial. While CCL2 recruits and activates pro-tumor macrophages, it is also reported to enhance neutrophil-mediated anti-tumor activity. Moreover, loss of CCL2 in early development enhances breast cancer progression. METHODS: To clarify these conflicting findings, we examined the ability of CCL2 to alter naïve and tumor entrained neutrophil production of ROS, release of granzyme-B, and killing of tumor cells in multiple mouse models of breast cancer. CCL2 was delivered intranasally in mice to elevate CCL2 levels in the lung and effects on seeding and growth of breast tumor cells were evaluated. The TCGA data base was queried for relationship between CCL2 expression and relapse free survival of breast cancer patients and compared to subsets of breast cancer patients. RESULTS: Even though each of the tumor cell lines studied produced approximately equal amounts of CCL2, exogenous delivery of CCL2 to co-cultures of breast tumor cells and neutrophils enhanced the ability of tumor-entrained neutrophils (TEN) to kill the less aggressive 67NR variant of 4T1 breast cancer cells. However, exogenous CCL2 did not enhance naïve or TEN neutrophil killing of more aggressive 4T1 or PyMT breast tumor cells. Moreover, this anti-tumor activity was not observed in vivo. Intranasal delivery of CCL2 to BALB/c mice markedly enhanced seeding and outgrowth of 67NR cells in the lung and increased the recruitment of CD4+ T cells and CD8+ central memory T cells into lungs of tumor bearing mice. There was no significant increase in the recruitment of CD19+ B cells, or F4/80+, Ly6G+ and CD11c + myeloid cells. CCL2 had an equal effect on CD206+ and MHCII+ populations of macrophages, thus balancing the pro- and anti-tumor macrophage cell population. Analysis of the relationship between CCL2 levels and relapse free survival in humans revealed that overall survival is not significantly different between high CCL2 expressing and low CCL2 expressing breast cancer patients grouped together. However, examination of the relationship between high CCL2 expressing basal-like, HER2+ and luminal B breast cancer patients revealed that higher CCL2 expressing tumors in these subgroups have a significantly higher probability of surviving longer than those expressing low CCL2. CONCLUSIONS: While our in vitro data support a potential anti-tumor role for CCL2 in TEN neutrophil- mediated tumor killing in poorly aggressive tumors, intranasal delivery of CCL2 increased CD4+ T cell recruitment to the pre-metastatic niche of the lung and this correlated with enhanced seeding and growth of tumor cells. These data indicate that effects of CCL2/CCR2 antagonists on the intratumoral leukocyte content should be monitored in ongoing clinical trials using these agents.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quimiocina CCL2/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neutrófilos/metabolismo , Neutrófilos/patología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Humanos , Leucocitos/metabolismo , Leucocitos/patología , Pulmón/metabolismo , Pulmón/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
7.
Biochem Soc Trans ; 44(4): 1117-34, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27528760

RESUMEN

Bone morphogenetic proteins (BMPs) belong to the transforming growth factor-ß (TGF-ß) family signalling pathway. Similar to TGF-ß, the complex roles of BMPs in development and disease are demonstrated by their dichotomous roles in various cancers and cancer stages. Although early studies implicated BMP signalling in tumour suppressive phenotypes, the results of more recent experiments recognize BMPs as potent tumour promoters. Many of these complexities are becoming illuminated by understanding the role of BMPs in their contextual role in unique cell types of cancer and the impact of their surrounding tumour microenvironment. Here we review the emerging roles of BMP signalling in cancer, with a focus on the molecular underpinnings of BMP signalling in individual cancers as a valid therapeutic target for cancer prevention and treatment.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Microambiente Tumoral , Animales , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/genética , Progresión de la Enfermedad , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/terapia
8.
J Immunol ; 193(6): 3155-64, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25127858

RESUMEN

There is growing evidence that generation of adenosine from ATP, which is mediated by the CD39/CD73 enzyme pair, predetermines immunosuppressive and proangiogenic properties of myeloid cells. We have previously shown that the deletion of the TGF-ß type II receptor gene (Tgfbr2) expression in myeloid cells is associated with decreased tumor growth, suggesting protumorigenic effect of TGF-ß signaling. In this study, we tested the hypothesis that TGF-ß drives differentiation of myeloid-derived suppressor cells into protumorigenic terminally differentiated myeloid mononuclear cells (TDMMCs) characterized by high levels of cell-surface CD39/CD73 expression. We found that TDMMCs represent a major cell subpopulation expressing high levels of both CD39 and CD73 in the tumor microenvironment. In tumors isolated from mice with spontaneous tumor formation of mammary gland and conditional deletion of the type II TGF-ß receptor in mammary epithelium, an increased level of TGF-ß protein was associated with further increase in number of CD39(+)CD73(+) TDMMCs compared with MMTV-PyMT/TGFßRII(WT) control tumors with intact TGF-ß signaling. Using genetic and pharmacological approaches, we demonstrated that the TGF-ß signaling mediates maturation of myeloid-derived suppressor cells into TDMMCs with high levels of cell surface CD39/CD73 expression and adenosine-generating capacity. Disruption of TGF-ß signaling in myeloid cells resulted in decreased accumulation of TDMMCs, expressing CD39 and CD73, and was accompanied by increased infiltration of T lymphocytes, reduced density of blood vessels, and diminished progression of both Lewis lung carcinoma and spontaneous mammary carcinomas. We propose that TGF-ß signaling can directly induce the generation of CD39(+)CD73(+) TDMMCs, thus contributing to the immunosuppressive, proangiogenic, and tumor-promoting effects of this pleiotropic effector in the tumor microenvironment.


Asunto(s)
5'-Nucleotidasa/biosíntesis , Antígenos CD/biosíntesis , Apirasa/biosíntesis , Células Mieloides/inmunología , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/inmunología , Animales , Células de la Médula Ósea/inmunología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patología , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular/inmunología , Femenino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/inmunología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/biosíntesis , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Transducción de Señal/inmunología , Linfocitos T/inmunología , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Curr Osteoporos Rep ; 14(4): 151-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27255469

RESUMEN

Bone metastatic disease remains a significant and frequent problem for cancer patients that can lead to increased morbidity and mortality. Unfortunately, despite decades of research, bone metastases remain incurable. Current studies have demonstrated that many properties and cell types within the bone and bone marrow microenvironment contribute to tumor-induced bone disease. Furthermore, they have pointed to the importance of understanding how tumor cells interact with their microenvironment in order to help improve both the development of new therapeutics and the prediction of response to therapy.


Asunto(s)
Células de la Médula Ósea/citología , Neoplasias Óseas/secundario , Huesos/citología , Microambiente Tumoral , Linfocitos B , Médula Ósea/irrigación sanguínea , Médula Ósea/metabolismo , Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/metabolismo , Huesos/irrigación sanguínea , Huesos/metabolismo , Microambiente Celular , Fibroblastos , Humanos , Células Asesinas Naturales , Macrófagos , Células Supresoras de Origen Mieloide , Osteoblastos , Osteoclastos , Osteocitos , Linfocitos T
10.
Breast Cancer Res Treat ; 149(1): 69-79, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25491778

RESUMEN

Activation of the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathway has been implicated in anti-estrogen resistance in breast cancer. We tested the therapeutic potential of the novel PI3K/mTOR dual inhibitor P7170 in a panel of anti-estrogen-sensitive and anti-estrogen-resistant models of ER+ breast cancer. Estrogen receptor-positive (ER+) breast cancer cells were treated ±P7170. Fresh cores from primary ER+/HER2- tumors from two patients were treated ±P7170 ex vivo. Mice bearing breast cancer xenografts were randomized to treatment with vehicle, fulvestrant, P7170, or combinations, and tumor volumes were measured. Tissues and cells were analyzed for markers of pathway activity, cell viability, and apoptosis. In cell lines, P7170 exhibited IC50 values in the range of 0.9-7 nM and induced apoptosis. P7170 potently inhibited mTOR activity (≤ 25 nM) and inhibited PI3K at higher concentrations (≥ 200 nM). P7170 completely inhibited MCF-7 tumor growth, significantly inhibited growth of fulvestrant-resistant T47D tumors, and suppressed tumor cell proliferation but did not induce apoptosis. While P7170 inhibits PI3K and mTOR in ER+/HER2- human breast cancer cells and tumors ex vivo, in vivo data indicate that the primary mechanism of P7170 anti-tumor action is inhibition of mTOR and cell proliferation. P7170 is a novel agent worthy of further investigation for the treatment of ER+ breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de las Glándulas Endocrinas/tratamiento farmacológico , Imidazoles/administración & dosificación , Fosfatidilinositol 3-Quinasas/genética , Quinolinas/administración & dosificación , Serina-Treonina Quinasas TOR/genética , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Neoplasias de las Glándulas Endocrinas/genética , Neoplasias de las Glándulas Endocrinas/patología , Estradiol/administración & dosificación , Estradiol/análogos & derivados , Femenino , Fulvestrant , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/administración & dosificación , Receptores de Estrógenos/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Proc Natl Acad Sci U S A ; 109(8): 2814-9, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-21576484

RESUMEN

Bone morphogenetic proteins (BMPs) are members of the TGF-ß superfamily of signaling molecules. BMPs can elicit a wide range of effects in many cell types and have previously been shown to induce growth inhibition in carcinoma cells as well as normal epithelia. Recently, it has been demonstrated that BMP4 and BMP7 are overexpressed in human breast cancers and may have tumor suppressive and promoting effects. We sought to determine whether disruption of the BMP receptor 2 (BMPR2) would alter mammary tumor progression in mice that express the Polyoma middle T antigen. Mice expressing Polyoma middle T antigen under the mouse mammary tumor virus promoter were combined with mice that have doxycycline-inducible expression of a dominant-negative (DN) BMPR2. We did not observe any differences in tumor latency. However, mice expressing the BMPR2-DN had a fivefold increase in lung metastases. We characterized several cell autonomous changes and found that BMPR2-DN-expressing tumor cells had higher rates of proliferation. We also identified unique changes in inflammatory cells and secreted chemokines/cytokines that accompanied BMPR2-DN-expressing tumors. By immunohistochemistry, it was found that BMPR2-DN primary tumors and metastases had an altered reactive stroma, indicating specific changes in the tumor microenvironment. Among the changes we discovered were increased myeloid derived suppressor cells and the chemokine CCL9. BMP was shown to directly regulate CCL9 expression. We conclude that BMPR2 has tumor-suppressive function in mammary epithelia and microenvironment and that disruption can accelerate mammary carcinoma metastases.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Comunicación Paracrina , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Movimiento Celular , Proliferación Celular , Quimiocinas/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Inflamación/patología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/irrigación sanguínea , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Células Mieloides/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Neovascularización Patológica , Transducción de Señal , Microambiente Tumoral
12.
J Environ Qual ; 44(5): 1605-16, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26436277

RESUMEN

The use of sediment color as a fingerprint property to determine sediment sources is an emerging technique that can provide a rapid and inexpensive means of investigating sediment sources. The present study aims to test the feasibility of color fingerprint properties to apportion sediment sources within the South Tobacco Creek Watershed (74 km) in Manitoba, Canada. Suspended sediment from 2009 to 2011 at six monitoring stations and potential source samples along the main stem of the creek were collected. Reflectance spectra of sediments and source materials were quantified using a diffuse reflectance spectrometry, and 16 color coefficients were derived from several color space models. Canonical discriminant analysis was used to reclassify and downsize sediment source groups. After the linear additive test and stepwise discriminant function analysis, four color coefficients were chosen to fit the Stable Isotope Analysis in R model. Consistent with the conventional fingerprinting approach, the color fingerprint results demonstrated a switch in the dominant sediment source between the headwaters and the outlet of the watershed, with the main sources being topsoil in the upper reaches, whereas outcrop shale and stream bank materials dominated in the lower reaches. The color fingerprinting approach can be integrated with conventional fingerprints (e.g., geochemical and fallout radionuclide properties) to improve source discrimination, which is a key component for source ascription modeling. We concluded that the use of color fingerprints is a promising, cost-effective technique for sediment source fingerprinting.

13.
Breast Cancer Res ; 16(5): 425, 2014 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-25280532

RESUMEN

INTRODUCTION: Transforming growth factor beta (TGFß) plays a major role in the regulation of tumor initiation, progression, and metastasis. It is depended on the type II TGFß receptor (TßRII) for signaling. Previously, we have shown that deletion of TßRII in mammary epithelial of MMTV-PyMT mice results in shortened tumor latency and increased lung metastases. However, active TGFß signaling increased the number of circulating tumor cells and metastases in MMTV-Neu mice. In the current study, we describe a newly discovered connection between attenuated TGFß signaling and human epidermal growth factor receptor 2 (HER2) signaling in mammary tumor progression. METHODS: All studies were performed on MMTV-Neu mice with and without dominant-negative TßRII (DNIIR) in mammary epithelium. Mammary tumors were analyzed by flow cytometry, immunohistochemistry, and immunofluorescence staining. The levels of secreted proteins were measured by enzyme-linked immunosorbent assay. Whole-lung mount staining was used to quantitate lung metastasis. The Cancer Genome Atlas (TCGA) datasets were used to determine the relevance of our findings to human breast cancer. RESULTS: Attenuated TGFß signaling led to a delay tumor onset, but increased the number of metastases in MMTVNeu/DNIIR mice. The DNIIR tumors were characterized by increased vasculogenesis, vessel leakage, and increased expression of vascular endothelial growth factor (VEGF). During DNIIR tumor progression, both the levels of CXCL1/5 and the number of CD11b+Gr1+ cells and T cells decreased. Analysis of TCGA datasets demonstrated a significant negative correlation between TGFBR2 and VEGF genes expression. Higher VEGFA expression correlated with shorter distant metastasis-free survival only in HER2+ patients with no differences in HER2-, estrogen receptor +/- or progesterone receptor +/- breast cancer patients. CONCLUSION: Our studies provide insights into a novel mechanism by which epithelial TGFß signaling modulates the tumor microenvironment, and by which it is involved in lung metastasis in HER2+ breast cancer patients. The effects of pharmacological targeting of the TGFß pathway in vivo during tumor progression remain controversial. The targeting of TGFß signaling should be a viable option, but because VEGF has a protumorigenic effect on HER2+ tumors, the targeting of this protein could be considered when it is associated with attenuated TGFß signaling.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Carcinogénesis/metabolismo , Quimiocinas/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/patología , Ratones Transgénicos , Neovascularización Patológica/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos T/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
JCI Insight ; 9(1)2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38193534

RESUMEN

Metastatic breast cancer (mBC) tissue in bone was systematically profiled to define the composition of the tumor microenvironment. Gene expression identified a high myeloid signature of patients with improved survival outcomes. Bone metastases were profiled by spatial proteomics to examine myeloid populations within the stroma that correlated with macrophage functions. Single-cell spatial analysis uncovered macrophage activation in the stroma of mBC bone lesions. Matched BC patient samples of primary breast tumor and bone metastasis tissues were compared for gene expression in the bone, where bone morphogenetic protein 2 (BMP2) was most significantly upregulated. Immune cell changes from breast to bone demonstrated a loss of lymphoid cells but a consistent population of macrophages. BMP-activated macrophages were increased uniquely in bone. Bone marrow-derived macrophage activation coupled with BMP inhibition increased inflammatory responses. Using experimental mouse models of mBC bone metastasis and trained immunity, we found that BMP inhibition restricts progression of metastases early in the macrophage activation state but not after tumors were established in the bone. This study revealed unique myeloid BMP activation states that are distinctly integrated with bone metastases.


Asunto(s)
Proteínas Morfogenéticas Óseas , Neoplasias Óseas , Neoplasias de la Mama , Macrófagos , Animales , Femenino , Humanos , Ratones , Huesos , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Fenotipo , Microambiente Tumoral , Proteínas Morfogenéticas Óseas/metabolismo
15.
J Environ Radioact ; 264: 107206, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37224721

RESUMEN

Cryoconite is a specific type of material found on the surface of glaciers and icesheets. Samples of cryoconite were collected from the Orwell Glacier and its moraines, together with suspended sediment from the proglacial stream on Signy Island, part of the South Orkney Islands, Antarctica. The activity concentrations of certain fallout radionuclides were determined in the cryoconite, moraine and suspended sediment, in addition to particle size composition and %C and %N. For cryoconite samples (n = 5), mean activity concentrations (±1SD) of 137Cs, 210Pbun and 241Am were 13.2 ± 20.9, 66.1 ± 94.0 and 0.32 ± 0.64 Bq kg-1, respectively. Equivalent values for the moraine samples (n = 7) were 2.56 ± 2.75, 14.78 ± 12.44 and <1.0 Bq kg-1, respectively. For the composite suspended sediment sample, collected over 3 weeks in the ablation season, the values (± counting uncertainty) for 137Cs, 210Pbun and 241Am were 2.64 ± 0.88, 49.2 ± 11.9 and <1.0 Bq kg-1, respectively. Thus, fallout radionuclide activity concentrations were elevated in cryoconite relative to moraine and suspended sediment. In the case of 40K, the highest value was for the suspended sediment (1423 ± 166 Bq kg-1). The fallout radionuclides in cryoconite were 1-2 orders of magnitude greater than values in soils collected from other locations in Antarctica. This work further demonstrates that cryoconite likely scavenges fallout radionuclides (dissolved and particulate) in glacial meltwater. In the case of 40K, the greater value in suspended sediment implies a subglacial source. These results are amongst the relatively few that demonstrate the presence of fallout radionuclides in cryoconites at remote locations in the Southern Hemisphere. This work adds to the growing contention that elevated activities of fallout radionuclides, and other contaminants, in cryoconites are a global phenomenon and may be a risk to downstream terrestrial and aquatic ecosystems.


Asunto(s)
Cubierta de Hielo , Monitoreo de Radiación , Ecosistema , Regiones Antárticas , Plomo , Radioisótopos de Cesio/análisis
16.
Cancer Rep (Hoboken) ; 6(2): e1707, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36054271

RESUMEN

BACKGROUND: Prostate cancer is a common cancer in men that annually results in more than 33 000 US deaths. Mortality from prostate cancer is largely from metastatic disease, reflecting on the great strides in the last century of treatments in care for the localized disease. Metastatic castrate resistant prostate cancer (mCRPC) will commonly travel to the bone, creating unique bone pathology that requires nuanced treatments in those sites with surgical, radio and chemotherapeutic interventions. The bone morphogenetic protein (BMP) pathway has been historically studied in the capacity to regulate the osteogenic nature of new bone. New mineralized bone generation is a frequent and common observation in mCRPC and referred to as blastic bone lesions. Less common are bone destructive lesions that are termed lytic. METHODS: We queried the cancer genome atlas (TCGA) prostate cancer databases for the expression of the BMP pathway and found that distinct gene expression of the ligands, soluble antagonists, receptors, and intracellular mediators were altered in localized versus metastatic disease. Human prostate cancer cell lines have an innate ability to promote blastic- or lytic-like bone lesions and we hypothesized that inhibiting BMP signaling in these cell lines would result in a distinct change in osteogenesis gene expression with BMP inhibition. RESULTS: We found unique and common changes by comparing these cell lines response and unique BMP pathway alterations. We treated human PCa cell lines with distinct bone pathologic phenotypes with the BMP inhibitor DMH1 and found distinct osteogenesis responses. We analyzed distinct sites of metastatic PCa in the TCGA and found that BMP signaling was selectively altered in commons sites such as lymph node, bone and liver compared to primary tumors. CONCLUSIONS: Overall we conclude that BMPs in metastatic prostate cancer are important signals and functional mediators of diverse processes that have potential for individualized precision oncology in mCRPC.


Asunto(s)
Osteogénesis , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Medicina de Precisión , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Línea Celular
17.
J Immunother Cancer ; 11(9)2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37734878

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC) is a devastating disease most often associated with tobacco consumption that induces a field of mutations from which a tumor arises. Identification of ways to prevent the emergence of cancer in high-risk patients is an ultimate goal for combatting all types of cancer, including OSCC. METHODS: Our study employs a mouse model of tongue carcinogenesis induced by tobacco carcinogen mimetic, 4-nitroquinoline 1-oxide (4NQO), to establish tongue dysplasia and OSCC. We use conventional histology, immunohistochemistry, multispectral imaging, mass cytometry, novel cell lines, pharmaceutical inhibition of PI3Kγ, T-cell suppression assays and mouse transplant models in our functional experimentation. RESULTS: In our study, we identify Ly6G+ granulocytes as the most abundant immune cell type in a model of tongue carcinogenesis induced by tobacco carcinogen mimetic 4NQO. Targeting Ly6G+ granulocytes with a pharmacologic inhibitor of PI3Kγ, an isoform of PI3K exclusively expressed by myeloid cells, resulted in reduced tongue dysplasia severity, and reduced rates of OSCC. Importantly, we performed functional assays with the Ly6G+ granulocytes induced in cell line models of 4NQO carcinogenesis to demonstrate that these granulocytes have increased polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) activity against T-cell proliferation and these PMN-MDSCs play a functional role in promoting tumor formation by inhibiting tumor regression in a PI3Kγ-dependent manner. CONCLUSIONS: Overall, our data suggest that recruitment of PMN-MDSCs to sites of dysplasia is critical to immune suppression of CD8 T cells, thereby permitting malignancy, and PI3Kγ inhibitors are one mechanism to reduce PMN-MDSC recruitment, immunosuppression and tumorigenesis in OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Células Supresoras de Origen Mieloide , Humanos , Animales , Ratones , Fosfatidilinositol 3-Quinasa , Carcinoma de Células Escamosas/inducido químicamente , Neoplasias de la Boca/inducido químicamente , Carcinogénesis , Carcinógenos/toxicidad , Carcinoma de Células Escamosas de Cabeza y Cuello , Fosfatidilinositoles
18.
Sci Total Environ ; 894: 164902, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37343877

RESUMEN

The accumulation of fallout radionuclides (FRNs) from nuclear weapons testing and nuclear accidents has been evaluated for over half a century in natural environments; however, until recently their distribution and abundance within glaciers have been poorly understood. Following a series of individual studies of FRNs, specifically 137Cs, 241Am and 210Pb, deposited on the surface of glaciers, we now understand that cryoconite, a material commonly found in the supraglacial environment, is a highly efficient accumulator of FRNs, both artificial and natural. However, the variability of FRN activity concentrations in cryoconite across the global cryosphere has never been assessed. This study thus aims to both synthesize current knowledge on FRNs in cryoconite and assess the controls on variability of activity concentrations. We present a global database of new and previously published data based on gamma spectrometry of cryoconite and proglacial sediments, and assess the extent to which a suite of environmental and physical factors can explain spatial variability in FRN activity concentrations in cryoconite. We show that FRNs are not only found in cryoconite on glaciers within close proximity to specific sources of radioactivity, but across the global cryosphere, and at activity concentrations up to three orders of magnitude higher than those found in soils and sediments in the surrounding environment. We also show that the organic content of cryoconite exerts a strong control on accumulation of FRNs, and that activity concentrations in cryoconite are some of the highest ever described in environmental matrices outside of nuclear exclusion zones, occasionally in excess of 10,000 Bq kg-1. These findings highlight a need for significant improvements in the understanding of the fate of legacy contaminants within glaciated catchments. Future interdisciplinary research is required on the mechanisms governing their accumulation, storage, and mobility, and their potential to create time-dependent impacts on downstream water quality and ecosystem sustainability.

19.
Pharmacol Ther ; 239: 108280, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36116682

RESUMEN

Breast cancer (BC) patient prognosis has improved over the past 2 decades with a 99% 5-year survival rate for localized BC, yet metastatic breast cancer (mBC) continues to cause high mortality with a 5-year survival rate of 29%. Approximately 70% of BC metastases occur in the bone, with estrogen receptor (ER) positive BC exhibiting a particular affinity to bone. Once BC metastasizes to the bone, curative treatments are not available, thus therapeutic approaches are focused on palliative care and prevention of skeletal related events while attempting to slow metastatic progression. Recent advances in molecularly targeted agents have enhanced the repertoire of options for mBC patients, but immunotherapies have not yet been fully translated to ER+ tumors. Thus mBC patients have yet to fully benefit from novel therapies, which is currently obstructing patient survival. The unique tumor microenvironment (TME) of mBC in bone offers an array of targets for therapeutic development. The mBC TME in bone presents a predominantly osteolytic or destructive bone pathology, where bone mineral loss is driven by increased resorption of bone by osteoclasts. We discuss therapeutics targeting the mBC cells, bone cells, immune cells, and other stromal cells in the bone TME, including treatments that are currently used in the clinic, under development, and are potential new avenues for therapy. Therapeutic advancements targeting the TME of mBC in bone could be applied to other bone-resident cancers, including myeloma and metastatic prostate cancer bone lesions. These precision oncology approaches to mBC treatment will improve the quality of life and clinical outcomes of patients with mBC bone lesions.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Mama , Masculino , Humanos , Microambiente Tumoral , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Calidad de Vida , Medicina de Precisión , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología
20.
Dev Cell ; 11(3): 301-12, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16950122

RESUMEN

To assess whether Smad signaling affects skin development, we generated transgenic mice in which a Smad antagonist, Smad7, was induced in keratinocytes, including epidermal stem cells. Smad7 transgene induction perturbed hair follicle morphogenesis and differentiation, but accelerated sebaceous gland morphogenesis. Further analysis revealed that independent of its role in anti-Smad signaling, Smad7 bound beta-catenin and induced beta-catenin degradation by recruiting an E3 ligase, Smurf2, to the Smad7/beta-catenin complex. Consequently, Wnt/beta-catenin signaling was suppressed in Smad7 transgenic hair follicles. Coexpression of the Smurf2 and Smad7 transgenes exacerbated Smad7-induced abnormalities in hair follicles and sebaceous glands. Conversely, when endogenous Smad7 was knocked down, keratinocytes exhibited increased beta-catenin protein and enhanced Wnt signaling. Our data reveal a mechanism for Smad7 in antagonizing Wnt/beta-catenin signaling, thereby shifting the skin differentiation program from forming hair follicles to sebaceous glands.


Asunto(s)
Epidermis/patología , Folículo Piloso/fisiología , Proteína smad7/fisiología , Células Madre/fisiología , beta Catenina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Regulación hacia Abajo , Epidermis/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinocitos/metabolismo , Ratones , Ratones Transgénicos , Morfogénesis , Glándulas Sebáceas/fisiología , Transducción de Señal , Proteína smad7/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA