Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Clin Invest ; 134(9)2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690739
3.
STAR Protoc ; 2(3): 100627, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34258591

RESUMEN

While isolating immune cells from spleens and lungs is routinely achieved using flow cytometry, it is challenging to isolate viable immune cells from skin. Here, we describe a step-by-step protocol for skin digestion using a murine melanoma model, which is amenable for detection of low abundant immune cell populations including group 2 innate lymphoid cells.


Asunto(s)
Inmunofenotipificación , Melanoma Experimental/inmunología , Neoplasias Cutáneas/inmunología , Piel/inmunología , Animales , Citometría de Flujo/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
4.
Cancer Res ; 81(14): 3876-3889, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33975880

RESUMEN

Breast cancer diagnosed within 10 years following childbirth is defined as postpartum breast cancer (PPBC) and is highly metastatic. Interactions between immune cells and other stromal cells within the involuting mammary gland are fundamental in facilitating an aggressive tumor phenotype. The MNK1/2-eIF4E axis promotes translation of prometastatic mRNAs in tumor cells, but its role in modulating the function of nontumor cells in the PPBC microenvironment has not been explored. Here, we used a combination of in vivo PPBC models and in vitro assays to study the effects of inactivation of the MNK1/2-eIF4E axis on the protumor function of select cells of the tumor microenvironment. PPBC mice deficient for phospho-eIF4E (eIF4ES209A) were protected against lung metastasis and exhibited differences in the tumor and lung immune microenvironment compared with wild-type mice. Moreover, the expression of fibroblast-derived IL33, an alarmin known to induce invasion, was repressed upon MNK1/2-eIF4E axis inhibition. Imaging mass cytometry on PPBC and non-PPBC patient samples indicated that human PPBC contains phospho-eIF4E high-expressing tumor cells and CD8+ T cells displaying markers of an activated dysfunctional phenotype. Finally, inhibition of MNK1/2 combined with anti-PD-1 therapy blocked lung metastasis of PPBC. These findings implicate the involvement of the MNK1/2-eIF4E axis during PPBC metastasis and suggest a promising immunomodulatory route to enhance the efficacy of immunotherapy by blocking phospho-eIF4E. SIGNIFICANCE: This study investigates the MNK1/2-eIF4E signaling axis in tumor and stromal cells in metastatic breast cancer and reveals that MNK1/2 inhibition suppresses metastasis and sensitizes tumors to anti-PD-1 immunotherapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Factor 4E Eucariótico de Iniciación/uso terapéutico , Terapia de Inmunosupresión/métodos , Animales , Modelos Animales de Enfermedad , Factor 4E Eucariótico de Iniciación/farmacología , Femenino , Humanos , Ratones , Metástasis de la Neoplasia , Periodo Posparto
5.
J Clin Invest ; 131(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33690225

RESUMEN

Melanomas commonly undergo a phenotype switch, from a proliferative to an invasive state. Such tumor cell plasticity contributes to immunotherapy resistance; however, the mechanisms are not completely understood and thus are therapeutically unexploited. Using melanoma mouse models, we demonstrated that blocking the MNK1/2-eIF4E axis inhibited melanoma phenotype switching and sensitized melanoma to anti-PD-1 immunotherapy. We showed that phospho-eIF4E-deficient murine melanomas expressed high levels of melanocytic antigens, with similar results verified in patient melanomas. Mechanistically, we identified phospho-eIF4E-mediated translational control of NGFR, a critical effector of phenotype switching. Genetic ablation of phospho-eIF4E reprogrammed the immunosuppressive microenvironment, exemplified by lowered production of inflammatory factors, decreased PD-L1 expression on dendritic cells and myeloid-derived suppressor cells, and increased CD8+ T cell infiltrates. Finally, dual blockade of the MNK1/2-eIF4E axis and the PD-1/PD-L1 immune checkpoint demonstrated efficacy in multiple melanoma models regardless of their genomic classification. An increase in the presence of intratumoral stem-like TCF1+PD-1+CD8+ T cells, a characteristic essential for durable antitumor immunity, was detected in mice given a MNK1/2 inhibitor and anti-PD-1 therapy. Using MNK1/2 inhibitors to repress phospho-eIF4E thus offers a strategy to inhibit melanoma plasticity and improve response to anti-PD-1 immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Factor 4E Eucariótico de Iniciación/inmunología , Inmunidad Celular , Sistema de Señalización de MAP Quinasas/inmunología , Melanoma Experimental/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Línea Celular Tumoral , Factor 4E Eucariótico de Iniciación/genética , Inmunoterapia , Sistema de Señalización de MAP Quinasas/genética , Melanoma Experimental/genética , Melanoma Experimental/terapia , Ratones , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Proteínas Serina-Treonina Quinasas/genética , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/inmunología
6.
Oncogene ; 39(18): 3650-3665, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32132651

RESUMEN

The BRAFV600E mutation occurs in more than 50% of cutaneous melanomas, and results in the constitutive activation of the mitogen-activated protein kinases (MAPK) pathway. MAP kinase-interacting serine/threonine-protein kinase 1 and 2 (MNK1/2) are downstream effectors of the activated MAPK pathway, and important molecular targets in invasive and metastatic cancer. Despite the well-known role of MNK1 in regulating mRNA translation, little is known concerning the impact of its aberrant activation on gene transcription. Here, we show that changes in the activity, or abundance, of MNK1 result in changes in the expression of pro-oncogenic and pro-invasive genes. Among the MNK1-upregulated genes, we identify Angiopoietin-like 4 (ANGPTL4), which in turn promotes an invasive phenotype via its ability to induce the expression of matrix metalloproteinases (MMPs). Using a pharmacologic inhibitor of MNK1/2, SEL201, we demonstrate that BRAFV600E-mutated cutaneous melanoma cells are reliant on MNK1/2 for invasion and lung metastasis.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares/genética , Melanoma/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Melanoma/genética , Melanoma/patología , Ratones , Mutación/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Transcriptoma/genética , Melanoma Cutáneo Maligno
7.
Toxicol Sci ; 170(1): 45-56, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30912803

RESUMEN

Tungsten is an emerging environmental toxicant associated with several pediatric leukemia clusters, although a causal association has not been established. Our previous work demonstrated that tungsten exposure resulted in an accumulation of pre-B cells in the bone marrow, the same cell type that accumulates in pediatric acute lymphoblastic leukemia (ALL). To better understand the relevant molecular mechanisms, we performed RNA-sequencing on flow sorted pre-B cells from control and tungsten-exposed mice. Tungsten decreased the expression of multiple genes critical for B cell development, including members of the interleukin-7 receptor (IL-7R) and pre-B cell receptor signaling pathways, such as Jak1, Stat5a, Pax5, Syk, and Ikzf3. These results were confirmed in an in vitro model of B cell differentiation, where tungsten arrested differentiation at the pro-B cell stage and inhibited proliferation. These changes were associated with decreased expression of multiple genes in the IL-7R signaling pathway and decreased percentage of IL-7R, phosphorylated STAT5 double-positive cells. Supplementation with IL-7 or overexpression of Pax5, the transcription factor downstream of IL-7R, rescued the tungsten-induced differentiation block. Together, these data support the hypothesis that IL-7R/Pax5 signaling axis is critical to tungsten-mediated effects on pre-B cell development. Importantly, many of these molecules are modulated in ALL.


Asunto(s)
Linfocitos B/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor de Transcripción PAX5/metabolismo , Receptores de Interleucina-7/metabolismo , Compuestos de Tungsteno/toxicidad , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Regulación hacia Abajo , Expresión Génica/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Factor de Transcripción PAX5/genética , Receptores de Interleucina-7/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Cancer Res ; 79(7): 1646-1657, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30659022

RESUMEN

The mechanisms by which breast cancers progress from relatively indolent ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) are not well understood. However, this process is critical to the acquisition of metastatic potential. MAPK-interacting serine/threonine-protein kinase 1 (MNK1) signaling can promote cell invasion. NODAL, a morphogen essential for embryogenic patterning, is often reexpressed in breast cancer. Here we describe a MNK1/NODAL signaling axis that promotes DCIS progression to IDC. We generated MNK1 knockout (KO) or constitutively active MNK1 (caMNK1)-expressing human MCF-10A-derived DCIS cell lines, which were orthotopically injected into the mammary glands of mice. Loss of MNK1 repressed NODAL expression, inhibited DCIS to IDC conversion, and decreased tumor relapse and metastasis. Conversely, caMNK1 induced NODAL expression and promoted IDC. The MNK1/NODAL axis promoted cancer stem cell properties and invasion in vitro. The MNK1/2 inhibitor SEL201 blocked DCIS progression to invasive disease in vivo. In clinical samples, IDC and DCIS with microinvasion expressed higher levels of phospho-MNK1 and NODAL versus low-grade (invasion-free) DCIS. Cumulatively, our data support further development of MNK1 inhibitors as therapeutics for preventing invasive disease. SIGNIFICANCE: These findings provide new mechanistic insight into progression of ductal carcinoma and support clinical application of MNK1 inhibitors to delay progression of indolent ductal carcinoma in situ to invasive ductal carcinoma.


Asunto(s)
Carcinoma de Mama in situ/patología , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Nodal/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Carcinoma de Mama in situ/metabolismo , Neoplasias de la Mama/metabolismo , Sistemas CRISPR-Cas , Carcinoma Ductal de Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/genética
10.
Toxicol Sci ; 150(2): 333-46, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26865663

RESUMEN

Tungsten is a naturally occurring metal that increasingly is being incorporated into industrial goods and medical devices, and is recognized as an emerging contaminant. Tungsten preferentially and rapidly accumulates in murine bone in a concentration-dependent manner; however the effect of tungsten deposition on bone biology is unknown. Other metals alter bone homeostasis by targeting bone marrow-derived mesenchymal stromal cell (MSC) differentiation, thus, we investigated the effects of tungsten on MSCsin vitroandin vivoIn vitro, tungsten shifted the balance of MSC differentiation by enhancing rosiglitazone-induced adipogenesis, which correlated with an increase in adipocyte content in the bone of tungsten-exposed, young, male mice. Conversely, tungsten inhibited osteogenesis of MSCsin vitro; however, we found no evidence that tungsten inhibited osteogenesisin vivo Interestingly, two factors known to influence adipogenesis are sex and age of mice. Both female and older mice have enhanced adipogenesis. We extended our study and exposed young female and adult (9-month) male and female mice to tungsten for 4 weeks. Although tungsten accumulated to a similar extent in young female mice, it did not promote adipogenesis. Interestingly, tungsten did not accumulate in the bone of older mice; it was undetectable in adult male mice, and just above the limit of detect in adult female mice. Surprisingly, tungsten enhanced adipogenesis in adult female mice. In summary, we found that tungsten alters bone homeostasis by altering differentiation of MSCs, which could have significant implications for bone quality, but is highly dependent upon sex and age.


Asunto(s)
Adipogénesis/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Fémur/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Caracteres Sexuales , Tungsteno/toxicidad , Envejecimiento/metabolismo , Animales , Médula Ósea/metabolismo , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/farmacocinética , Femenino , Fémur/metabolismo , Fémur/patología , Técnicas In Vitro , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Tungsteno/farmacocinética
11.
Toxicol Sci ; 143(1): 165-77, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25324207

RESUMEN

The number of individuals exposed to high levels of tungsten is increasing, yet there is limited knowledge of the potential human health risks. Recently, a cohort of breast cancer patients was left with tungsten in their breasts following testing of a tungsten-based shield during intraoperative radiotherapy. While monitoring tungsten levels in the blood and urine of these patients, we utilized the 66Cl4 cell model, in vitro and in mice to study the effects of tungsten exposure on mammary tumor growth and metastasis. We still detect tungsten in the urine of patients' years after surgery (mean urinary tungsten concentration at least 20 months post-surgery = 1.76 ng/ml), even in those who have opted for mastectomy, indicating that tungsten does not remain in the breast. In addition, standard chelation therapy was ineffective at mobilizing tungsten. In the mouse model, tungsten slightly delayed primary tumor growth, but significantly enhanced lung metastasis. In vitro, tungsten did not enhance 66Cl4 proliferation or invasion, suggesting that tungsten was not directly acting on 66Cl4 primary tumor cells to enhance invasion. In contrast, tungsten changed the tumor microenvironment, enhancing parameters known to be important for cell invasion and metastasis including activated fibroblasts, matrix metalloproteinases, and myeloid-derived suppressor cells. We show, for the first time, that tungsten enhances metastasis in an animal model of breast cancer by targeting the microenvironment. Importantly, all these tumor microenvironmental changes are associated with a poor prognosis in humans.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Pulmonares/secundario , Microambiente Tumoral , Compuestos de Tungsteno/toxicidad , Animales , Biopsia , Carga Corporal (Radioterapia) , Neoplasias de la Mama/sangre , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/orina , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quelantes/uso terapéutico , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/orina , Mamografía , Ratones Endogámicos BALB C , Invasividad Neoplásica , Medición de Riesgo , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Compuestos de Tungsteno/sangre , Compuestos de Tungsteno/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA