Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 25(7): 1257-1269, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38806707

RESUMEN

The circadian clock is a critical regulator of immunity, and this circadian control of immune modulation has an essential function in host defense and tumor immunosurveillance. Here we use a single-cell RNA sequencing approach and a genetic model of colorectal cancer to identify clock-dependent changes to the immune landscape that control the abundance of immunosuppressive cells and consequent suppression of cytotoxic CD8+ T cells. Of these immunosuppressive cell types, PD-L1-expressing myeloid-derived suppressor cells (MDSCs) peak in abundance in a rhythmic manner. Disruption of the epithelial cell clock regulates the secretion of cytokines that promote heightened inflammation, recruitment of neutrophils and the subsequent development of MDSCs. We also show that time-of-day anti-PD-L1 delivery is most effective when synchronized with the abundance of immunosuppressive MDSCs. Collectively, these data indicate that circadian gating of tumor immunosuppression informs the timing and efficacy of immune checkpoint inhibitors.


Asunto(s)
Antígeno B7-H1 , Relojes Circadianos , Inhibidores de Puntos de Control Inmunológico , Células Supresoras de Origen Mieloide , Animales , Ratones , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Relojes Circadianos/inmunología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Ratones Endogámicos C57BL , Ritmo Circadiano/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales/tratamiento farmacológico , Microambiente Tumoral/inmunología , Tolerancia Inmunológica , Humanos , Femenino , Línea Celular Tumoral , Análisis de la Célula Individual , Terapia de Inmunosupresión , Citocinas/metabolismo , Masculino
2.
Nat Genet ; 55(4): 595-606, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36914836

RESUMEN

Women with germline BRCA1 mutations (BRCA1+/mut) have increased risk for hereditary breast cancer. Cancer initiation in BRCA1+/mut is associated with premalignant changes in breast epithelium; however, the role of the epithelium-associated stromal niche during BRCA1-driven tumor initiation remains unclear. Here we show that the premalignant stromal niche promotes epithelial proliferation and mutant BRCA1-driven tumorigenesis in trans. Using single-cell RNA sequencing analysis of human preneoplastic BRCA1+/mut and noncarrier breast tissues, we show distinct changes in epithelial homeostasis including increased proliferation and expansion of basal-luminal intermediate progenitor cells. Additionally, BRCA1+/mut stromal cells show increased expression of pro-proliferative paracrine signals. In particular, we identify pre-cancer-associated fibroblasts (pre-CAFs) that produce protumorigenic factors including matrix metalloproteinase 3 (MMP3), which promotes BRCA1-driven tumorigenesis in vivo. Together, our findings demonstrate that precancerous stroma in BRCA1+/mut may elevate breast cancer risk through the promotion of epithelial proliferation and an accumulation of luminal progenitor cells with altered differentiation.


Asunto(s)
Neoplasias de la Mama , Glándulas Mamarias Humanas , Femenino , Humanos , Mutación , Proteína BRCA1/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/metabolismo , Glándulas Mamarias Humanas/metabolismo , Carcinogénesis/patología , Células del Estroma/patología
3.
Commun Biol ; 4(1): 1268, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34741115

RESUMEN

Metastasis is a fatal disease where research progress has been hindered by a lack of authentic experimental models. Here, we develop a 3D tumor sphere culture-transplant system that facilitates the growth and engineering of patient-derived xenograft (PDX) tumor cells for functional metastasis assays in vivo. Orthotopic transplantation and RNA sequencing (RNA-seq) analyses show that PDX tumor spheres maintain tumorigenic potential, and the molecular marker and global transcriptome signatures of native tumor cells. Tumor spheres display robust capacity for lentiviral engineering and dissemination in spontaneous and experimental metastasis assays in vivo. Inhibition of pathways previously reported to attenuate metastasis also inhibit metastasis after sphere culture, validating our approach for authentic investigations of metastasis. Finally, we demonstrate a new role for the metabolic enzyme NME1 in promoting breast cancer metastasis, providing proof-of-principle that our culture-transplant system can be used for authentic propagation and engineering of patient tumor cells for functional studies of metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Xenoinjertos , Metástasis de la Neoplasia , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Neoplasias Experimentales , Microambiente Tumoral
4.
Commun Biol ; 4(1): 685, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083739

RESUMEN

Foreign body response (FBR) to biomaterials compromises the function of implants and leads to medical complications. Here, we report a hybrid alginate microcapsule (AlgXO) that attenuated the immune response after implantation, through releasing exosomes derived from human Umbilical Cord Mesenchymal Stem Cells (XOs). Upon release, XOs suppress the local immune microenvironment, where xenotransplantation of rat islets encapsulated in AlgXO led to >170 days euglycemia in immunocompetent mouse model of Type 1 Diabetes. In vitro analyses revealed that XOs suppressed the proliferation of CD3/CD28 activated splenocytes and CD3+ T cells. Comparing suppressive potency of XOs in purified CD3+ T cells versus splenocytes, we found XOs more profoundly suppressed T cells in the splenocytes co-culture, where a heterogenous cell population is present. XOs also suppressed CD3/CD28 activated human peripheral blood mononuclear cells (PBMCs) and reduced their cytokine secretion including IL-2, IL-6, IL-12p70, IL-22, and TNFα. We further demonstrate that XOs mechanism of action is likely mediated via myeloid cells and XOs suppress both murine and human macrophages partly by interfering with NFκB pathway. We propose that through controlled release of XOs, AlgXO provide a promising new platform that could alleviate the local immune response to implantable biomaterials.


Asunto(s)
Diabetes Mellitus Experimental/cirugía , Diabetes Mellitus Tipo 1/cirugía , Exosomas/inmunología , Inmunidad/inmunología , Factores Inmunológicos/inmunología , Trasplante de Islotes Pancreáticos/métodos , Animales , Células Cultivadas , Técnicas de Cocultivo , Citocinas/inmunología , Citocinas/metabolismo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Exosomas/metabolismo , Humanos , Huésped Inmunocomprometido/inmunología , Factores Inmunológicos/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratas , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Heterólogo
5.
Methods Mol Biol ; 2236: 177-187, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33237548

RESUMEN

Single-cell transcriptomics is a powerful tool to study previously unrealized cellular heterogeneity at the resolution of individual cells. Most of the previous knowledge in cell biology is based on data generated by bulk analysis methods, which provide averaged readouts that usually mask cellular heterogeneity. This approach is challenging when the biological effect of interest is limited to a subpopulation within a cell type. This may particularly apply immune cell populations as these cells are highly mobile and swiftly respond to changes in cytokines or chemokines. For example, in cancer certain subset of myeloid immune cells may acquire immunosuppressive features to suppress antitumor immune responses, and thus described as myeloid-derived suppressor cells (MDSCs). Advances in single-cell RNA sequencing (scRNAseq) allowed scientists to overcome this limitation and enable in-depth interrogation of these subsets of immune cells including MDSCs. Here, we provide a detailed protocol for using scRNAseq to explore MDSCs in the context of splenic myeloid cells from breast tumor-bearing mice in comparison to wildtype controls to define the unique molecular features of immunosuppressive myeloid cells.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Neoplasias Mamarias Animales/patología , Células Supresoras de Origen Mieloide/metabolismo , Análisis de la Célula Individual/métodos , Bazo/patología , Animales , Biología Computacional , Femenino , Ratones Transgénicos , Control de Calidad , Reproducibilidad de los Resultados , Flujo de Trabajo
6.
Nat Cell Biol ; 22(3): 310-320, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32144411

RESUMEN

Although metastasis remains the cause of most cancer-related mortality, mechanisms governing seeding in distal tissues are poorly understood. Here, we establish a robust method for the identification of global transcriptomic changes in rare metastatic cells during seeding using single-cell RNA sequencing and patient-derived-xenograft models of breast cancer. We find that both primary tumours and micrometastases display transcriptional heterogeneity but micrometastases harbour a distinct transcriptome program conserved across patient-derived-xenograft models that is highly predictive of poor survival of patients. Pathway analysis revealed mitochondrial oxidative phosphorylation as the top pathway upregulated in micrometastases, in contrast to higher levels of glycolytic enzymes in primary tumour cells, which we corroborated by flow cytometric and metabolomic analyses. Pharmacological inhibition of oxidative phosphorylation dramatically attenuated metastatic seeding in the lungs, which demonstrates the functional importance of oxidative phosphorylation in metastasis and highlights its potential as a therapeutic target to prevent metastatic spread in patients with breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Transcriptoma , Animales , Neoplasias de la Mama/metabolismo , Metabolismo Energético , Femenino , Humanos , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/metabolismo , Metástasis de la Neoplasia , Fosforilación Oxidativa , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcripción Genética
7.
Sci Immunol ; 5(44)2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32086381

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are innate immune cells that acquire the capacity to suppress adaptive immune responses during cancer. It remains elusive how MDSCs differ from their normal myeloid counterparts, which limits our ability to specifically detect and therapeutically target MDSCs during cancer. Here, we sought to determine the molecular features of breast cancer-associated MDSCs using the widely studied mouse model based on the mouse mammary tumor virus (MMTV) promoter-driven expression of the polyomavirus middle T oncoprotein (MMTV-PyMT). To identify MDSCs in an unbiased manner, we used single-cell RNA sequencing to compare MDSC-containing splenic myeloid cells from breast tumor-bearing mice with wild-type controls. Our computational analysis of 14,646 single-cell transcriptomes revealed that MDSCs emerge through an aberrant neutrophil maturation trajectory in the spleen that confers them an immunosuppressive cell state. We establish the MDSC-specific gene signature and identify CD84 as a surface marker for improved detection and enrichment of MDSCs in breast cancers.


Asunto(s)
Neoplasias de la Mama/patología , Células Supresoras de Origen Mieloide/patología , Análisis de la Célula Individual , Transcriptoma , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Neoplasias de la Mama/inmunología , Diferenciación Celular/genética , Femenino , Humanos , Ratones , Ratones Endogámicos , Ratones Transgénicos , Células Supresoras de Origen Mieloide/inmunología , ARN Neoplásico/genética , ARN Neoplásico/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología
8.
Lab Chip ; 18(18): 2776-2786, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30090895

RESUMEN

Tissues are increasingly being analyzed at the single cell level in order to characterize cellular diversity and identify rare cell types. Single cell analysis efforts are greatly limited, however, by the need to first break down tissues into single cell suspensions. Current dissociation methods are inefficient, leaving a significant portion of the tissue as aggregates that are filtered away or left to confound results. Here, we present a simple and inexpensive microfluidic device that simultaneously filters large tissue fragments and dissociates smaller aggregates into single cells, thereby improving single cell yield and purity. The device incorporates two nylon mesh membranes with well-defined, micron-sized pores that operate on aggregates of different size scales. We also designed the device so that the first filtration could be performed under tangential flow to minimize clogging. Using cancer cell lines, we demonstrated that aggregates were effectively dissociated using high flow rates and pore sizes that were smaller than a single cell. However, pore sizes that were less than half the cell size caused significant damage. We then improved results by passing the sample through two filter devices in series, with single cell yield and purity predominantly determined by the pore size of the second membrane. Next, we optimized performance using minced and digested murine kidney tissue samples, and determined that the combination of 50 and 15 µm membranes was optimal. Finally, we integrated these two membranes into a single filter device and performed validation experiments using minced and digested murine kidney, liver, and mammary tumor tissue samples. The dual membrane microfluidic filter device increased single cell numbers by at least 3-fold for each tissue type, and in some cases by more than 10-fold. These results were obtained in minutes without affecting cell viability, and additional filtering would not be required prior to downstream applications. In future work, we will create complete tissue analysis platforms by integrating the dual membrane microfluidic filter device with additional upstream tissue processing technologies, as well as downstream operations such as cell sorting and detection.


Asunto(s)
Agregación Celular , Separación Celular/instrumentación , Filtración/instrumentación , Dispositivos Laboratorio en un Chip , Membranas Artificiales , Nylons , Animales , Humanos , Riñón/citología , Células MCF-7 , Ratones , Análisis de la Célula Individual
9.
Nanomedicine ; 13(2): 383-390, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27558349

RESUMEN

A nanobiosensor for arginase detection was designed and synthesized. It features a central dopamine-coated iron/iron oxide nanoparticle to which sulfonated cyanine 7.0 is tethered via a stable amide bond. Cyanine 5.5 is linked to the N-terminal of the peptide sequence GRRRRRRRG. Arginine (R) reacts to ornithine (O) in the presence of arginase. Based on calibration with commercially obtained arginase II, the limit of detection (LOD) is picomolar. It is noteworthy that the nanobiosensor for arginase detection does not show a fluorescence increase when incubated with the enzyme NO-reductase, which also uses arginase as substrate, but is indicative of an inflammatory response by the host to cancer and infections. Arginase activity was determined in a syngeneic mouse model for aggressive breast cancer (4T1 tumors in BALB/c mice). It was found that the arginase activity is systemically enhanced, but especially pronounced in the active tumor regions.


Asunto(s)
Arginasa/metabolismo , Técnicas Biosensibles , Nanopartículas del Metal , Animales , Arginina , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/enzimología , Ratones Endogámicos BALB C , Óxido Nítrico , Ornitina
10.
PLoS One ; 10(5): e0128144, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26011247

RESUMEN

The recent WHO report on antibiotic resistances shows a dramatic increase of microbial resistance against antibiotics. With only a few new antibiotics in the pipeline, a different drug delivery approach is urgently needed. We have obtained evidence demonstrating the effectiveness of a cell based drug delivery system that utilizes the innate immune system as targeting carrier for antibacterial drugs. In this study we show the efficient loading of neutrophil granulocytes with chlorhexidine and the complete killing of E. coli as well as Fusobacterium necrophorum in in-vitro studies. Fusobacterium necrophorum causes hepatic abscesses in cattle fed high grain diets. We also show in a mouse model that this delivery system targets infections of F. necrophorum in the liver and reduces the bacterial burden by an order of magnitude from approximately 2•106 to 1•105.


Asunto(s)
Antiinfecciosos Locales/administración & dosificación , Enfermedades de los Bovinos/terapia , Clorhexidina/administración & dosificación , Absceso Hepático/veterinaria , Micrococcus luteus/química , Neutrófilos/trasplante , Animales , Antiinfecciosos Locales/uso terapéutico , Bovinos , Enfermedades de los Bovinos/microbiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Clorhexidina/uso terapéutico , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Escherichia coli/efectos de los fármacos , Fusobacterium necrophorum/efectos de los fármacos , Absceso Hepático/microbiología , Absceso Hepático/terapia , Ratones , Neutrófilos/química , Neutrófilos/microbiología
11.
J Photochem Photobiol B ; 127: 223-8, 2013 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-24077442

RESUMEN

Polymorphonuclear neutrophils (PMNs) are the most abundant circulating blood leukocytes. They are part of the innate immune system and provide a first line of defense by migrating toward areas of inflammation in response to chemical signals released from the site. Some solid tumors, such as breast cancer, also cause recruitment and activation of PMNs and release of myeloperoxidase. In this study, we demonstrate that administration of luminol to mice that have been transplanted with 4T1 mammary tumor cells permits the detection of myeloperoxidase activity, and consequently, the location of the tumor. Luminol allowed detection of activated PMNs only two days after cancer cell transplantation, even though tumors were not yet palpable. In conclusion, luminol-bioluminescence imaging (BLI) can provide a pathway towards detection of solid tumors at an early stage in preclinical tumor models.


Asunto(s)
Sustancias Luminiscentes/metabolismo , Mediciones Luminiscentes , Luminol/metabolismo , Neoplasias Mamarias Experimentales/patología , Imagen Molecular , Animales , Benzotiazoles/metabolismo , Línea Celular Tumoral , Femenino , Cinética , Neoplasias Mamarias Experimentales/enzimología , Ratones , Ratones Endogámicos BALB C , Peroxidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA