Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Immunity ; 38(4): 818-30, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23562160

RESUMEN

In comparison to murine dendritic cells (DCs), less is known about the function of human DCs in tissues. Here, we analyzed, by using lung tissues from humans and humanized mice, the role of human CD1c(+) and CD141(+) DCs in determining the type of CD8(+) T cell immunity generated to live-attenuated influenza virus (LAIV) vaccine. We found that both lung DC subsets acquired influenza antigens in vivo and expanded specific cytotoxic CD8(+) T cells in vitro. However, lung-tissue-resident CD1c(+) DCs, but not CD141(+) DCs, were able to drive CD103 expression on CD8(+) T cells and promoted CD8(+) T cell accumulation in lung epithelia in vitro and in vivo. CD1c(+) DCs induction of CD103 expression was dependent on membrane-bound cytokine TGF-ß1. Thus, CD1c(+) and CD141(+) DCs generate CD8(+) T cells with different properties, and CD1c(+) DCs specialize in the regulation of mucosal CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Pulmón/inmunología , Subgrupos de Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos CD1/metabolismo , Antígenos Virales/inmunología , Diferenciación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Glicoproteínas/metabolismo , Humanos , Inmunidad Mucosa , Memoria Inmunológica , Vacunas contra la Influenza/inmunología , Cadenas alfa de Integrinas/metabolismo , Pulmón/virología , Activación de Linfocitos , Ratones , Ratones SCID , Análisis por Micromatrices
2.
J Immunol ; 193(9): 4335-43, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25246496

RESUMEN

Dendritic cells (DCs) play the central role in the priming of naive T cells and the differentiation of unique effector T cells. In this study, using lung tissues and blood from both humans and humanized mice, we analyzed the response of human CD1c(+) and CD141(+) DC subsets to live-attenuated influenza virus. Specifically, we analyzed the type of CD4(+) T cell immunity elicited by live-attenuated influenza virus-exposed DCs. Both DC subsets induce proliferation of allogeneic naive CD4(+) T cells with the capacity to secrete IFN-γ. However, CD141(+) DCs are uniquely able to induce the differentiation of IL-4- and IL-13-producing CD4(+) T cells. CD141(+) DCs induce IL-4- and IL-13-secreting CD4(+) T cells through OX40 ligand. Thus, CD141(+) DCs demonstrate remarkable plasticity in guiding adaptive immune responses.


Asunto(s)
Antígenos de Superficie/metabolismo , Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Animales , Antígenos CD1/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/metabolismo , Diferenciación Celular , Células Cultivadas , Células Dendríticas/metabolismo , Glicoproteínas/metabolismo , Humanos , Inmunofenotipificación , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ligando OX40/metabolismo , Orthomyxoviridae/inmunología , Fenotipo , Transducción de Señal , Células Th2/inmunología , Células Th2/metabolismo , Trombomodulina
3.
iScience ; 27(3): 109238, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38433905

RESUMEN

Pre-clinical use of humanized mice transplanted with CD34+ hematopoietic stem and progenitor cells (HSPCs) is limited by insufficient engraftment with adult non-mobilized HSPCs. Here, we developed a novel immunodeficient mice based on NOD-SCID-Il2γc-/- (NSG) mice to support long-term engraftment with human adult HSPCs. As both Flt3L and IL-6 are critical for many aspects of hematopoiesis, we knock-out mouse Flt3 and knock-in human IL6 gene. The resulting mice showed an increase in the availability of mouse Flt3L to human cells and a dose-dependent production of human IL-6 upon activation. Upon transplantation with low number of human HSPCs from adult bone marrow, these humanized mice demonstrated a significantly higher engraftment with multilineage differentiation of human lymphoid and myeloid cells, and tissue colonization at one year after adult HSPC transplant. Thus, these mice enable studies of human hematopoiesis and tissue colonization over time and may facilitate building autologous models for immuno-oncology studies.

4.
STAR Protoc ; 5(3): 103155, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-38935509

RESUMEN

Humanized mice, defined as mice with human immune systems, have become an emerging model to study human hematopoiesis, infectious disease, and cancer. Here, we describe the techniques to generate humanized NSGF6 mice using adult human CD34+ hematopoietic stem and progenitor cells (HSPCs). We describe steps for constructing and monitoring the engraftment of humanized mice. We then detail procedures for tissue processing and immunophenotyping by flow cytometry to evaluate the multilineage hematopoietic differentiation. For complete details on the use and execution of this protocol, please refer to Yu et al.1.


Asunto(s)
Antígenos CD34 , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Animales , Células Madre Hematopoyéticas/citología , Ratones , Humanos , Antígenos CD34/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Citometría de Flujo/métodos , Inmunofenotipificación/métodos
5.
J Exp Med ; 204(5): 1037-47, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17438063

RESUMEN

We previously reported (Bell, D., P. Chomarat, D. Broyles, G. Netto, G.M. Harb, S. Lebecque, J. Valladeau, J. Davoust, K.A. Palucka, and J. Banchereau. 1999. J. Exp. Med. 190: 1417-1426) that breast cancer tumors are infiltrated with mature dendritic cells (DCs), which cluster with CD4(+) T cells. We now show that CD4(+) T cells infiltrating breast cancer tumors secrete type 1 (interferon gamma) as well as high levels of type 2 (interleukin [IL] 4 and IL-13) cytokines. Immunofluorescence staining of tissue sections revealed intense IL-13 staining on breast cancer cells. The expression of phosphorylated signal transducer and activator of transcription 6 in breast cancer cells suggests that IL-13 actually delivers signals to cancer cells. To determine the link between breast cancer, DCs, and CD4(+) T cells, we implanted human breast cancer cell lines in nonobese diabetic/LtSz-scid/scid beta2 microglobulin-deficient mice engrafted with human CD34(+) hematopoietic progenitor cells and autologous T cells. There, CD4(+) T cells promote early tumor development. This is dependent on DCs and can be partially prevented by administration of IL-13 antagonists. Thus, breast cancer targets DCs to facilitate its development.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Linfocitos T CD4-Positivos/metabolismo , Células Dendríticas/metabolismo , Interleucina-13/metabolismo , Transducción de Señal/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Trasplante de Células Madre Hematopoyéticas , Humanos , Interferón gamma/metabolismo , Interleucina-13/inmunología , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factor de Transcripción STAT6/metabolismo
6.
bioRxiv ; 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37873457

RESUMEN

Pre-clinical use of humanized mice transplanted with CD34 + hematopoietic progenitor cells (HPCs) is limited by insufficient engraftment with adult HPCs. Here, we developed a novel immunodeficient mice based in NOD-SCID- Il2γc -/- (NSG) mice to support long-term engraftment with human adult HPCs and tissue colonization with human myeloid cells. As both Flt3L and IL-6 are critical for many aspects of hematopoiesis, we knock-out mouse Flt3 and knock-in human IL6 gene. The resulting mice showed an increase in the availability of mouse Flt3L to human cells, and a dose-dependent production of human IL-6 upon activation. Upon transplantation with low number of human HPCs from adult bone marrow, these humanized mice demonstrated a significantly higher engraftment with multilineage differentiation of human lymphoid and myeloid cells. Furthermore, higher frequencies of human lymphoid and myeloid cells were detected in tissues at one year after adult HPC transplant. Thus, these mice enable studies of human hematopoiesis and tissue colonization over time. Summary: Pre-clinical use of humanized mice is limited by insufficient engraftment with adult hematopoietic progenitor cells (HPCs). Here, we developed a novel immunodeficient mice which support long-term engraftment with adult bone marrow HPCs and facilitate building autologous models for immuno-oncology studies.

7.
STAR Protoc ; 4(4): 102735, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37991921

RESUMEN

Primary human lung organoid-derived air-liquid interface (ALI) cultures serve as a physiologically relevant model to study human airway epithelium in vitro. Here, we present a protocol for establishing these cultures from cryopreserved human lung tissue. We describe steps for lung tissue cryostorage, tissue dissociation, lung epithelial organoid generation, and ALI culture differentiation. We also include quality control steps and technical readouts for monitoring virus response. This protocol demonstrates severe acute respiratory syndrome coronavirus 2 infection in these cultures as an example of their utility. For complete details on the use and execution of this protocol, please refer to Diana Cadena Castaneda et al. (2023).1.


Asunto(s)
Células Epiteliales , Pulmón , Humanos , Células Cultivadas , Organoides
8.
bioRxiv ; 2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37034597

RESUMEN

The COVID-19 pandemic continues to be a health crisis with major unmet medical needs. The early responses from airway epithelial cells, the first target of the virus regulating the progression towards severe disease, are not fully understood. Primary human air-liquid interface cultures representing the broncho-alveolar epithelia were used to study the kinetics and dynamics of SARS-CoV-2 variants infection. The infection measured by nucleoprotein expression, was a late event appearing between day 4-6 post infection for Wuhan-like virus. Other variants demonstrated increasingly accelerated timelines of infection. All variants triggered similar transcriptional signatures, an "early" inflammatory/immune signature preceding a "late" type I/III IFN, but differences in the quality and kinetics were found, consistent with the timing of nucleoprotein expression. Response to virus was spatially organized: CSF3 expression in basal cells and CCL20 in apical cells. Thus, SARS-CoV-2 virus triggers specific responses modulated over time to engage different arms of immune response.

9.
iScience ; 26(8): 107374, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37520727

RESUMEN

The COVID-19 pandemic continues to be a health crisis with major unmet medical needs. The early responses from airway epithelial cells, the first target of the virus regulating the progression toward severe disease, are not fully understood. Primary human air-liquid interface cultures representing the broncho-alveolar epithelia were used to study the kinetics and dynamics of SARS-CoV-2 variants infection. The infection measured by nucleoprotein expression, was a late event appearing between day 4-6 post infection for Wuhan-like virus. Other variants demonstrated increasingly accelerated timelines of infection. All variants triggered similar transcriptional signatures, an "early" inflammatory/immune signature preceding a "late" type I/III IFN, but differences in the quality and kinetics were found, consistent with the timing of nucleoprotein expression. Response to virus was spatially organized: CSF3 expression in basal cells and CCL20 in apical cells. Thus, SARS-CoV-2 virus triggers specific responses modulated over time to engage different arms of immune response.

10.
STAR Protoc ; 3(4): 101698, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36149794

RESUMEN

We describe a pipeline for optimized and streamlined multiplexed immunofluorescence-guided laser capture microdissection allowing the harvest of individual cells based on their phenotype and tissue localization for transcriptomic analysis with next-generation RNA sequencing. Here, we analyze transcriptomes of CD3+ T cells, CD14+ monocytes/macrophages, and melanoma cells in non-dissociated metastatic melanoma tissue. While this protocol is described for melanoma tissues, we successfully applied it to human tonsil, skin, and breast cancer tissues as well as mouse lung tissues. For complete details on the use and execution of this protocol, please refer to Martinek et al. (2022).


Asunto(s)
Captura por Microdisección con Láser , Melanoma , Animales , Humanos , Ratones , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica/métodos , Captura por Microdisección con Láser/métodos , Melanoma/genética , Melanoma/cirugía , Transcriptoma/genética
11.
Cell Rep Med ; 3(5): 100621, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35584631

RESUMEN

Modulation of immune function at the tumor site could improve patient outcomes. Here, we analyze patient samples of metastatic melanoma, a tumor responsive to T cell-based therapies, and find that tumor-infiltrating T cells are primarily juxtaposed to CD14+ monocytes/macrophages rather than melanoma cells. Using immunofluorescence-guided laser capture microdissection, we analyze transcriptomes of CD3+ T cells, CD14 + monocytes/macrophages, and melanoma cells in non-dissociated tissue. Stromal CD14+ cells display a specific transcriptional signature distinct from CD14+ cells within tumor nests. This signature contains LY75, a gene linked with antigen capture and regulation of tolerance and immunity in dendritic cells (DCs). When applied to TCGA cohorts, this gene set can distinguish patients with significantly prolonged survival in metastatic cutaneous melanoma and other cancers. Thus, the stromal CD14+ cell signature represents a candidate biomarker and suggests that reprogramming of stromal macrophages to acquire DC function may offer a therapeutic opportunity for metastatic cancers.


Asunto(s)
Melanoma , Neoplasias Primarias Secundarias , Neoplasias Cutáneas , Humanos , Macrófagos , Melanoma/genética , Fenotipo , Neoplasias Cutáneas/genética , Linfocitos T
12.
J Exp Med ; 218(6)2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33857287

RESUMEN

Metastasis of melanoma significantly worsens prognosis; thus, therapeutic interventions that prevent metastasis could improve patient outcomes. Here, we show using humanized mice that colonization of distant visceral organs with melanoma is dependent upon a human CD33+CD11b+CD117+ progenitor cell subset comprising <4% of the human CD45+ leukocytes. Metastatic tumor-infiltrating CD33+ cells from patients and humanized (h)NSG-SGM3 mice showed converging transcriptional profiles. Single-cell RNA-seq analysis identified a gene signature of a KIT/CD117-expressing CD33+ subset that correlated with decreased overall survival in a TCGA melanoma cohort. Thus, human CD33+CD11b+CD117+ myeloid cells represent a novel candidate biomarker as well as a therapeutic target for metastatic melanoma.


Asunto(s)
Melanoma/metabolismo , Melanoma/patología , Células Mieloides/metabolismo , Células Mieloides/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Biomarcadores/metabolismo , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Estudios de Cohortes , Humanos , Antígenos Comunes de Leucocito/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , Ratones , Ratones Endogámicos NOD , Pronóstico
13.
Blood ; 112(9): 3671-8, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18713944

RESUMEN

The development of novel human vaccines would be greatly facilitated by the development of in vivo models that permit preclinical analysis of human immune responses. Here, we show that nonobese diabetic severe combined immunodeficiency (NOD/SCID) beta(2) microglobulin(-/-) mice, engrafted with human CD34+ hematopoietic progenitors and further reconstituted with T cells, can mount specific immune responses against influenza virus vaccines. Live attenuated trivalent influenza virus vaccine induces expansion of CD8+ T cells specific to influenza matrix protein (FluM1) and nonstructural protein 1 in blood, spleen, and lungs. On ex vivo exposure to influenza antigens, antigen-specific CD8+ T cells produce IFN-gamma and express cell-surface CD107a. FluM1-specific CD8+ T cells can be also expanded in mice vaccinated with inactivated trivalent influenza virus vaccine. Expansion of antigen-specific CD8+ T cells is dependent on reconstitution of the human myeloid compartment. Thus, this humanized mouse model permits preclinical testing of vaccines designed to induce cellular immunity, including those against influenza virus. Furthermore, this work sets the stage for systematic analysis of the in vivo functions of human DCs. This, in turn, will allow a new approach to the rational design and preclinical testing of vaccines that cannot be tested in human volunteers.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra la Influenza/farmacología , Traslado Adoptivo , Animales , Células Dendríticas/inmunología , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunidad Celular , Vacunas contra la Influenza/inmunología , Transfusión de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Linfocitos T/trasplante , Toxoide Tetánico/inmunología , Toxoide Tetánico/farmacología , Trasplante Heterólogo , Proteínas de la Matriz Viral/inmunología , Proteínas no Estructurales Virales/inmunología , Microglobulina beta-2/deficiencia , Microglobulina beta-2/genética
14.
Methods Enzymol ; 636: 351-368, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32178826

RESUMEN

Mouse models of human cancer have been used extensively to circumvent the complexity in human patients. However, murine models often inadequately recapitulate the human cancer-immune interface partly due to important differences between mouse and human immune systems. Immunodeficient mice when transplanted with CD34+ hematopoietic progenitor cells (HPCs) develop multilineage human immune cells. While there remain limitations, efforts have been made to improve the function of human immune system. Thus, humanized mice, defined as mice with human immune system, have become an emerging model to study human cancers. Humanized mouse models have been used for various areas of cancer research including adoptive transfer of chimeric antigen receptor (CAR)-modified T cells, neoantigen vaccination to increase T cell repertoire and reprograming tumor microenvironment. Here, we describe the essential techniques to generate humanized mouse models for immuno-oncology studies.


Asunto(s)
Neoplasias , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Neoplasias/terapia , Linfocitos T , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Res ; 78(18): 5243-5258, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30012670

RESUMEN

Inflammation affects tumor immune surveillance and resistance to therapy. Here, we show that production of IL1ß in primary breast cancer tumors is linked with advanced disease and originates from tumor-infiltrating CD11c+ myeloid cells. IL1ß production is triggered by cancer cell membrane-derived TGFß. Neutralizing TGFß or IL1 receptor prevents breast cancer progression in humanized mouse model. Patients with metastatic HER2- breast cancer display a transcriptional signature of inflammation in the blood leukocytes, which is attenuated after IL1 blockade. When present in primary breast cancer tumors, this signature discriminates patients with poor clinical outcomes in two independent public datasets (TCGA and METABRIC).Significance: IL1ß orchestrates tumor-promoting inflammation in breast cancer and can be targeted in patients using an IL1 receptor antagonist. Cancer Res; 78(18); 5243-58. ©2018 AACRSee related commentary by Dinarello, p. 5200.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Interleucina-1beta/metabolismo , Transcripción Genética , Animales , Neoplasias de la Mama/tratamiento farmacológico , Antígeno CD11c/metabolismo , Capecitabina/administración & dosificación , Línea Celular Tumoral , Membrana Celular/metabolismo , Femenino , Furanos/administración & dosificación , Humanos , Inflamación , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Cetonas/administración & dosificación , Leucocitos Mononucleares/citología , Macrófagos/metabolismo , Ratones , Ratones SCID , Células Mieloides/metabolismo , Metástasis de la Neoplasia , Trasplante de Neoplasias , Paclitaxel/administración & dosificación , Proyectos Piloto , Factor de Crecimiento Transformador beta/metabolismo
16.
Vaccine ; 34(41): 4857-4865, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27595442

RESUMEN

The targeting of vaccine antigens to antigen presenting cells (APC), such as dendritic cells (DCs), is a promising strategy for boosting vaccine immunogenicity and, in turn, protective and/or therapeutic efficacy. However, in vivo systems are needed to evaluate the potential of this approach for testing human vaccines. To this end, we examined human CD8(+) T-cell expansion to novel DC-targeting vaccines in vitro and in vivo in humanized mice. Vaccines incorporating the influenza matrix protein-1 (FluM1) antigen fused to human specific antibodies targeting different DC receptors, including DEC-205, DCIR, Dectin-1, and CD40, elicited human CD8(+) T-cell responses, as defined by the magnitude of specific CD8(+) T-cells to the targeted antigen. In vitro we observed differences in response to the different vaccines, particularly between the weakly immunogenic DEC-205-targeted and more strongly immunogenic CD40-targeted vaccines, consistent with previous studies. However, in humanized mice adoptively transferred (AT) with mature human T cells (HM-T), vaccines that performed weakly in vitro (i.e., DEC-205, DCIR, and Dectin-1) gave stronger responses in vivo, some resembling those of the strongly immunogenic CD40-targeted vaccine. These results demonstrate the utility of the humanized mouse model as a platform for studies of human vaccines.


Asunto(s)
Traslado Adoptivo , Anticuerpos Monoclonales/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Vacunas contra la Influenza/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Presentación de Antígeno , Antígenos CD/inmunología , Antígenos CD40/inmunología , Reactividad Cruzada , Epítopos/inmunología , Humanos , Inmunidad Celular , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Antígenos de Histocompatibilidad Menor/inmunología , Receptores de Superficie Celular/inmunología , Proteínas Recombinantes de Fusión/inmunología
17.
Int J Parasitol ; 32(8): 969-77, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12076626

RESUMEN

Toxoplasma gondii is a medically important, obligate intracellular parasite. Little is known regarding factors that regulate its replication within cells. Such knowledge would further understanding of T. gondii pathogenesis, and might lead to novel therapeutic strategies. Mitogen-activated protein kinases (MAPKs) govern diverse cellular processes including proliferation and differentiation. We now show that treatment of T. gondii-infected cells with SB203580 or SB202190, substituted pyridinylimidazoles that are potent inhibitors of human p38 MAPK, inhibits intracellular T. gondii replication. Several independent experimental approaches suggest that the anti-proliferative effects of pyridinylimidazoles depend on direct action on tachyzoites, not the host cell: (i) selective inhibition of host p38 MAPK using recombinant adenoviruses had little effect on tachyzoite replication, (ii) pyridinylimidazole-treated tachyzoites developed abnormal morphology suggesting defective parasite division, and (iii) pyridinylimidazole-resistant mutant tachyzoites were developed through culture in progressively higher drug concentrations. We hypothesise that pyridinylimidazoles target a human p38 MAPK homologue in tachyzoites that regulates their replication. Phylogenetic data suggest that T. gondii likely encodes a p38 MAPK homologue, but such a homologue is absent from the incomplete Toxoplasma genomic data base. As all eukaryotic pathogens, including agents of malaria, leishmaniasis and trypanosomiasis encode endogenous MAPKs, drugs inhibiting endogenous MAPK activation may represent a novel, potentially broadly-acting class of anti-parasitic agents. Pyridinylimidazoles also represent tools to elucidate factors governing intracellular tachyzoite replication.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Imidazoles/farmacología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Toxoplasma/efectos de los fármacos , Toxoplasma/crecimiento & desarrollo , Animales , Butadienos/farmacología , Células Cultivadas , Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/parasitología , Concentración 50 Inhibidora , Mutación/genética , Nitrilos/farmacología , Piridinas/farmacología , Toxoplasma/enzimología , Toxoplasma/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
18.
Nat Biotechnol ; 32(4): 364-72, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24633240

RESUMEN

Mice repopulated with human hematopoietic cells are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, existing humanized mouse models cannot support development of human innate immune cells, including myeloid cells and natural killer (NK) cells. Here we describe two mouse strains called MITRG and MISTRG, in which human versions of four genes encoding cytokines important for innate immune cell development are knocked into their respective mouse loci. The human cytokines support the development and function of monocytes, macrophages and NK cells derived from human fetal liver or adult CD34(+) progenitor cells injected into the mice. Human macrophages infiltrated a human tumor xenograft in MITRG and MISTRG mice in a manner resembling that observed in tumors obtained from human patients. This humanized mouse model may be used to model the human immune system in scenarios of health and pathology, and may enable evaluation of therapeutic candidates in an in vivo setting relevant to human physiology.


Asunto(s)
Inmunidad Innata/genética , Inmunidad Innata/inmunología , Células Asesinas Naturales , Células Mieloides , Animales , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/fisiología , Infiltración Leucémica/genética , Infiltración Leucémica/inmunología , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/fisiología , Neoplasias Experimentales , Trasplante Heterólogo
19.
Cancer Immunol Res ; 2(5): 487-500, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24795361

RESUMEN

Our studies showed that tumor-infiltrating dendritic cells (DC) in breast cancer drive inflammatory Th2 (iTh2) cells and protumor inflammation. Here, we show that intratumoral delivery of the ß-glucan curdlan, a ligand of dectin-1, blocks the generation of iTh2 cells and prevents breast cancer progression in vivo. Curdlan reprograms tumor-infiltrating DCs via the ligation of dectin-1, enabling the DCs to become resistant to cancer-derived thymic stromal lymphopoietin (TSLP), to produce IL-12p70, and to favor the generation of Th1 cells. DCs activated via dectin-1, but not those activated with TLR-7/8 ligand or poly I:C, induce CD8+ T cells to express CD103 (αE integrin), a ligand for cancer cells, E-cadherin. Generation of these mucosal CD8+ T cells is regulated by DC-derived integrin αvß8 and TGF-ß activation in a dectin-1-dependent fashion. These CD103+ CD8+ mucosal T cells accumulate in the tumors, thereby increasing cancer necrosis and inhibiting cancer progression in vivo in a humanized mouse model of breast cancer. Importantly, CD103+ CD8+ mucosal T cells elicited by reprogrammed DCs can reject established cancer. Thus, reprogramming tumor-infiltrating DCs represents a new strategy for cancer rejection.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Membrana Mucosa/inmunología , Animales , Neoplasias de la Mama/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Lectinas Tipo C/metabolismo , Ratones , Membrana Mucosa/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th2/efectos de los fármacos , Células Th2/inmunología , Factor de Crecimiento Transformador beta/metabolismo , beta-Glucanos/inmunología , beta-Glucanos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA