Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
iScience ; 27(3): 109238, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38433905

RESUMO

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.

2.
STAR Protoc ; 4(4): 102735, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37991921

RESUMO

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.


Assuntos
Células Epiteliais , Pulmão , Humanos , Células Cultivadas , Organoides
3.
bioRxiv ; 2023 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-37873457

RESUMO

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.

4.
iScience ; 26(8): 107374, 2023 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-37520727

RESUMO

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.

5.
bioRxiv ; 2023 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-37034597

RESUMO

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.

6.
STAR Protoc ; 3(4): 101698, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36149794

RESUMO

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).


Assuntos
Microdissecção e Captura a Laser , Melanoma , Animais , Humanos , Camundongos , Imunofluorescência , Perfilação da Expressão Gênica/métodos , Microdissecção e Captura a Laser/métodos , Melanoma/genética , Melanoma/cirurgia , Transcriptoma/genética
7.
Cell Rep Med ; 3(5): 100621, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35584631

RESUMO

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.


Assuntos
Melanoma , Segunda Neoplasia Primária , Neoplasias Cutâneas , Humanos , Macrófagos , Melanoma/genética , Fenótipo , Neoplasias Cutâneas/genética , Linfócitos T
8.
J Exp Med ; 218(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33857287

RESUMO

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.


Assuntos
Melanoma/metabolismo , Melanoma/patologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Biomarcadores/metabolismo , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Estudos de Coortes , Humanos , Antígenos Comuns de Leucócito/metabolismo , Leucócitos/metabolismo , Leucócitos/patologia , Camundongos , Camundongos Endogâmicos NOD , Prognóstico
9.
Methods Enzymol ; 636: 351-368, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32178826

RESUMO

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.


Assuntos
Neoplasias , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Neoplasias/terapia , Linfócitos T , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 78(18): 5243-5258, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30012670

RESUMO

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.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-1beta/metabolismo , Transcrição Gênica , Animais , Neoplasias da Mama/tratamento farmacológico , Antígeno CD11c/metabolismo , Capecitabina/administração & dosagem , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Feminino , Furanos/administração & dosagem , Humanos , Inflamação , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Cetonas/administração & dosagem , Leucócitos Mononucleares/citologia , Macrófagos/metabolismo , Camundongos , Camundongos SCID , Células Mieloides/metabolismo , Metástase Neoplásica , Transplante de Neoplasias , Paclitaxel/administração & dosagem , Projetos Piloto , Fator de Crescimento Transformador beta/metabolismo
12.
Vaccine ; 34(41): 4857-4865, 2016 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-27595442

RESUMO

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.


Assuntos
Transferência Adotiva , Anticorpos Monoclonais/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Vacinas contra Influenza/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Apresentação de Antígeno , Antígenos CD/imunologia , Antígenos CD40/imunologia , Apresentação Cruzada , Epitopos/imunologia , Humanos , Imunidade Celular , Lectinas Tipo C/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Antígenos de Histocompatibilidade Menor/imunologia , Receptores de Superfície Celular/imunologia , Proteínas Recombinantes de Fusão/imunologia
13.
J Immunol ; 193(9): 4335-43, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25246496

RESUMO

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.


Assuntos
Antígenos de Superfície/metabolismo , Linfócitos T CD4-Positivos/imunologia , Citocinas/biossíntese , Células Dendríticas/imunologia , Animais , Antígenos CD1/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD40/metabolismo , Diferenciação Celular , Células Cultivadas , Células Dendríticas/metabolismo , Glicoproteínas/metabolismo , Humanos , Imunofenotipagem , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Ligante OX40/metabolismo , Orthomyxoviridae/imunologia , Fenótipo , Transdução de Sinais , Células Th2/imunologia , Células Th2/metabolismo , Trombomodulina
14.
Cancer Immunol Res ; 2(5): 487-500, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24795361

RESUMO

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.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Mucosa/imunologia , Animais , Neoplasias da Mama/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Lectinas Tipo C/metabolismo , Camundongos , Mucosa/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Fator de Crescimento Transformador beta/metabolismo , beta-Glucanas/imunologia , beta-Glucanas/farmacologia
15.
Nat Biotechnol ; 32(4): 364-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24633240

RESUMO

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.


Assuntos
Imunidade Inata/genética , Imunidade Inata/imunologia , Células Matadoras Naturais , Células Mieloides , Animais , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/fisiologia , Infiltração Leucêmica/genética , Infiltração Leucêmica/imunologia , Camundongos , Camundongos Transgênicos , Células Mieloides/imunologia , Células Mieloides/fisiologia , Neoplasias Experimentais , Transplante Heterólogo
16.
Immunity ; 38(4): 818-30, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23562160

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Pulmão/imunologia , Subpopulações de Linfócitos T/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos CD1/metabolismo , Antígenos Virais/imunologia , Diferenciação Celular , Células Cultivadas , Citotoxicidade Imunológica , Glicoproteínas/metabolismo , Humanos , Imunidade nas Mucosas , Memória Imunológica , Vacinas contra Influenza/imunologia , Cadeias alfa de Integrinas/metabolismo , Pulmão/virologia , Ativação Linfocitária , Camundongos , Camundongos SCID , Análise em Microsséries
17.
J Exp Med ; 208(3): 479-90, 2011 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-21339324

RESUMO

The human breast tumor microenvironment can display features of T helper type 2 (Th2) inflammation, and Th2 inflammation can promote tumor development. However, the molecular and cellular mechanisms contributing to Th2 inflammation in breast tumors remain unclear. Here, we show that human breast cancer cells produce thymic stromal lymphopoietin (TSLP). Breast tumor supernatants, in a TSLP-dependent manner, induce expression of OX40L on dendritic cells (DCs). OX40L(+) DCs are found in primary breast tumor infiltrates. OX40L(+) DCs drive development of inflammatory Th2 cells producing interleukin-13 and tumor necrosis factor in vitro. Antibodies neutralizing TSLP or OX40L inhibit breast tumor growth and interleukin-13 production in a xenograft model. Thus, breast cancer cell-derived TSLP contributes to the inflammatory Th2 microenvironment conducive to breast tumor development by inducing OX40L expression on DCs.


Assuntos
Neoplasias da Mama/fisiopatologia , Citocinas/fisiologia , Inflamação/fisiopatologia , Células Th2/fisiologia , Animais , Anticorpos Antineoplásicos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/fisiologia , Camundongos , Transplante de Neoplasias , Ligante OX40/fisiologia , Células Th2/imunologia , Linfopoietina do Estroma do Timo
18.
Blood ; 112(9): 3671-8, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18713944

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas contra Influenza/farmacologia , Transferência Adotiva , Animais , Células Dendríticas/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunidade Celular , Vacinas contra Influenza/imunologia , Transfusão de Linfócitos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Linfócitos T/transplante , Toxoide Tetânico/imunologia , Toxoide Tetânico/farmacologia , Transplante Heterólogo , Proteínas da Matriz Viral/imunologia , Proteínas não Estruturais Virais/imunologia , Microglobulina beta-2/deficiência , Microglobulina beta-2/genética
19.
J Exp Med ; 204(5): 1037-47, 2007 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-17438063

RESUMO

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.


Assuntos
Neoplasias da Mama/fisiopatologia , Linfócitos T CD4-Positivos/metabolismo , Células Dendríticas/metabolismo , Interleucina-13/metabolismo , Transdução de Sinais/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Citometria de Fluxo , Imunofluorescência , Transplante de Células-Tronco Hematopoéticas , Humanos , Interferon gama/metabolismo , Interleucina-13/imunologia , Interleucina-4/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fator de Transcrição STAT6/metabolismo
20.
Blood ; 102(9): 3302-10, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12869510

RESUMO

Distinct human dendritic cell (DC) subsets differentially control immunity. Thus, insights into their in vivo functions are important to understand the launching and modulation of immune responses. We show that nonobese diabetic/LtSz-scid/scid (NOD/SCID) mice engrafted with human CD34+ hematopoietic progenitors develop human myeloid and plasmacytoid DCs. The skin displays immature DCs expressing Langerin, while other tissues display interstitial DCs. Myeloid DCs from these mice induce proliferation of allogeneic CD4 T cells in vitro, and bone marrow human cells containing plasmacytoid DCs release interferon-alpha (IFN-alpha) upon influenza virus exposure. Injection of influenza virus into reconstituted mice triggers IFN-alpha release and maturation of mDCs. Thus, these mice may provide a model to study the pathophysiology of human DC subsets.


Assuntos
Antígenos CD34 , Células Dendríticas/citologia , Transplante de Células-Tronco Hematopoéticas , Animais , Células Sanguíneas , Células da Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/classificação , Células Dendríticas/imunologia , Humanos , Interferon-alfa/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos SCID , Orthomyxoviridae/imunologia , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA