Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Int Immunol ; 33(3): 183-189, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33027513

RESUMEN

Food allergy is a common disease caused by intake of allergen-containing foods, such as milk, eggs, peanuts and wheat. Systemic anaphylaxis is a severe hypersensitive allergic reaction resulting from degranulation of mast cells or basophils after cross-linking of surface high-affinity IgE receptors (Fcε-RI) with allergen-specific IgE and allergens. In this study, we developed a novel human mast cell/basophil-engrafted mouse model that recapitulates systemic anaphylaxis triggered by ß-lactoglobulin (BLG), a major allergen found in cow's milk. Human CD34+ hematopoietic stem cells were transferred into NOG (non-Tg) or NOG hIL-3/hGM-CSF transgenic (Tg) mice. After 14-16 weeks, bovine BLG-specific human IgE was intravenously injected into humanized mice, followed by intravenous or oral bovine BLG exposure 1 day later. Body temperature in Tg, but not in non-Tg, mice gradually decreased within 10 min, and 80% of Tg mice died within 1 h by intravenous BLG exposure. Serum histamine levels and anaphylaxis scores in Tg mice were markedly increased compared to non-Tg mice. Furthermore, these allergic symptoms were significantly inhibited by epinephrine treatment of the Tg mice. Therefore, the current NOG hIL-3/hGM-CSF Tg mouse model may be useful for development of novel anaphylaxis drugs for treatment of food allergies and for safety assessment of low-allergenicity extensively hydrolyzed cow's milk whey protein-based infant formulas.


Asunto(s)
Anafilaxia/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Inmunoglobulina E/inmunología , Lactoglobulinas/inmunología , Hipersensibilidad a la Leche/inmunología , Anafilaxia/mortalidad , Animales , Basófilos/inmunología , Bovinos , Modelos Animales de Enfermedad , Epinefrina/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Histamina/sangre , Humanos , Interleucina-3/genética , Interleucina-3/metabolismo , Mastocitos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos
2.
Cancer Sci ; 111(9): 3386-3394, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32639672

RESUMEN

Cell line-derived xenograft (CDX) models created by implanting cancer cell lines into immunodeficient mice have contributed largely to the development of cancer drug therapies. However, cell lines often lose their original biological characteristics through many passages and cancer tissues in CDX models have many cancer cells and few cancer stromal cells, therefore CDX models are currently considered not suitable for predicting the results of clinical studies. Conversely, patient-derived xenograft (PDX) models are gaining importance, as human cancer biological characteristics and microenvironments are recreated by implanting tumor tissue into immunodeficient mice. These highly expected, evidently beneficial PDX models have been used in some basic research and are becoming more generalized. However, quality control and quality assurance criteria have not been established for them, and challenges and problems in the utilization of valuable PDX models in drug development have yet to be clarified. In this report, we conducted a questionnaire survey among researchers in Japanese academic institutions and pharmaceutical companies to understand the current status of PDX models in Japan. Based on the questionnaire results, we summarized the situations surrounding respondent's utilization and quality control in the development of anticancer drugs and proposed several measures to facilitate the utilization of PDX models in the development of anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Modelos Animales de Enfermedad , Desarrollo de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Japón , Ratones , Especificidad de la Especie
3.
Biochem Biophys Res Commun ; 516(2): 480-485, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31230747

RESUMEN

Although Th17 cells are closely linked to cutaneous graft-versus-host-disease (GVHD) in mouse models, this association remains unclear in human GVHD. In this study, we established a novel xenogeneic cutaneous GVHD model using humanized mice. To induce the differentiation of human Th17 cells, we created transgenic NOG mice expressing human IL-1ß and IL-23 cytokines (hIL-1ß/23 Tg) and transplanted with human CD4+ T cells. The pathologies of cutaneous GVHD, such as a decrease in body weight, alopecia, and T cell inflammation in the skin, were observed much earlier in hIL-1ß/23 Tg mice compared with non-Tg mice after human CD4+ T cell transplantation. In the skin of Tg mice, IL-17- and IFNγ-producing pathogenic Th17 cells were significantly accumulated. Furthermore, high infiltration of murine neutrophils was seen in the skin of Tg mice, but not non-Tg mice, which may have been the cause of the severe alopecia. CD4+ T-cell-transferred hIL-1ß/23 Tg mice were therefore highly sensitive models for inducing cutaneous GVHD mediated by human pathogenic Th17 cells.


Asunto(s)
Progresión de la Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Interleucina-1beta/metabolismo , Interleucina-23/metabolismo , Trasplante de Piel/efectos adversos , Células Th17/patología , Animales , Humanos , Interferón gamma/metabolismo , Recuento de Linfocitos , Ratones Transgénicos
4.
Int J Mol Sci ; 20(11)2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167385

RESUMEN

The prevalence rates of allergic diseases are increasing worldwide, particularly in industrial countries. To date, many mouse models have been generated for allergy research; studies conducted using these models have suggested the importance of cross-talk between immune cells and tissue-resident non-immune cells in the onset of allergic diseases. However, there are several differences between the immune systems of rodents and humans, and human studies are limited. Thus, mice reconstituted with human immune cells are a novel tool for the preclinical evaluation of the efficacy and safety of developing drugs. Genetic technologies for generating humanized mice have improved markedly in recent years. In this review, we will discuss recent progress in allergy research using humanized mice and introduce our recent humanized mouse model of airway inflammation in human immune cells.


Asunto(s)
Hipersensibilidad/inmunología , Investigación , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Ratones , Ratones Transgénicos
5.
J Immunol ; 195(4): 1883-90, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26170385

RESUMEN

Immunodeficient hosts exhibit high acceptance of xenogeneic or neoplastic cells mainly due to lack of adaptive immunity, although it still remains to be elucidated how innate response affects the engraftment. IL-2R common γ-chain (IL-2Rγc) signaling is required for development of NK cells and a subset of dendritic cells producing IFN-γ. To better understand innate response in the absence of adaptive immunity, we examined amounts of metastatic foci in the livers after intrasplenic transfer of human colon cancer HCT116 cells into NOD/SCID versus NOD/SCID/IL-2Rγc (null) (NOG) hosts. The intravital microscopic imaging of livers in the hosts depleted of NK cells and/or macrophages revealed that IL-2Rγc function critically contributes to elimination of cancer cells without the need for NK cells and macrophages. In the absence of IL-2Rγc, macrophages play a role in the defense against tumors despite the NOD Sirpa allele, which allows human CD47 to bind to the encoded signal regulatory protein α to inhibit macrophage phagocytosis of human cells. Analogous experiments using human pancreas cancer MIA PaCa-2 cells provided findings roughly similar to those from the experiments using HCT116 cells except for lack of suppression of metastases by macrophages in NOG hosts. Administration of mouse IFN-γ to NOG hosts appeared to partially compensate lack of IL-2Rγc-dependent elimination of transferred HCT116 cells. These results provide insights into the nature of innate response in the absence of adaptive immunity, aiding in developing tumor xenograft models in experimental oncology.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Subunidad gamma Común de Receptores de Interleucina/genética , Neoplasias/genética , Neoplasias/inmunología , Receptores de Interleucina-2/genética , Animales , Puntos de Control del Ciclo Celular , Modelos Animales de Enfermedad , Células HCT116 , Humanos , Interferón gamma/administración & dosificación , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias Hepáticas/secundario , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/patología
6.
J Immunol ; 194(7): 3513-25, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25712215

RESUMEN

We generated a severe immunodeficient NOD/Shi-scid-IL-2Rγ(null) (NOG) mouse substrain expressing the transgenic human IL-2 gene (NOG-IL-2 Tg). Upon transfer of human cord blood-derived hematopoietic stem cells (HSCs), CD3(-)CD56(high)CD16(+/-) cells developed unexpectedly, predominantly in the NOG-IL-2 Tg (hu-HSC NOG-IL-2 Tg). These cells expressed various NK receptors, including NKp30, NKp44, NKp46, NKG2D, and CD94, as well as a diverse set of killer cell Ig-like receptor molecules at levels comparable to normal human NK cells from the peripheral blood, which is evidence of their maturity. They produced levels of granzyme A as high as in human peripheral blood-derived NK cells, and a considerable amount of perforin protein was detected in the plasma. Human NK cells in hu-HSC NOG-IL-2 Tg produced IFN-γ upon stimulation, and IL-2, IL-15, or IL-12 treatment augmented the in vitro cytotoxicity. Inoculation of K562 leukemia cells into hu-HSC NOG-IL-2 Tg caused complete rejection of the tumor cells, whereas inoculation into hu-HSC NOG fully reconstituted with human B, T, and some NK cells did not. Moreover, when a CCR4(+) Hodgkin's lymphoma cell line was inoculated s.c. into hu-HSC NOG-IL-2 Tg, the tumor growth was significantly suppressed by treatment with a therapeutic humanized anti-CCR4 Ab (mogamulizumab), suggesting that the human NK cells in the mice exerted active Ab-dependent cellular cytotoxicity in vivo. Taken together, these data suggest that the new NOG-IL-2 Tg strain is a unique model that can be used to investigate the biological and pathological functions of human NK cells in vivo.


Asunto(s)
Interleucina-2/biosíntesis , Interleucina-2/genética , Células Asesinas Naturales/inmunología , Ratones Transgénicos , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos de Superficie/metabolismo , Diferenciación Celular , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Fenotipo , Receptores KIR/genética , Receptores KIR/metabolismo
7.
Biochem Biophys Res Commun ; 456(1): 219-24, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25462565

RESUMEN

The development of severe immunodeficient mouse strains containing various human genes, including cytokines or HLA, has enabled the reconstitution of functional human immune systems after transplantation of human hematopoietic stem cells (HSC). Accumulating evidence has suggested that HLA-restricted antigen-specific human T-cell responses can be generated in these humanized mice. To directly monitor immune responses of human CD4(+) T cells, we introduced ß-lactoglobulin (BLG)-specific T cell receptor (TCR) genes derived from CD4(+) T-cell clones of cow-milk allergy patients into HSCs, and subsequently transplanted them into NOG-HLA-DR4 transgenic/I-Aß deficient mice (NOG-DR4/I-A(o)). In the thymus, thymocytes with BLG-specific TCR preferentially differentiated into CD4(+)CD8(-) single-positive cells. Adoptive transfer of mature CD4(+) T cells expressing the TCR into recipient NOG-DR4/I-A(o) mice demonstrated that human CD4(+) T cells proliferated in response to antigenic stimulation and produced IFN-γ in vivo, suggesting that functional T-cell reactions (especially Th1-skewed responses) were induced in humanized mice.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Regulación de la Expresión Génica , Antígeno HLA-DR4/genética , Receptores de Antígenos de Linfocitos T/genética , Traslado Adoptivo , Animales , Antígenos CD34/metabolismo , Proliferación Celular , Trasplante de Células , Ensayo de Immunospot Ligado a Enzimas , Células HEK293 , Células Madre Hematopoyéticas/citología , Humanos , Interferón gamma/metabolismo , Lactoglobulinas/metabolismo , Lentivirus/genética , Leucocitos Mononucleares/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Timocitos/citología
8.
J Immunol ; 191(6): 2890-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23956433

RESUMEN

The development of animal models that mimic human allergic responses is crucial to study the pathophysiology of disease and to generate new therapeutic methodologies. Humanized mice reconstituted with human immune systems are essential to study human immune reactions in vivo and are expected to be useful for studying human allergies. However, application of this technology to the study of human allergies has been limited, largely because of the poor development of human myeloid cells, especially granulocytes and mast cells, which are responsible for mediating allergic diseases, in conventional humanized mice. In this study, we developed a novel transgenic (Tg) strain, NOD/Shi-scid-IL2rγ(null) (NOG), bearing human IL-3 and GM-CSF genes (NOG IL-3/GM-Tg). In this strain, a large number of human myeloid cells of various lineages developed after transplantation of human CD34⁺ hematopoietic stem cells. Notably, mature basophils and mast cells expressing FcεRI were markedly increased. These humanized NOG IL-3/GM-Tg mice developed passive cutaneous anaphylaxis reactions when administered anti-4-hydroxy-3-nitrophenylacetyl IgE Abs and 4-hydroxy-3-nitrophenylacetyl. More importantly, a combination of serum from Japanese cedar pollinosis patients and cedar pollen extract also elicited strong passive cutaneous anaphylaxis responses in mice. Thus, to our knowledge, our NOG IL-3/GM-Tg mice are the first humanized mouse model to enable the study of human allergic responses in vivo and are excellent tools for preclinical studies of allergic diseases.


Asunto(s)
Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Hipersensibilidad/inmunología , Interleucina-3/inmunología , Animales , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunohistoquímica , Interleucina-3/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
J Immunol ; 189(9): 4313-20, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23018460

RESUMEN

Xenograft animal models using immunodeficient mice have been widely applied in medical research on various human diseases. NOD/Shi-scid-IL2rγ(null) (NOG) mice are known to show an extremely high engraftment rate of xenotransplants compared with conventional immunodeficient mice. This high engraftment rate of xenotransplants in NOG mice was substantially suppressed by the transfer of spleen cells from NOD-scid mice that were devoid of NK cells. These results indicate that cell types other than splenic NK cells present in NOD-scid mice but not in NOG mice may be involved in this suppression. To identify the cell types responsible for this effect, we transferred subpopulations of spleen cells from NOD-scid mice into NOG mice and assessed the levels of human cell engraftment after human PBMC (hPBMC) transplantation. These experiments revealed that CD11c(+)B220(+) plasmacytoid dendritic cells (pDCs) from NOD-scid mice markedly inhibited engraftment of human cells. The CD11c(+)B220(+)CD122(+) cells further fractionated from the pDCs based on the expression of CD122, which is an NK cell marker strongly inhibited during hPBMC engraftment in NOG mice. Moreover, the CD122(+) cells in the pDC fraction were morphologically distinguishable from conventional CD122(+) NK cells and showed a higher rejection efficiency. The current results suggest that CD11c(+)B220(+)CD122(+) cells play an important role in xenograft rejection, and their absence in NOG mice may be critical in supporting the successful engraftment of xenotransplants.


Asunto(s)
Antígeno CD11c , Supervivencia de Injerto/inmunología , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad beta del Receptor de Interleucina-2/deficiencia , Antígenos Comunes de Leucocito/deficiencia , Trasplante Heterólogo/métodos , Animales , Antígeno CD11c/biosíntesis , Antígeno CD11c/genética , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Supervivencia de Injerto/genética , Humanos , Huésped Inmunocomprometido , Subunidad gamma Común de Receptores de Interleucina/biosíntesis , Subunidad beta del Receptor de Interleucina-2/biosíntesis , Subunidad beta del Receptor de Interleucina-2/genética , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Bazo/inmunología , Bazo/metabolismo , Bazo/trasplante
10.
Int Immunol ; 24(4): 243-52, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22402880

RESUMEN

Mounting evidence has demonstrated that NOD-Shi/scid/γc(null) (NOG) mice are one of the most suitable mouse strains for humanized mouse technologies, in which various human cells or tissues can be engrafted without rejection and autonomously maintained. We have characterized and analyzed various features of the human immune system reconstituted in NOG mice by transplanting human hematopoietic stem cells (hu-HSC). One of the problems of the quasi-immune system in these hu-HSC NOG mice is that the quality of immune responses is not always sufficient, as demonstrated by the lack of IgG production in response to antigen challenge. In this study, we established a novel transgenic NOG sub-strain of mice bearing the HLA-DRA and HLA-DRB1:0405 genes, which specifically expresses HLA-DR4 molecules in MHC II-positive cells. This mouse strain enabled us to match the haplotype of HLA-DR between the recipient mice and human donor HSC. We demonstrated that T-cell homeostasis was differentially regulated in HLA-matched hu-HSC NOG mice compared with HLA-mismatched control mice, and antibody class switching was induced after immunization with exogenous antigens in HLA-matched mice. This novel mouse strain improves the reconstituted human immune systems that develop in humanized mice and will contribute to future studies of human humoral immune responses.


Asunto(s)
Antígeno HLA-DR4/inmunología , Cadenas HLA-DRB1/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunidad Humoral , Linfocitos T/inmunología , Animales , Antígenos CD34/análisis , Células Cultivadas , Femenino , Antígeno HLA-DR4/genética , Cadenas HLA-DRB1/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Linfocitos T/metabolismo , Trasplante Heterólogo
11.
Cell Rep ; 41(12): 111841, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36543125

RESUMEN

Neutrophils are critical mediators during the early stages of innate inflammation in response to bacterial or fungal infections. A human hematopoietic system reconstituted in humanized mice aids in the study of human hematology and immunology. However, the poor development of human neutrophils is a well-known limitation of humanized mice. Here, we generate a human granulocyte colony-stimulating factor (hG-CSF) knockin (KI) NOD/Shi-scid-IL2rgnull (NOG) mouse in which hG-CSF is systemically expressed while the mouse G-CSF receptor is disrupted. These mice generate high numbers of mature human neutrophils, which can be readily mobilized into the periphery, compared with conventional NOG mice. Moreover, these neutrophils exhibit infection-mediated emergency granulopoiesis and are capable of efficient phagocytosis and reactive oxygen species production. Thus, hG-CSF KI mice provide a useful model for studying the development of human neutrophils, emergency granulopoiesis, and a potential therapeutic model for sepsis.


Asunto(s)
Mercurio , Neutrófilos , Humanos , Ratones , Animales , Factor Estimulante de Colonias de Granulocitos , Ratones Endogámicos NOD , Hematopoyesis
12.
Front Immunol ; 12: 671648, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34386001

RESUMEN

Despite recent advances in immunodeficient mouse models bearing human red blood cells (hRBCs), the elimination of circulating hRBCs by residual innate immune systems remains a significant challenge. In this study, we evaluated the role of mouse complement C3 in the elimination of circulating hRBCs by developing a novel NOG substrain harboring a truncated version of the murine C3 gene (NOG-C3ΔMG2-3). Genetic C3 deletion prolonged the survival of transfused hRBCs in the circulation. Chemical depletion and functional impairment of mouse macrophages, using clodronate liposomes (Clo-lip) or gadolinium chloride (GdCl3), respectively, further extended the survival of hRBCs in NOG-C3ΔMG2-3 mice. Low GdCl3 toxicity allowed the establishment of hRBC-bearing mice, in which hRBCs survived for more than 4 weeks with transfusion once a week. In addition, erythropoiesis of human hematopoietic stem cells (hHSCs) was possible in NOG-C3ΔMG2-3/human GM-CSF-IL-3 transgenic mice with Clo-lip treatment. These findings indicate that mouse models harboring hRBCs can be achieved using NOG-C3ΔMG2-3 mice, which could facilitate studies of human diseases associated with RBCs.


Asunto(s)
Complemento C3/deficiencia , Eritrocitos , Modelos Animales , Animales , Transfusión de Eritrocitos/métodos , Eritrocitos/inmunología , Eritropoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos
13.
Sci Rep ; 11(1): 21087, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702924

RESUMEN

Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer in the clinic. Further discovery of novel drugs or therapeutic protocols that enhance efficacy requires reliable animal models that recapitulate human immune responses to ICI treatment in vivo. In this study, we utilized an immunodeficient NOG mouse substrain deficient for mouse FcγR genes, NOG-FcγR-/- mice, to evaluate the anti-cancer effects of nivolumab, an anti-programmed cell death-1 (PD-1) antibody. After reconstitution of human immune systems by human hematopoietic stem cell transplantation (huNOG-FcγR-/- mice), four different programmed death-ligand 1 (PD-L1)-positive human cancer cell lines were tested. Among them, the growth of three cell lines was strongly suppressed by nivolumab in huNOG-FcγR-/- mice, but not in conventional huNOG mice. Accordingly, immunohistochemistry demonstrated the enhanced infiltration of human T cells into tumor parenchyma in only nivolumab-treated huNOG-FcγR-/- mice. Consistently, the number of human T cells was increased in the spleen in huNOG-FcγR-/- mice by nivolumab but not in huNOG mice. Furthermore, human PD-L1 expression was strongly induced in the spleen of huNOG-FcγR-/- mice. Collectively, our results suggest that the anti-cancer effects of anti-PD-1 antibodies can be detected more clearly in NOG-FcγR-/- mice than in NOG mice.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de Neoplasias , Neoplasias Experimentales , Nivolumab/farmacología , Receptor de Muerte Celular Programada 1 , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología
14.
Immunol Lett ; 229: 55-61, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33253759

RESUMEN

Humanized mice are widely used to study the human immune system in vivo and develop therapies for various human diseases. Human peripheral blood mononuclear cells (PBMC)-engrafted NOD/Shi-scid IL2rγnull (NOG) mice are useful models for characterization of human T cells. However, the development of graft-versus-host disease (GVHD) limits the use of NOG PBMC models. We previously established a NOG-major histocompatibility complex class I/II double knockout (dKO) mouse model. Although humanized dKO mice do not develop severe GVHD, they have impaired reproductive performance and reduced chimerism of human cells. In this study, we established a novel beta-2 microglobulin (B2m) KO mouse model using CRISPR/Cas9. By crossing B2m KO mice with I-Ab KO mice, we established a modified dKO (dKO-em) mouse model. Reproductivity was slightly improved in dKO-em mice, compared with conventional dKO (dKO-tm) mice. dKO-em mice showed no signs of GVHD after the transfer of human PBMCs; they also exhibited high engraftment efficiency. Engrafted human PBMCs survived significantly longer in the peripheral blood and spleens of dKO-em mice, compared with dKO-tm mice. In conclusion, dKO-em mice might constitute a promising PBMC-based humanized mouse model for the development and preclinical testing of novel therapeutics for human diseases.


Asunto(s)
Sistemas CRISPR-Cas , Trasplante de Células , Técnicas de Inactivación de Genes , Antígenos de Histocompatibilidad/genética , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Animales , Biomarcadores , Trasplante de Células/efectos adversos , Trasplante de Células/métodos , Edición Génica , Marcación de Gen , Sitios Genéticos , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/etiología , Humanos , Inmunohistoquímica , Inmunofenotipificación , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Modelos Animales , Índice de Severidad de la Enfermedad , Bazo/inmunología , Bazo/metabolismo
15.
Int Immunol ; 21(7): 843-58, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19515798

RESUMEN

'Humanized mice' are anticipated to be a valuable tool for studying the human immune system, but the reconstituted human immune cells have not yet been well characterized. Here, we extensively investigated the differentiation and functions of human B and T cells in a supra-immunodeficient mouse strain, NOD/shi-scid/gammac(null) (NOG) reconstituted with CD34(+) hematopoietic stem cells obtained from umbilical cord blood. In these hu-HSC NOG mice, the development of human B cells was partially blocked, and a significant number of B-cell progenitors accumulated in the spleen. The mature CD19(+)IgM(+)IgD(+) human B cells of the hu-HSC NOG mice could produce IgG in vivo and in vitro by antigenic stimulation. In contrast, although human T cells with an apparently normal phenotype developed, most of them could neither proliferate nor produce IL-2 in response to antigenic stimulation by anti-CD3 and anti-CD28 antibodies in vitro. The positive selection of human T cells in the thymus was sufficiently functional, if not complete, and mainly mediated by mouse class II, suggesting that the human T cells lost their function in the periphery. We found that multiple mechanisms were involved in the T-cell abnormalities. Collectively, our results demonstrate that further improvements are necessary before humanized mice with a functional human immune system are achieved.


Asunto(s)
Linfocitos B/inmunología , Proteínas Portadoras/inmunología , Células Madre Hematopoyéticas/inmunología , Células Precursoras de Linfocitos B/inmunología , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Anticuerpos/inmunología , Antígenos CD34/inmunología , Linfocitos B/efectos de los fármacos , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Complejo CD3/inmunología , Complejo CD3/metabolismo , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular/inmunología , Sangre Fetal/inmunología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Interleucina-2/farmacología , Interleucinas/farmacología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células Precursoras de Linfocitos B/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/efectos de los fármacos
16.
Front Immunol ; 11: 532684, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117338

RESUMEN

We generated an NOD/Shi-scid-IL2Rγ null (NOG) mouse deficient for the Fcer1g and Fcgr2b genes (NOG-FcγR-/- mice), in which monocytes/macrophages do not express activating (FcγRI, III, and IV) or inhibitory (FcγRIIB) Fcγ receptors. Antibody-dependent cellular cytotoxicity (ADCC) by innate immune cells was strongly reduced in this strain. Thus, while the growth of xenogeneic human tumors engrafted in conventional NOG mice was suppressed by innate cells upon specific antibody treatment, such growth inhibition was abrogated in NOG-FcγR-/- mice. Using this novel strain, we further produced NOG-FcγR-/--mice expressing human IL-15 (NOG-FcγR-/--hIL-15 Tg). The mice inherited unique features from each strain, i.e., the long-term sustenance of human natural killer (NK) cells, and the elimination of mouse innate cell-mediated ADCC. As a result, segregation of human NK cell-mediated ADCC from mouse cell-mediated ADCC was possible in the NOG-FcγR-/--hIL-15 Tg mice. Our results suggest that NOG-FcγR-/--hIL-15 Tg mice are useful for validating the in vivo function of antibody drug candidates.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Proteínas Portadoras/genética , Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Receptores de IgG/deficiencia , Animales , Proteínas Portadoras/inmunología , Humanos , Interleucina-15/genética , Ratones , Ratones Noqueados , Receptores de IgG/inmunología
17.
Mol Neurodegener ; 14(1): 12, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30832693

RESUMEN

BACKGROUND: Microglia are the principal innate immune defense cells of the centeral nervous system (CNS) and the target of the human immunodeficiency virus type one (HIV-1). A complete understanding of human microglial biology and function requires the cell's presence in a brain microenvironment. Lack of relevant animal models thus far has also precluded studies of HIV-1 infection. Productive viral infection in brain occurs only in human myeloid linage microglia and perivascular macrophages and requires cells present throughout the brain. Once infected, however, microglia become immune competent serving as sources of cellular neurotoxic factors leading to disrupted brain homeostasis and neurodegeneration. METHODS: Herein, we created a humanized bone-marrow chimera producing human "microglia like" cells in NOD.Cg-PrkdcscidIl2rgtm1SugTg(CMV-IL34)1/Jic mice. Newborn mice were engrafted intrahepatically with umbilical cord blood derived CD34+ hematopoietic stem progenitor cells (HSPC). After 3 months of stable engraftment, animals were infected with HIV-1ADA, a myeloid-specific tropic viral isolate. Virologic, immune and brain immunohistology were performed on blood, peripheral lymphoid tissues, and brain. RESULTS: Human interleukin-34 under the control of the cytomegalovirus promoter inserted in NSG mouse strain drove brain reconstitution of HSPC derived peripheral macrophages into microglial-like cells. These human cells expressed canonical human microglial cell markers that included CD14, CD68, CD163, CD11b, ITGB2, CX3CR1, CSFR1, TREM2 and P2RY12. Prior restriction to HIV-1 infection in the rodent brain rested on an inability to reconstitute human microglia. Thus, the natural emergence of these cells from ingressed peripheral macrophages to the brain could allow, for the first time, the study of a CNS viral reservoir. To this end we monitored HIV-1 infection in a rodent brain. Viral RNA and HIV-1p24 antigens were readily observed in infected brain tissues. Deep RNA sequencing of these infected mice and differential expression analysis revealed human-specific molecular signatures representative of antiviral and neuroinflammatory responses. CONCLUSIONS: This humanized microglia mouse reflected human HIV-1 infection in its known principal reservoir and showed the development of disease-specific innate immune inflammatory and neurotoxic responses mirroring what can occur in an infected human brain.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Interleucinas , Microglía/virología , Animales , Diferenciación Celular , VIH-1 , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Ratones Endogámicos NOD
18.
Immunol Lett ; 119(1-2): 97-102, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606456

RESUMEN

It remains unclear how superantigen induces unresponsiveness in stimulated T cells. We analyzed the chromatin status of the interleukin-2 (IL-2) promoter region in T cells stimulated with toxic shock syndrome toxin-1 (TSST-1) superantigen using T cell receptor-transgenic T cells responding to ovalbumin (OVA) and TSST-1. Compared to OVA stimulation, naïve T cells cultured with TSST-1 showed lower IL-2 expression after transient enhancement. Coincidentally, the acetylated histone H3 (AcH3) level at the IL-2 promoter region was first enhanced, and then decreased, in TSST-1-stimulated T cells. At the reduction stage of AcH3, histone deacetylase-1 (HDAC1) was markedly associated with the IL-2 promoter region in a TSST-1-specific manner without HDAC1 over-expression. The enhancement of HDAC1 association and IL-2 suppression was prevented by pre-treatment with HDAC inhibitor, but not once the anergy status was established. These results suggest that recruitment of HDAC1 in the IL-2 promoter region at the early stimulation stage with TSST-1 plays a pivotal role in sAg-induced anergy.


Asunto(s)
Toxinas Bacterianas/administración & dosificación , Linfocitos T CD4-Positivos/metabolismo , Enterotoxinas/administración & dosificación , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Activación de Linfocitos/inmunología , Regiones Promotoras Genéticas , Superantígenos/administración & dosificación , Acetilación/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Anergia Clonal/efectos de los fármacos , Histona Desacetilasa 1 , Histona Desacetilasas/inmunología , Histonas/antagonistas & inhibidores , Histonas/genética , Interleucina-2/biosíntesis , Interleucina-2/genética , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Unión Proteica , Receptores de Antígenos de Linfocitos T/genética
19.
Front Immunol ; 9: 152, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29456539

RESUMEN

The tumor microenvironment contains unique immune cells, termed myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs) that suppress host anti-tumor immunity and promote tumor angiogenesis and metastasis. Although these cells are considered a key target of cancer immune therapy, in vivo animal models allowing differentiation of human immunosuppressive myeloid cells have yet to be established, hampering the development of novel cancer therapies. In this study, we established a novel humanized transgenic (Tg) mouse strain, human interleukin (hIL)-6-expressing NOG mice (NOG-hIL-6 transgenic mice). After transplantation of human hematopoietic stem cells (HSCs), the HSC-transplanted NOG-hIL-6 Tg mice (HSC-NOG-hIL-6 Tg mice) showed enhanced human monocyte/macrophage differentiation. A significant number of human monocytes were negative for HLA-DR expression and resembled immature myeloid cells in the spleen and peripheral blood from HSC-NOG-hIL-6 Tg mice, but not from HSC-NOG non-Tg mice. Engraftment of HSC4 cells, a human head and neck squamous cell carcinoma-derived cell line producing various factors including IL-6, IL-1ß, macrophage colony-stimulating factor (M-CSF), and vascular endothelial growth factor (VEGF), into HSC-NOG-hIL-6 Tg mice induced a significant number of TAM-like cells, but few were induced in HSC-NOG non-Tg mice. The tumor-infiltrating macrophages in HSC-NOG-hIL-6 Tg mice expressed a high level of CD163, a marker of immunoregulatory myeloid cells, and produced immunosuppressive molecules such as arginase-1 (Arg-1), IL-10, and VEGF. Such cells from HSC-NOG-hIL-6 Tg mice, but not HSC-NOG non-Tg mice, suppressed human T cell proliferation in response to antigen stimulation in in vitro cultures. These results suggest that functional human TAMs can be developed in NOG-hIL-6 Tg mice. This mouse model will contribute to the development of novel cancer immune therapies targeting immunoregulatory/immunosuppressive myeloid cells.


Asunto(s)
Interleucina-6/genética , Células Mieloides/citología , Células Supresoras de Origen Mieloide/citología , Animales , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Ratones Transgénicos , Monocitos/citología , Microambiente Tumoral/inmunología
20.
Cell Mol Immunol ; 15(11): 953-962, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29151581

RESUMEN

Immunodeficient mice engrafted with human peripheral blood cells are promising tools for in vivo analysis of human patient individual immune responses. However, when human peripheral blood mononuclear cells (PBMCs) are transferred into NOG (NOD/Shi-scid, IL-2rgnull) mice, severe graft versus host disease (GVHD) hinders long term detailed analysis. Administration of human PBMCs into newly developed murine MHC class I- and class II-deficient NOG (NOG-dKO; NOG- Iab, B2m-double-knockout) mice showed sufficient engraftment of human immune cells with little sign of GVHD. Immunization with influenza vaccine resulted in an increase in influenza-specific human IgG Ab, indicating induction of antigen-specific B cells in the NOG-dKO mice. Immunization with human dendritic cells pulsed with HLA-A2 restricted cytomegalovirus peptide induced specific cytotoxic T cells, indicating the induction of antigen-specific T cells in the NOG-dKO mice. Adoptive cell therapies (ACTs) using melanoma antigen recognized by T cells (MART-1)-specific TCR-transduced activated T cells showed strong tumor growth inhibition in NOG-dKO mice without any sign of GVHD accompanied by preferential expansion of the transferred MART-1-specific T cells. ACTs using cultured human melanoma infiltrating T cells also showed anti-tumor effects against autologous melanoma cells in NOG-dKO mice, in which changes in human cancer phenotypes by immune intervention, such as increased CD271 expression, could be evaluated. Therefore, NOG-dKO mice are useful tools for more detailed analysis of both the induction and effector phases of T-cell and B-cell responses for a longer period than regular NOG mice.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Leucocitos Mononucleares , Modelos Inmunológicos , Traslado Adoptivo , Animales , Xenoinjertos , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/trasplante , Antígeno MART-1/genética , Antígeno MART-1/inmunología , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA