Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 15(1): 182, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38902833

RESUMO

Human hematopoietic stem cell (HSC)-transferred humanized mice are valuable models for exploring human hematology and immunology. However, sufficient recapitulation of human hematopoiesis in mice requires large quantities of enriched human CD34+ HSCs and total-body irradiation for adequate engraftment. Recently, we generated a NOG mouse strain with a point mutation in the c-kit tyrosine kinase domain (W41 mutant; NOGW mice). In this study, we examined the ability of NOGW mice to reconstitute human hematopoietic cells. Irradiated NOGW mice exhibited high engraftment levels of human CD45+ cells in the peripheral blood, even when only 5,000-10,000 CD34+ HSCs were transferred. Efficient engraftment of human CD45+ cells was also observed in non-irradiated NOGW mice transferred with 20,000-40,000 HSCs. The bone marrow (BM) of NOGW mice exhibited significantly more engrafted human HSCs or progenitor cells (CD34+CD38- or CD34+CD38+ cells) than the BM of NOG mice. Furthermore, we generated a human cytokine (interleukin-3 and granulocyte-macrophage colony-stimulating factor) transgenic NOG-W41 (NOGW-EXL) mouse to achieve multilineage reconstitution with sufficient engraftment of human hematopoietic cells. Non-irradiated NOGW-EXL mice showed significantly higher engraftment levels of human CD45+ and myeloid lineage cells, particularly granulocytes and platelets/megakaryocytes, than non-irradiated NOGW or irradiated NOG-EXL mice after human CD34+ cell transplantation. Serial BM transplantation experiments revealed that NOGW mice exhibited the highest potential for long-term HSC compared with other strains. Consequently, c-kit mutant NOGW-EXL humanized mice represent an advanced model for HSC-transferred humanized mice and hold promise for widespread applications owing to their high versatility.


Assuntos
Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Proteínas Proto-Oncogênicas c-kit , Animais , Humanos , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Camundongos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Transplante de Células-Tronco Hematopoéticas/métodos , Camundongos Transgênicos , Linhagem da Célula , Antígenos CD34/metabolismo , Interleucina-3/metabolismo , Interleucina-3/genética , Mutação
2.
Exp Anim ; 72(3): 402-412, 2023 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-37019665

RESUMO

Humanized mice are widely used to study the human immune system in vivo and investigate therapeutic targets for various human diseases. Immunodeficient NOD/Shi-scid-IL2rγnull (NOG) mice transferred with human hematopoietic stem cells are a useful model for studying human immune systems and analyzing engrafted human immune cells. The gut microbiota plays a significant role in the development and function of immune cells and the maintenance of immune homeostasis; however, there is currently no available animal model that has been reconstituted with human gut microbiota and immune systems in vivo. In this study, we established a new model of CD34+ cell-transferred humanized germ-free NOG mice using an aseptic method. Flow cytometric analysis revealed that the germ-free humanized mice exhibited a lower level of human CD3+ T cells than the SPF humanized mice. Additionally, we found that the human CD3+ T cells slightly increased after transplanting human gut microbiota into the germ-free humanized mice, suggesting that the human microbiota supports T cell proliferation or maintenance in humanized mice colonized by the gut microbiota. Consequently, the dual-humanized mice may be useful for investigating the physiological role of the gut microbiota in human immunity in vivo and for application as a new humanized mouse model in cancer immunology.


Assuntos
Microbioma Gastrointestinal , Sistema Imunitário , Camundongos , Animais , Humanos , Camundongos Endogâmicos NOD , Células-Tronco Hematopoéticas , Modelos Animais de Doenças , Camundongos SCID
3.
Cell Rep ; 41(12): 111841, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36543125

RESUMO

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.


Assuntos
Mercúrio , Neutrófilos , Humanos , Camundongos , Animais , Fator Estimulador de Colônias de Granulócitos , Camundongos Endogâmicos NOD , Hematopoese
4.
Sci Rep ; 12(1): 14907, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-36050438

RESUMO

Chimeric TK-NOG mice with a humanized liver (normal Hu-liver) are a unique animal model for predicting drug metabolism in humans. However, residual mouse hepatocytes occasionally prevent the precise evaluation of human drug metabolism. Herein, we developed a novel humanized liver TK-NOG mouse with a conditional knockout of liver-specific cytochrome P450 oxidoreductase (POR cKO Hu-liver). Immunohistochemical analysis revealed only a few POR-expressing cells around the portal vein in POR cKO mouse livers. NADPH-cytochrome c reductase and cytochrome P450 (P450)-mediated drug oxidation activity in liver microsomes from POR cKO mice was negligible. After the intravenous administration of S-warfarin, high circulating and urinary levels of S-7-hydroxywarfarin (a major human metabolite) were observed in POR cKO Hu-liver mice. Notably, the circulating and urinary levels of S-4'-hydroxywarfarin (a major warfarin metabolite in mice) were much lower in POR cKO Hu-liver mice than in normal Hu-liver mice. POR cKO Hu-liver mice with minimal interference from mouse hepatic P450 oxidation activity are a valuable model for predicting human drug metabolism.


Assuntos
Sistema Enzimático do Citocromo P-450 , Fígado , Varfarina , Animais , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Camundongos Knockout , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Varfarina/metabolismo , Varfarina/farmacologia
5.
J Toxicol Pathol ; 35(1): 19-24, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35221492

RESUMO

The rasH2 mouse was developed as a model for carcinogenicity studies in regulatory science. Its phenotype is stable during high-volume production and over successive generations. To produce rasH2 mice, three strains of mice (C57BL/6J-TgrasH2, C57BL/6J, and BALB/cByJ) were maintained individually. Since the homozygous c-HRAS genotype is lethal, hemizygous transgenic mice were maintained by crossing with inbred C57BL/6J mice. After breeding, male B6-transgenic mice were mated with female BALB/cByJ mice to obtain transgenic mice. Pups that were rasH2-Tg (tg/wt) or rasH2-Wt (wt/wt) were confirmed by genotyping. Frozen embryos were preserved by the Central Institute for Experimental Animals (CIEA) and sent to two facilities, CLEA Japan and Taconic Biosciences, where the mice were produced. Production colonies are created in both facilities and supplied to customers worldwide. To prevent genetic drift, the colonies were renewed for up to 10 generations, and renewals were carried out four times every five years from 2005 to 2021. To ensure the uniformity and maintenance of the phenotype of rasH2 mice, the carcinogen susceptibilities were monitored in every renewal of colonies by CIEA based on a standard protocol of the short-term carcinogenicity study using the positive control compound N-methyl-N-nitrosourea (MNU). Furthermore, simple carcinogenicity monitoring targeting the forestomach, the organ most sensitive to MNU, was performed approximately once a year. Based on the optimally designed production and monitoring systems, the quality of rasH2 mice with reproducibility and stability of carcinogenicity is maintained and supplied globally.

6.
Sci Rep ; 11(1): 11770, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083640

RESUMO

Genetically modified mice are commonly used in biologic, medical, and drug discovery research, but conventional microinjection methods used for genetic modification require extensive training and practical experience. Here we present a fully automated system for microinjection into the pronucleus to facilitate genetic modification. We first developed software that automatically controls the microinjection system hardware. The software permits automatic rotation of the zygote to move the pronucleus to the injection pipette insertion position. We also developed software that recognizes the pronucleus in 3-dimensional coordinates so that the injection pipette can be automatically inserted into the pronucleus, and achieved a 94% insertion rate by linking the 2 pieces of software. Next, we determined the optimal solution injection conditions (30 hPa, 0.8-2.0 s) by examining the survival rate of injected zygotes. Finally, we produced transgenic (traditional DNA injection and piggyBac Transposon system) and knock-in (genomic editing) mice using our newly developed Integrated Automated Embryo Manipulation System (IAEMS). We propose that the IAEMS will simplify highly reproducible pronuclear stage zygote microinjection procedures.


Assuntos
Pesquisas com Embriões , Embrião de Mamíferos , Engenharia Genética/métodos , Camundongos Transgênicos , Animais , Automação , Biologia Computacional/métodos , Genômica/métodos , Camundongos , Microinjeções/métodos , Software , Fluxo de Trabalho
7.
Nat Biomed Eng ; 5(5): 429-440, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34002062

RESUMO

Avoiding the immune rejection of transplanted T cells is central to the success of allogeneic cancer immunotherapies. One solution to protecting T-cell grafts from immune rejection involves the deletion of allogeneic factors and of factors that activate cytotoxic immune cells. Here we report the generation of hypoimmunogenic cancer-antigen-specific T cells derived from induced pluripotent stem cells (iPSCs) lacking ß2-microglobulin, the class-II major histocompatibility complex (MHC) transactivator and the natural killer (NK) cell-ligand poliovirus receptor CD155, and expressing single-chain MHC class-I antigen E. In mouse models of CD20-expressing leukaemia or lymphoma, differentiated T cells expressing a CD20 chimeric antigen receptor largely escaped recognition by NKG2A+ and DNAM-1+ NK cells and by CD8 and CD4 T cells in the allogeneic recipients while maintaining anti-tumour potency. Hypoimmunogenic iPSC-derived T cells may contribute to the creation of off-the-shelf T cell immunotherapies.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Leucemia/terapia , Linfoma/terapia , Receptores Virais/genética , Linfócitos T/transplante , Microglobulina beta-2/genética , Animais , Antígenos de Diferenciação de Linfócitos T/metabolismo , Diferenciação Celular , Linhagem Celular , Técnicas de Inativação de Genes , Engenharia Genética , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Leucemia/imunologia , Linfoma/imunologia , Masculino , Camundongos , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Sci Adv ; 7(10)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658193

RESUMO

Organisms use l-amino acids (l-aa) for most physiological processes. Unlike other organisms, bacteria chiral-convert l-aa to d-configurations as essential components of their cell walls and as signaling molecules in their ecosystems. Mammals recognize microbe-associated molecules to initiate immune responses, but roles of bacterial d-amino acids (d-aa) in mammalian immune systems remain largely unknown. Here, we report that amino acid chirality balanced by bacteria-mammal cross-talk modulates intestinal B cell fate and immunoglobulin A (IgA) production. Bacterial d-aa stimulate M1 macrophages and promote survival of intestinal naïve B cells. Mammalian intestinal d-aa catabolism limits the number of B cells and restricts growth of symbiotic bacteria that activate T cell-dependent IgA class switching of the B cells. Loss of d-aa catabolism results in excessive IgA production and dysbiosis with altered IgA coating on bacteria. Thus, chiral conversion of amino acids is linked to bacterial recognition by mammals to control symbiosis with bacteria.


Assuntos
Aminoácidos , Ecossistema , Aminoácidos/química , Animais , Bactérias/metabolismo , Disbiose , Imunoglobulina A/metabolismo , Intestinos , Mamíferos/metabolismo
9.
Lab Anim ; 55(1): 13-20, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32507045

RESUMO

Severe immunodeficient mice are an essential tool for the examination of the efficacy and safety of new therapeutic technologies as a humanized model. Previously, non-obese diabetic (NOD)/Shi-scid IL2rγnull (NOG) mice were established as immunodeficient mice by combining interleukin-2 receptor-γ chain-knockout mice and NOD/Shi-scid mice. The NOG mice are used frequently in the research of therapeutic monoclonal antibodies and regenerative medicine for human diseases. Establishment of an efficient production system of NOG mice, using optimized reproductive techniques, is required to accelerate research. In this study, we investigated the efficacy of the superovulation technique using equine chorionic gonadotropin (eCG) and inhibin antiserum (IAS) in NOG mice of various ages (4, 8, 12, 24, or 54 weeks). Additionally, we examined the fertilizing and developmental ability of the oocytes through in-vitro fertilization using frozen-thawed sperm, embryo culture and embryo transfer. The results showed that NOG mice produced the highest number of oocytes at 12 weeks old following the co-administration of eCG and IAS (collectively IASe) (70 oocytes/female). IASe was more effective in increasing the number of oocytes v. eCG at all ages. The IASe-derived oocytes demonstrated the ability to fertilize and develop into blastocysts and pups. Finally, we demonstrated that three strains of genetically modified NOG mice were efficiently produced through the optimized reproductive techniques. In summary, we developed an efficient system for the production of immunodeficient mice using 12-week-old, IASe-treated female NOG mice.


Assuntos
Cruzamento/métodos , Gonadotropinas Equinas/química , Soros Imunes/química , Inibinas/química , Camundongos , Superovulação , Animais , Feminino , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID
10.
Exp Anim ; 70(2): 177-184, 2021 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-33239489

RESUMO

To avoid microbial contamination risk, vinyl film isolators are generally used in animal microbiome experiments involving germ-free (GF) mice and/or gnotobiotic (GB) mice. However, it can take several months to gain expertise in operating the isolator competently. Furthermore, sterilization and sterility testing, which are essential for isolator preparation, can take more than 20 days. Hence, we built an experimental rearing environment that combines an individual ventilation cage system and a bioBUBBLE clean room enclosure to easily set up an experimental animal microbiome environment for animal facilities. In this work, a three-step evaluation was conducted. First, we examined whether GF mice can be maintained in this rearing environment without bacterial contamination. Next, we examined whether GF and GB mice can be maintained without cross-contamination in one individual ventilation cage rack. Finally, we tested whether GF mice can be maintained in a biological safety cabinet controlled by negative pressure. In our series of experiments, no microbial contamination occurred over more than 3 months. These results indicated that our rearing system that combines the individual ventilation cage and bioBUBBLE systems can be used not only for experiments with GF mice but also for Biosafety Level 2 experiments that handle bacteria. Our system can mitigate various disadvantages of using vinyl film isolators. In conclusion, we established an experimental method with improved working time and efficiency compared with those of the previous vinyl isolator method.


Assuntos
Criação de Animais Domésticos/instrumentação , Vida Livre de Germes , Abrigo para Animais , Camundongos/microbiologia , Microbiota , Experimentação Animal , Animais , Animais de Laboratório/microbiologia , Camundongos Endogâmicos ICR , Ventilação
11.
Immunol Lett ; 229: 55-61, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33253759

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Transplante de Células , Técnicas de Inativação de Genes , Antígenos de Histocompatibilidade/genética , Subunidade gama Comum de Receptores de Interleucina/deficiência , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Animais , Biomarcadores , Transplante de Células/efeitos adversos , Transplante de Células/métodos , Edição de Genes , Marcação de Genes , Loci Gênicos , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/diagnóstico , Doença Enxerto-Hospedeiro/etiologia , Humanos , Imuno-Histoquímica , Imunofenotipagem , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Modelos Animais , Índice de Gravidade de Doença , Baço/imunologia , Baço/metabolismo
12.
Biochem Biophys Res Commun ; 516(2): 480-485, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31230747

RESUMO

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.


Assuntos
Progressão da Doença , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Interleucina-1beta/metabolismo , Interleucina-23/metabolismo , Transplante de Pele/efeitos adversos , Células Th17/patologia , Animais , Humanos , Interferon gama/metabolismo , Contagem de Linfócitos , Camundongos Transgênicos
13.
Exp Anim ; 68(3): 361-370, 2019 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-30996149

RESUMO

In Japan, it is possible to generate chimeric animals from specified embryos by combining animal blastocysts with human pluripotent stem (PS) cells (animal-human PS chimera). However, the production of animal-human PS chimeras has been restricted because of ethical concerns, such as the development of human-like intelligence and formation of humanized gametes in the animals, owing to the contributions of human PS cells to the brain and reproductive organs. To solve these problems, we established a novel blastocyst complementation technology that does not contribute to the gametes or the brain. First, we established GFP-expressing mouse embryonic stem cells (G-mESCs) in which the Prdm14 and Otx2 genes were knocked out and generated chimeric mice by injecting them into PDX-1-deficient blastocysts. The results showed that the G-mESCs did not contribute to the formation of gametes and the brain. Therefore, in the PDX-1-deficient mice complemented by G-mESCs without the Prdm14 and Otx2 genes, the germline was not transmitted to the next generations. This approach could address concerns regarding the development of both human gametes and a human-like brain upon mouse blastocyst complementation using human stem cells.


Assuntos
Blastocisto/citologia , Diferenciação Celular/fisiologia , Técnicas de Cultura Embrionária/métodos , Transferência Embrionária/métodos , Células-Tronco Embrionárias Murinas/citologia , Animais , Encéfalo/fisiologia , Feminino , Células Germinativas/fisiologia , Japão , Masculino , Camundongos , Camundongos Endogâmicos ICR
14.
RSC Adv ; 9(45): 26429-26434, 2019 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-35530992

RESUMO

Metal complexes with planar structures have been utilized as DNA intercalators that can be inserted into the base pairs of DNA strands, and have potential applications in DNA-targeting drug therapies. When designing the intercalator metal complexes, controlling their interactions with DNA is important, and has been performed by modifying the chemical structure of the metal ligand. Herein, we designed a graft copolymer segment having Pt complexes with bipyridine and poly(ethylene glycol) (p(PEGMA-co-BPyMA-Pt)) as another strategy to control the interaction with DNA via a multivalent effect. The p(PEGMA-co-BPyMA-Pt) increased not only the binding constant as one macromolecule but also the apparent binding constant per intercalator unit compared to the Pt complex with bipyridine (BPy-Pt). Moreover, p(PEGMA-co-BPyMA-Pt) induced a larger change in DNA structure using lower amounts of Pt than BPy-Pt. These observed properties of p(PEGMA-co-BPyMA-Pt) suggest that grafting intercalators on polymer segments is a promising approach for designing novel types of intercalators.

15.
JCI Insight ; 3(21)2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30385714

RESUMO

Asthma is one of the most common immunological diseases and is characterized by airway hyperresponsiveness (AHR), mucus overproduction, and airway eosinophilia. Although mouse models have provided insight into the mechanisms by which type-2 cytokines induce asthmatic airway inflammation, differences between the rodent and human immune systems hamper efforts to improve understanding of human allergic diseases. In this study, we aim to establish a preclinical animal model of asthmatic airway inflammation using humanized IL-3/GM-CSF or IL-3/GM-CSF/IL-5 Tg NOD/Shi-scid-IL2rγnull (NOG) mice and investigate the roles of human type-2 immune responses in the asthmatic mice. Several important characteristics of asthma - such as AHR, goblet cell hyperplasia, T cell infiltration, IL-13 production, and periostin secretion - were induced in IL-3/GM-CSF Tg mice by intratracheally administered human IL-33. In addition to these characteristics, human eosinophilic inflammation was observed in IL-3/GM-CSF/IL-5 Tg mice. The asthmatic mechanisms of the humanized mice were driven by activation of human Th2 and mast cells by IL-33 stimulation. Furthermore, treatment of the humanized mice with an anti-human IL-13 antibody significantly suppressed these characteristics. Therefore, the humanized mice may enhance our understanding of the pathophysiology of allergic disorders and facilitate the preclinical development of new therapeutics for IL-33-mediated type-2 inflammation in asthma.


Assuntos
Asma/imunologia , Inflamação/imunologia , Interleucina-13/administração & dosagem , Interleucina-33/administração & dosagem , Animais , Asma/fisiopatologia , Citocinas/imunologia , Modelos Animais de Doenças , Eosinófilos/imunologia , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-13/imunologia , Interleucina-13/farmacologia , Interleucina-33/imunologia , Interleucina-33/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/fisiopatologia
16.
Stem Cell Reports ; 10(3): 1059-1074, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29503091

RESUMO

Increasing demand for clinical retinal degeneration therapies featuring human ESC/iPSC-derived retinal tissue and cells warrants proof-of-concept studies. Here, we established two mouse models of end-stage retinal degeneration with immunodeficiency, NOG-rd1-2J and NOG-rd10, and characterized disease progress and immunodeficient status. We also transplanted human ESC-derived retinal sheets into NOG-rd1-2J and confirmed their long-term survival and maturation of the structured graft photoreceptor layer, without rejection or tumorigenesis. We recorded light responses from the host ganglion cells using a multi-electrode array system; this result was consistent with whole-mount immunostaining suggestive of host-graft synapse formation at the responding sites. This study demonstrates an application of our mouse models and provides a proof of concept for the clinical use of human ESC-derived retinal sheets.


Assuntos
Células-Tronco Embrionárias/patologia , Retina/patologia , Degeneração Retiniana/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Células Fotorreceptoras/patologia , Transplante de Células-Tronco/métodos
17.
J Vet Med Sci ; 80(4): 662-666, 2018 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-29445073

RESUMO

Expression of peroxisome proliferator-activated receptor (PPAR) α was investigated in adiponectin knockout mice to elucidate the relationship between PPARα and adiponectin deficiency-induced diabetes. Adiponectin knockout (Adp-/-) mice were generated by gene targeting. Glucose tolerance test (GTT), insulin tolerance test (ITT), and organ sampling were performed in Adp-/- mice at the age of 10 weeks. PPARα, insulin, triglyceride, free fatty acid (FFA), and tumor necrosis factor α (TNFα) were analyzed from the sampled organs. Adp-/- mice showed impaired glucose tolerance and insulin resistance. Additionally, PPARα levels were decreased and plasma concentration of triglyceride, FFA and TNFα were increased. These data may indicate that insulin resistance in Adp-/- mice is likely caused by an increase in concentrations of TNFα and FFA via downregulation of PPARα.


Assuntos
Adiponectina/genética , Diabetes Mellitus/metabolismo , Regulação para Baixo/fisiologia , Ácidos Graxos não Esterificados/metabolismo , PPAR alfa/metabolismo , Animais , Diabetes Mellitus/genética , Regulação da Expressão Gênica/fisiologia , Intolerância à Glucose , Insulina/sangue , Camundongos , Camundongos Knockout , PPAR alfa/genética , Fator de Necrose Tumoral alfa
18.
Exp Anim ; 66(4): 425-435, 2017 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-28679969

RESUMO

Severely immunodeficient NOD/Shi-scid, IL-2Rγnull (NOG) mice provide an in vivo model for human cell/tissue transplantation studies. NOG mice were established by combining interleukin-2 receptor-γ chain knockout mice and NOD/Shi-scid mice. They exhibit a high incidence of thymic lymphomas and immunoglobulin (Ig) leakiness. In this study, we assessed the incidence of malignant lymphomas and the occurrence of leakiness in 2,184 non-experimental NOG retired breeder mice aged 16-40 weeks. We established that the total incidence of lymphomas was only 0.60% (13/2,184). Most lymphomas (10/13) occurred in female mice by the age of around 25 weeks. No mice developed Ig leakiness. All lymphomas were derived from the thymus, and consisted mainly of CD3-positive and CD45R-negative lymphoblastic-like cells. Therefore, based on the absence of Ig leakiness and a very low incidence of lymphomas, including thymic lymphomas, NOG mice may be useful in regeneration medicine for xenotransplantation of human embryonic stem (ES) cells or induced pluripotent stem (iPS) cells, and in transplantation experiments involving tumor cells.


Assuntos
Linfoma , Neoplasias do Timo , Animais , Complexo CD3 , Células-Tronco Embrionárias/transplante , Humanos , Incidência , Células-Tronco Pluripotentes Induzidas/transplante , Subunidade gama Comum de Receptores de Interleucina/genética , Antígenos Comuns de Leucócito , Linfoma/epidemiologia , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Modelos Animais , Transplante de Neoplasias , Neoplasias do Timo/epidemiologia , Transplante Heterólogo
19.
Biochem Biophys Res Commun ; 478(3): 1254-60, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27545600

RESUMO

Most in vivo studies on the conversion to insulin-producing cells with AAV carrying PDX1 gene are performed in rodents. However, there is little information regarding Adeno-associated virus (AAV) carrying PDX1 gene transduced to human liver in vivo because accidental death caused by unpredicted factors cannot be denied, such as the hypoglycemic agent troglitazone with hepatic failure. Here we aim to confirm insulin secretion from human liver transduced with AAV carrying PDX1 gene in vivo and any secondary effect using a humanized liver mouse. As the results, AAV2-PG succeeded to improve the hyperglycemia of STZ-induced diabetic humanized liver mice. Then, the analysis of humanized liver mice revealed that the AAV2-PG was more transducible to humanized liver area than to mouse liver area. In conclusion, the humanized liver mouse model could be used to examine AAV transduction of human hepatocytes in vivo and better predict clinical transduction efficiency than nonhumanized mice.


Assuntos
Dependovirus/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/terapia , Terapia Genética , Animais , Diabetes Mellitus Experimental/complicações , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Hiperglicemia/complicações , Hiperglicemia/terapia , Insulina/metabolismo , Camundongos , Camundongos Transgênicos , Transativadores/metabolismo , Transdução Genética
20.
Lab Anim Res ; 31(2): 93-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26155204

RESUMO

We developed pancreatic and duodenal homeobox1 (Pdx1) knockout mice to improve a compensatory hyperinsulinemia, which was induced by hyperplasia in the ß cells or Langerhans' islands, as the diabetic model mice. For targeting of Pdx1 gene by homologous recombination, ES cells derived from a 129(+Ter) /SvJcl×C57BL/6JJcl hybrid mouse were electroporated and subjected to positive-negative selection with hygromycin B and ganciclovir. As these results, one of the three chimeric mice succeeded to produce the next or F1 generation. Then, the mouse fetuses were extracted from the mother's uterus and analyzed immunohistologically for the existence of a pancreas. The fetuses were analyzed at embryonic day 14.5 (E14.5) because Pdx1 knockout could not alive after birth in this study. Immunohistochemical staining revealed that 10 fetuses out of 26 did not have any PDX1 positive primordium of the pancreas and that the PDX1 expresses in both the interior and exterior regions of intestine. In particular, one the exterior of the intestine PDX1 was expressed in glands that would be expected to form the pancreas. The result of PCR genotyping with extracted DNA from the paraffin sections showed existence of 10 Pdx1-knockout mice and corresponded to results of immunostaining. Thus, we succeeded to establish a Pdx1-knockout (Pdx1 (-/-)) mice.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA