Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 3242, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217706

RESUMO

Blastocyst complementation is an intriguing way of generating humanized animals for organ preparation in regenerative medicine and establishing novel models for drug development. Confirming that complemented organs and cells work normally in chimeric animals is critical to demonstrating the feasibility of blastocyst complementation. Here, we generated thymus-complemented chimeric mice, assessed the efficacy of anti-PD-L1 antibody in tumor-bearing chimeric mice, and then investigated T-cell function. Thymus-complemented chimeric mice were generated by injecting C57BL/6 (B6) embryonic stem cells into Foxn1nu/nu morulae or blastocysts. Flow cytometry data showed that the chimeric mouse thymic epithelial cells (TECs) were derived from the B6 cells. T cells appeared outside the thymi. Single-cell RNA-sequencing analysis revealed that the TEC gene-expression profile was comparable to that in B6 mice. Splenic T cells of chimeric mice responded very well to anti-CD3 stimulation in vitro; CD4+ and CD8+ T cells proliferated and produced IFNγ, IL-2, and granzyme B, as in B6 mice. Anti-PD-L1 antibody treatment inhibited MC38 tumor growth in chimeric mice. Moreover, in the chimeras, anti-PD-L1 antibody restored T-cell activation by significantly decreasing PD-1 expression on T cells and increasing IFNγ-producing T cells in the draining lymph nodes and tumors. T cells produced by complemented thymi thus functioned normally in vitro and in vivo. To successfully generate humanized animals by blastocyst complementation, both verification of the function and gene expression profiling of complemented organs/cells in interspecific chimeras will be important in the near future.


Assuntos
Blastocisto , Linfócitos T CD8-Positivos , Animais , Blastocisto/metabolismo , Quimera/genética , Células-Tronco Embrionárias , Camundongos , Camundongos Endogâmicos C57BL
2.
Arthritis Rheumatol ; 71(2): 222-231, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30079992

RESUMO

OBJECTIVE: To elucidate the role of the fractalkine (FKN)/CX3 CR1 pathway in joint destruction in rheumatoid arthritis. METHODS: We examined the effect of treatment with anti-mouse FKN (anti-mFKN) monoclonal antibody (mAb) on joint destruction and the migration of osteoclast precursors (OCPs) into the joint, using the collagen-induced arthritis (CIA) model. DBA/1 mice were immunized with bovine type II collagen to induce arthritis, and then treated with anti-mFKN mAb. Disease severity was monitored by arthritis score, and joint destruction was evaluated by soft x-ray and histologic analyses. Plasma levels of joint destruction markers were assessed by enzyme-linked immunosorbent assay. FKN expression on endothelial cells was detected by immunohistochemistry. Bone marrow-derived OCPs were labeled with fluorescein and transferred to mice with CIA, and the migration of the OCPs to the joints was then analyzed. RESULTS: Both prophylactic and therapeutic treatment with anti-mFKN mAb significantly decreased the arthritis and soft x-ray scores. Plasma levels of cartilage oligomeric matrix protein and matrix metalloproteinase 3 decreased after treatment with anti-mFKN mAb. Histologic analysis revealed that anti-mFKN mAb inhibited synovitis, pannus formation, and cartilage destruction, as well as suppressed bone damage, with a marked reduction in the number of tartrate-resistant acid phosphatase-positive osteoclasts. Anti-mFKN mAb strongly inhibited the migration of bone marrow-derived OCPs into the affected synovium. CONCLUSION: Anti-mFKN mAb notably ameliorates arthritis and joint destruction in the CIA model, as well as inhibits migration of OCPs into the synovium. These results suggest that inhibition of the FKN/CX3 CR1 pathway could be a novel strategy for treatment of both synovitis and joint destruction in rheumatoid arthritis.


Assuntos
Anticorpos Monoclonais/farmacologia , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Receptor 1 de Quimiocina CX3C/imunologia , Movimento Celular/efeitos dos fármacos , Quimiocina CX3CL1/antagonistas & inibidores , Osteoclastos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Proteína de Matriz Oligomérica de Cartilagem/efeitos dos fármacos , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Quimiocina CX3CL1/imunologia , Metaloproteinase 3 da Matriz/efeitos dos fármacos , Metaloproteinase 3 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Osteoclastos/metabolismo , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Sinovite/patologia , Fosfatase Ácida Resistente a Tartarato/metabolismo
3.
Transgenic Res ; 27(2): 193-201, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29546522

RESUMO

DNA site-specific recombination by Cre/loxP is a powerful tool for gene manipulation in experimental animals. VCre/VloxP and SCre/SloxP are novel site-specific recombination systems, consisting of a recombinase and its specific recognition sequences, which function in a manner similar to Cre/loxP. Previous reports using Escherichia coli and Oryzias latipes demonstrated the existence of stringent specificity between each recombinase and its target sites; VCre/VloxP, SCre/SloxP, and Cre/loxP have no cross-reactivity with each other. In this study, we established four novel knock-in (KI) mouse strains in which VloxP-EGFP, SloxP-tdTomato, CAG-VCre, and CAG-SCre genes were inserted into the ROSA26 locus. VloxP-EGFP and SloxP-tdTomato KI mice were reporter mice carrying EGFP or tdTomato genes posterior to the stop codon, which was floxed by VloxP or SloxP fragments, respectively. CAG-VCre and CAG-SCre KI mice carried VCre or SCre genes that were expressed ubiquitously. These two reporter mice were crossed with three different deleter mice, CAG-VCre KI, CAG-SCre KI, and Cre-expressing transgenic mice. Through these matings, we found that VCre/VloxP and SCre/SloxP systems were functional in mice similar to Cre/loxP, and that the recombinases showed tight specificity for their recognition sequences. Our results suggest that these novel recombination systems allow highly sophisticated genome manipulations and will be useful for tracing the fates of multiple cell lineages or elucidating complex spatiotemporal regulations of gene expression.


Assuntos
Deleção de Genes , Genes Reporter/genética , Integrases/genética , Recombinação Genética , Animais , Linhagem da Célula/genética , DNA Nucleotidiltransferases/genética , Regulação da Expressão Gênica/genética , Técnicas de Introdução de Genes , Genoma/genética , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos
4.
Sci Rep ; 7(1): 17230, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29222435

RESUMO

We generated a novel mouse strain expressing transgenic human interleukin-15 (IL-15) using the severe immunodeficient NOD/Shi-scid-IL-2Rγ null (NOG) mouse genetic background (NOG-IL-15 Tg). Human natural killer (NK) cells, purified from the peripheral blood (hu-PB-NK) of normal healthy donors, proliferated when transferred into NOG-IL-15 Tg mice. In addition, the cell number increased, and the hu-PB-NK cells persisted for 3 months without signs of xenogeneic graft versus host diseases (xGVHD). These in vivo-expanded hu-PB-NK cells maintained the original expression patterns of various surface antigens, including NK receptors and killer cell immunoglobulin-like receptor (KIR) molecules. They also contained significant amounts of granzyme A and perforin. Inoculation of K562 leukemia cells into hu-PB-NK-transplanted NOG-IL-15 Tg mice resulted in significant suppression of tumor growth compared with non-transplanted mice. Furthermore, NOG-IL-15 Tg mice allowed for engraftment of in vitro-expanded NK cells prepared for clinical cell therapy. These cells exerted antibody-dependent cell-mediated cytotoxicity (ADCC) on Her2-positive gastric cancer cells in the presence of therapeutic anti-Her2 antibody, and subsequently suppressed tumor growth. Our results collectively suggest that the NOG-IL-15 Tg mice are a useful model for studying human NK biology and evaluating human NK cell-mediated in vivo cytotoxicity.


Assuntos
Interleucina-15/sangue , Interleucina-15/genética , Células Matadoras Naturais/citologia , Animais , Humanos , Camundongos , Camundongos Transgênicos
5.
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.

6.
J Immunol ; 194(7): 3513-25, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25712215

RESUMO

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.


Assuntos
Interleucina-2/biossíntese , Interleucina-2/genética , Células Matadoras Naturais/imunologia , Camundongos Transgênicos , Animais , Citotoxicidade Celular Dependente de Anticorpos , Antígenos de Superfície/metabolismo , Diferenciação Celular , Citotoxicidade Imunológica , Modelos Animais de Doenças , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunofenotipagem , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Fenótipo , Receptores KIR/genética , Receptores KIR/metabolismo
7.
Cryobiology ; 70(2): 150-5, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25661709

RESUMO

Strain preservation of experimental animals is crucial for experimental reproducibility. Maintaining complete animal strains, however, is costly and there is a risk for genetic mutations as well as complete loss due to disasters or illness. Therefore, the development of effective vitrification techniques for cryopreservation of multiple experimental animal strains is important. We examined whether a vitrification method using cryoprotectant solutions, P10 and PEPeS, is suitable for preservation of multiple inbred and outbred mouse strains. First, we investigated whether our vitrification method using cryoprotectant solutions was suitable for two-cell stage mouse embryos. In vitro development of embryos exposed to the cryoprotectant solutions was similar to that of fresh controls. Further, the survival rate of the vitrified embryos was extremely high (98.1%). Next, we collected and vitrified two-cell stage embryos of 14 mouse strains. The average number of embryos obtained from one female was 7.3-33.3. The survival rate of vitrified embryos ranged from 92.8% to 99.1%, with no significant differences among mouse strains. In vivo development did not differ significantly between fresh controls and vitrified embryos of each strain. For strain preservation using cryopreserved embryos, two offspring for inbred lines and one offspring for outbred lines must be produced from two-cell stage embryos collected from one female. The expected number of surviving fetuses obtained from embryos collected from one female of either the inbred or outbred strains ranged from 2.9 to 19.5. The findings of the present study indicated that this vitrification method is suitable for strain preservation of multiple mouse strains.


Assuntos
Criopreservação/métodos , Embrião de Mamíferos/fisiologia , Preservação Biológica/métodos , Animais , Crioprotetores/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos , Reprodutibilidade dos Testes , Taxa de Sobrevida , Vitrificação
8.
Exp Anim ; 63(3): 321-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25077762

RESUMO

We have developed NOD-Rag2(null) IL-2Rγ(null) (NR2G) mice similar to NOD-scidIL-2Rγ(null) (NOG) mice that are known as an excellent host to generate humanized mice. To evaluate the usefulness of NR2G mice as a host for humanized mice, the engraftment rates and differentiation of human cells after human hematopoietic stem cell (HSC) transplantation were compared among NR2G, NOG, and NOD-scid mice. For this purpose, the appropriate irradiation doses to expand the niche for human stem cells in the bone marrow were first determined. As a result, 8 and 2.5 Gy in adult, and 4 and 1 Gy in newborn NR2G and NOG mice, respectively, were found to be appropriate. Next, 5 × 10(4) human umbilical cord blood CD34(+) cells were intravenously inoculated into irradiated adult or newborn of the immunodeficient mice. These HSC transplantation experiments demonstrated that both NR2G and NOG mice showed high engraftment rates compared with NOD-scid mice, although NOG mice showed a slightly higher engraftment rate than that for NR2G mice. However, no difference was found in the human cell populations differentiated from HSCs between NR2G and NOG mice. The HSC transplantation experiments to adults and newborns of two immunodeficient mice also revealed that the HSC transplantation into newborn mice resulted in higher engraftment rate than those into adults. These results showed that NR2G mice could be used as an alternative host to NOG mice to generate humanized mice.


Assuntos
Modelos Animais de Doenças , Animais , Animais Recém-Nascidos , Antígenos CD34 , Proteínas de Ligação a DNA/deficiência , Sangue Fetal/transplante , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Doses de Radiação , Receptores de Interleucina-2/deficiência , Irradiação Corporal Total
9.
Cryobiology ; 68(1): 147-51, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24462541

RESUMO

The present study was performed to develop a suitable cryoprotectant solution for cryopreservation of rat two-cell stage embryos. First, we examined the cell permeability of several cryoprotectants; propylene glycol had the fastest permeability compared to dimethyl sulfoxide, ethylene glycol, and glycerol. Embryos were then exposed to a solution containing propylene glycol to evaluate its effects on fetal development. As the development was similar to that of fresh embryos, P10 (10% v/v propylene glycol in PB1) was used as a pretreatment solution. Next, the effects of the vitrification solution components (sucrose, propylene glycol, ethylene glycol, and Percoll) were examined by observing the vitrification status; 10% v/v propylene glycol, 30% v/v ethylene glycol, 0.3 mol sucrose, and 20% v/v Percoll in PB1 (PEPeS) was the minimum essential concentration for effective vitrification without the formation of ice crystals or freeze fractures. A new vitrification method using P10 and PEPeS was tested using rat embryos. The survival rate of vitrified embryos after exposure to P10 for 120, 300, or 600 s ranged from 95.9% to 98.3%. The fetal developmental rate ranged from 57.7% to 65.2%, which was not significantly different from that of fresh embryos. The experimental results indicated that vitrification using a combination of P10 and PEPeS was suitable for cryopreservation of rat early stage embryos.


Assuntos
Criopreservação , Crioprotetores/farmacologia , Embrião de Mamíferos/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Vitrificação , Animais , Permeabilidade da Membrana Celular , Sobrevivência Celular/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/fisiologia , Etilenoglicol/farmacologia , Feminino , Glicerol/farmacologia , Masculino , Concentração Osmolar , Povidona/farmacologia , Propilenoglicol/farmacologia , Ratos , Dióxido de Silício/farmacologia , Sacarose/farmacologia
10.
J Immunol ; 191(6): 2890-9, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23956433

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Hipersensibilidade/imunologia , Interleucina-3/imunologia , Animais , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Transplante de Células-Tronco Hematopoéticas , Humanos , Imuno-Histoquímica , Interleucina-3/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
11.
J Immunol ; 189(9): 4313-20, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23018460

RESUMO

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.


Assuntos
Antígeno CD11c , Sobrevivência de Enxerto/imunologia , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade beta de Receptor de Interleucina-2/deficiência , Antígenos Comuns de Leucócito/deficiência , Transplante Heterólogo/métodos , Animais , Antígeno CD11c/biossíntese , Antígeno CD11c/genética , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/genética , Humanos , Hospedeiro Imunocomprometido , Subunidade gama Comum de Receptores de Interleucina/biossíntese , Subunidade beta de Receptor de Interleucina-2/biossíntese , Subunidade beta de Receptor de Interleucina-2/genética , Antígenos Comuns de Leucócito/biossíntese , Antígenos Comuns de Leucócito/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Baço/imunologia , Baço/metabolismo , Baço/transplante
12.
Exp Hematol ; 40(11): 953-963.e3, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22771497

RESUMO

NOD/Shi-scid IL2rγnull (NOG) mice with severe immunodeficiency are excellent recipients to generate "humanized" mice by the transplantation of human CD34(+) hematopoietic stem cells (HSCs). In this study, we developed NOG mice carrying a human Delta-like1 (DLL1) gene, which is a ligand of the Notch receptor and is known to be important in HSC maintenance and self-renewal. We also analyzed the effect of DLL1 signaling on human hematopoiesis and HSC maintenance using humanized DLL1 transgenic NOG mice. To develop DLL1 transgenic NOG (NOG-D1-Tg) mice, a transgenic vector consisting of a human DLL1 complementary DNA fragment placed downstream of the α1(I) collagen (Col1a1) promoter for expression specifically in osteoblasts was constructed. Human CD34(+) HSCs were transplanted into NOG-D1-Tg mice, and differentiation of lymphoid or myeloid lineage cells from human HSCs and maintenance of HSCs in bone marrow were analyzed. Severe osteosclerosis accompanied by increased bone mass and a decreased number of bone marrow cells were observed in NOG-D1-Tg mice. After human HSC transplantation, development of human B lymphocytes, but not T lymphocytes, was significantly suppressed in both bone marrow and the periphery of NOG-D1-Tg mice. Contrary to the initial expectation, retention of human CD34(+) HSCs was inhibited in the bone marrow of NOG-D1-Tg mice. In conclusion, our data suggest that the development of human B lymphocytes and HSC maintenance in osteosclerotic bone may be suppressed by introducing DLL1. These unique humanized mice with sclerotic bone reconstituted by human HSCs are useful models of hematopoiesis in patients with osteosclerosis, such as osteopetrosis, and for investigation of osteogenesis via Notch signaling.


Assuntos
Proteínas de Transporte/genética , Hematopoese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Osteoblastos/patologia , Osteosclerose/patologia , Animais , Proteínas de Ligação ao Cálcio , Camundongos , Camundongos Transgênicos
13.
Exp Anim ; 59(4): 521-4, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20660999

RESUMO

The effects of progesterone (P4) used in physiological studies and in delayed parturition in reproductive engineering were examined. A dose of 0.25, 0.5, 1, or 2 mg of P4 was repeatedly administered to Jcl:MCH(ICR) mice on days 17 and 18 of gestation, and plasma concentrations of P4 were investigated. The P4 concentrations in mothers and fetuses after administration of exogenous P4 were no differences between doses of 1 and 2 mg. Jcl:MCH(ICR) mothers administered a P4 dose of 1 mg did not give birth. Therefore, we consider 2 mg of P4 is an overdose and that it is evident that a dose of 1 mg P4 is sufficient to induce delayed parturition.


Assuntos
Parto/efeitos dos fármacos , Progesterona/farmacologia , Progestinas/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Feto/metabolismo , Idade Gestacional , Trabalho de Parto/sangue , Trabalho de Parto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Parto/sangue , Gravidez , Complicações na Gravidez/sangue , Complicações na Gravidez/induzido quimicamente , Progesterona/sangue , Progestinas/sangue
14.
Transplantation ; 87(11): 1654-8, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19502956

RESUMO

BACKGROUND: Several animal models for xenogenic (xeno) graft versus host disease (GVHD) have been developed in immunodeficient mice, such as C.B-17-scid and nonobese diabetes (NOD)/severe combined immunodeficiency (SCID), by human peripheral blood mononuclear cell (hPBMC) transplantation. However, these models pose problems because they require sublethal total body irradiation of the mice and a large number of hPBMCs to induce GVHD, and the timing of onset of GVHD is also unstable. The aim of this study is to establish improved murine models of xeno-GVHD using novel immunodeficient NOD/Shi-scid IL2r gamma null (NOG) mice. METHODS: In three strains of immunodeficient mice, NOG, BALB/cA-RAG2 IL2r gamma null, and NOD/SCID mice, GVHD was induced by transplantation of hPBMCs with or without total body irradiation, and the GVHD symptoms in these strains were compared. RESULTS: After intravenous transplantation of hPBMCs, NOG mice showed early onset of GVHD symptoms and a small number of hPBMCs (2.5 x 10(6)) was sufficient to induce GVHD when compared with BALB/cA-RAG2 null IL2r gamma null and NOD/SCID mice. In addition, total body irradiation was not always necessary in the present model. CONCLUSIONS: These results indicate that our model using the NOG mouse is a useful tool to investigate GVHD and to develop effective drugs for GVHD.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Leucócitos Mononucleares/transplante , Transplante Heterólogo/imunologia , Animais , Citometria de Fluxo , Doença Enxerto-Hospedeiro/mortalidade , Doença Enxerto-Hospedeiro/patologia , Humanos , Imuno-Histoquímica , Subunidade gama Comum de Receptores de Interleucina/deficiência , Rim/patologia , Fígado/patologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Irradiação Corporal Total
15.
Exp Anim ; 57(4): 407-11, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18633164

RESUMO

Efficient reproduction using natural mating and reproduction technology [in vitro fertilization (IVF) and embryo transfer (ET)] was investigated in IRS2 deficient mice with C57BL/6JJcl genetic background (Irs2(-/-) mice) as a typical type 2 diabetes model. From the results using various combinations of Irs2(-/-) and Irs2(-/+) mice, the combination of female Irs2(-/+) x male Irs2(-/-) was found to be more efficient than other combinations. In applications of reproduction technology using IVF and ET, the combination of female Irs2(-/+) x male Irs2(-/-) involves the possibility of Irs2(-/-) production by repeats using female Irs2(-/+) mice. However, reproductive continuity using this combination is difficult because of dependence on human technique and the cost of ET. Therefore, we concluded that Irs2(-/-) mice should be produced by embryo transfer using Irs2(-/-) mice from a colony consisting of female Irs2(-/+) x male Irs2(-/-).


Assuntos
Transferência Embrionária/métodos , Fertilização in vitro/métodos , Camundongos Endogâmicos C57BL/genética , Animais , Diabetes Mellitus Tipo 2/genética , Feminino , Masculino , Camundongos
16.
Exp Anim ; 54(2): 137-41, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15897622

RESUMO

In most strains of rats, the effects of treatment for the induction of superovulation show major strain differences and are strongly influenced by the stage of the estrous cycle. This study demonstrated, however, that superovulation was easily induced in Wistar strain Brl Han:WIST@Jcl(GALAS) rats by PMSG and hCG administration. To confirm the effects of such treatment, we studied age differences in egg collection efficiency. After superovulation was induced by intraperitoneal administration of 150 IU/kg PMSG and 75 IU/kg hCG given 48 h apart, the mean numbers of oocytes obtained from rats at 4, 8, 12, 20 and 28 weeks of age were 38.9, 33.5, 46.1, 26.9 and 21.3, respectively. No differences caused by the estrous stage at the PMSG administration were observed. In an embryo transfer experiment, fertilized eggs obtained from superovulated rats at each week of age showed equivalent viability until full-term to those from untreated rats. These results suggest that estrous stage-independent superovulation is effective not only in the pre-pubertal stage but also in adult rats.


Assuntos
Gonadotropina Coriônica/farmacologia , Gonadotropinas Equinas/farmacologia , Superovulação/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Gonadotropina Coriônica/administração & dosagem , Transferência Embrionária , Ciclo Estral/fisiologia , Feminino , Gonadotropinas Equinas/administração & dosagem , Injeções Intraperitoneais , Masculino , Oócitos , Óvulo , Ratos , Ratos Endogâmicos , Estimulação Química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...