Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 213
Filtrar
1.
Biomed Res Int ; 2021: 8851986, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34805408

RESUMO

OBJECTIVE: Natural killer (NK) cell-deficient mice are useful models in biomedical research. NOD/SCID mice have been used as a model of this type in research. However, the actual status of NK cells in NOD/SCID mice and CB17/SCID mice in comparison with that in BALB/c mice has not been sufficiently evaluated. METHODS: Splenocytes from naïve or poly(I:C)-treated mice were isolated for phenotyping and analysis of cytotoxicity-related molecules and inhibitory receptors; for cytotoxicity assay, purified NK cells were also used. RESULTS: The proportion of splenic NK cells did not differ significantly between NOD/SCID and CB17/SCID mice. The perforin levels in NK cells were similar between the poly(I:C)-treated CB17/SCID and NOD/SCID mice, while the granzyme B and NKG2A/C/E levels in NK cells from NOD/SCID mice were significantly lower than those from CB17/SCID mice. Moreover, the NKG2D and Ly49A levels in NK cells from NOD/SCID mice were higher than those from CB17/SCID. The splenocytes from CB17/SCID mice showed higher cytotoxicity than those from NOD/SCID mice, while the cytotoxicity of purified NK cells basically did not differ between the two strains. After in vitro stimulation with cytokines, the splenocytes from CB17/SCID mice showed higher IFN-γ production than those from NOD/SCID mice; however, NK cells did not. CONCLUSION: There was no significant difference in the proportion of splenic NK cells between CB17/SCID and NOD/SCID mice, and the function of NK cells was only partially compromised in NOD/SCID mice. Caution should be taken when considering the use of NOD/SCID mice as an NK-deficient model.


Assuntos
Células Matadoras Naturais/imunologia , Camundongos Endogâmicos NOD/imunologia , Camundongos SCID/imunologia , Animais , Antígenos Ly/metabolismo , Citocinas/biossíntese , Citotoxicidade Imunológica , Imunidade Inata , Técnicas In Vitro , Células Matadoras Naturais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Animais , Modelos Imunológicos , Subfamília B de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptores Semelhantes a Lectina de Células NK/metabolismo , Especificidade da Espécie
2.
Int Immunopharmacol ; 101(Pt A): 108206, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34626875

RESUMO

Though it has long been thought that the immune system is implicated in the pathophysiology of heat stroke, the underlying mechanisms are still poorly understood. As it has been reported in the literature that lymphocyte disturbance occurs in heat stroke patients or animals, we attempted to seek experimental evidence to define the role of lymphocytes in the pathophysiology of heat stroke. In our study, we used male Balb/c mice to establish a passive heat stroke model. We found that lymphocyte-deficient Severe combined immunodeficient (SCID) mice exposed to heat stress exhibited exacerbated heat stroke severity, which could be indicated by increased rates of mortality and serum levels of inflammatory cytokines compared to wildtype control mice. We further showed, through the depletion of T lymphocytes in wildtype mice and the transfer of wildtype lymphocytes into SCID mice, respectively, that T lymphocytes were both necessary and sufficient to alleviate the severity of heat stroke by inhibiting the early inflammatory response. Moreover, we found that the severity of heat injuries in heat-stressed wildtype mice showed great inter-individual variability, and the early number of T lymphocytes could be negatively associated with the severity of heat stroke. Our results suggest that lack of T lymphocytes could exacerbate the severity of heat stroke by augmenting inflammatory response, and the early circulating T lymphocytes may serve as a potential biomarker for the diagnosis of heat stroke.


Assuntos
Golpe de Calor/imunologia , Inflamação/imunologia , Linfócitos/fisiologia , Animais , Citocinas/metabolismo , Citometria de Fluxo , Golpe de Calor/patologia , Inflamação/patologia , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID/imunologia , Gravidade do Paciente , Linfócitos T/imunologia , Linfócitos T/fisiologia
3.
PLoS One ; 15(9): e0239231, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32997686

RESUMO

It is controversially discussed whether immune-deficient mice experience severity in the absence of infection. Because a comprehensive analysis of the well-being of immune-deficient mice under specific pathogen free conditions is missing, we used a multi-parametric test analyzing, corticosterone, weight, nest building and facial expression over a period of 9 month to determine the well-being of two immune-deficient mouse lines (recombination activating gene 2- and interferon gamma receptor-deficient mice). We do not find evidence for severity when comparing immune-deficient mice to their heterozygous immune-competent littermates. Our data challenge the assumption that immune-deficiency per se regardless of housing conditions causes severity. Based on our study we propose to use objective non-invasive parameters determined by laboratory animal science for decisions concerning severity of immune-deficient mice.


Assuntos
Corticosterona/genética , Proteínas de Ligação a DNA/genética , Interferon gama/genética , Camundongos SCID/genética , Animais , Linfócitos B/imunologia , Corticosterona/imunologia , Humanos , Infecções/genética , Infecções/imunologia , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos SCID/imunologia , Dor/genética , Dor/patologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais/genética , Linfócitos T/imunologia , Testosterona/genética
4.
Exp Anim ; 68(4): 471-482, 2019 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-31118345

RESUMO

Biological background data up to 11 weeks of age and tumorigenic susceptibility to xenotransplantation with HeLa cells were compared between severely immuno-deficient NOG and NSG mice. The body weight was lower in NOG mice than in NSG mice. Severe depletion of peripheral blood lymphocytes and lymphoid hypoplasia that are well-known characteristics of these mice were equally observed. No lymphoproliferative lesions developed in any mouse of either strain. The occurrence of ectopic exocrine gland and cyst was a common finding in the thymus of both strains. In addition, minimal spongiotic change was observed in the medulla oblongata and spinal cord in both strains, and its incidence in female NOG mice was a little higher than that in NSG mice. In the adrenal, subcapsular cell hyperplasia that is known as an age-related change in non-genetically modified mice developed earlier and its incidence was higher in NSG mice than in NOG mice. The development of female genital organs of NOG mice was slightly retarded in comparison with that of NSG mice. To evaluate tumorigenic susceptibility to xenotransplantation, female mice were implanted in the dorsal subcutis with 1×103 to 1×106 cells/head of HeLa cells, and were checked up to 16 weeks after implantation. As a result, there was no significant strain difference on tumor formation rate and tumor volume. In conclusion, the present study clearly demonstrated that NOG and NSG mice showed no distinct strain differences in either biological features or biological disadvantages.


Assuntos
Carcinogênese/imunologia , Camundongos Endogâmicos NOD/fisiologia , Camundongos SCID/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos NOD/imunologia , Camundongos SCID/imunologia , Especificidade da Espécie , Transplante Heterólogo
5.
G3 (Bethesda) ; 7(2): 321-331, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28040777

RESUMO

Epizootic bovine abortion (EBA), or "foothill abortion," is the leading cause of beef cattle abortion in California and has also been reported in Nevada and Oregon. In the 1970s, the soft-shelled tick Ornithodoros coriaceus, or "pajaroello tick," was confirmed as the disease-transmitting vector. In 2005, a novel Deltaproteobacterium was discovered as the etiologic agent of EBA (aoEBA), recently named Pajaroellobacter abortibovis This organism cannot be grown in culture using traditional microbiological techniques; it can only be grown in experimentally-infected severe combined immunodeficient (SCID) mice. The objectives of this study were to perform a de novo genome assembly for P. abortibovis and identify and validate potential antigenic proteins as candidates for future recombinant vaccine development. DNA and RNA were extracted from spleen tissue collected from experimentally-infected SCID mice following exposure to P. abortibovis This combination of mouse and bacterial DNA was sequenced and aligned to the mouse genome. Mouse sequences were subtracted from the sequence pool and the remaining sequences were de novo assembled at 50x coverage into a 1.82 Mbp complete closed circular Deltaproteobacterial genome containing 2250 putative protein-coding sequences. Phylogenetic analysis of P. abortibovis predicts that this bacterium is most closely related to the organisms of the order Myxococcales, referred to as Myxobacteria. In silico prediction of vaccine candidates was performed using a reverse vaccinology approach resulting in the identification and ranking of the top 10 candidate proteins that are likely to be antigenic. Immunologic testing of these candidate proteins confirmed antigenicity of seven of the nine expressed protein candidates using serum from P. abortibovis immunized mice.


Assuntos
Aborto Animal/genética , Aborto Animal/microbiologia , Antígenos de Bactérias/genética , Myxococcales/genética , Aborto Animal/imunologia , Aborto Animal/prevenção & controle , Animais , Antígenos de Bactérias/isolamento & purificação , California , Bovinos , Deltaproteobacteria/genética , Deltaproteobacteria/imunologia , Deltaproteobacteria/patogenicidade , Feminino , Genoma Bacteriano , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Camundongos SCID/imunologia , Camundongos SCID/microbiologia , Myxococcales/imunologia , Filogenia , Gravidez , Vacinação
6.
Physiol Rep ; 4(21)2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27856731

RESUMO

Mycobacterium avium (M. avium) causes significant pulmonary infection, especially in immunocompromised hosts. Alveolar macrophages (AMs) represent the first line of host defense against infection in the lung. Interferon gamma (IFN-γ) activation of AMs enhances in vitro killing of pathogens such as M. avium We hypothesized that airway delivery of AMs into the lungs of immunodeficient mice infected with M. avium will inhibit M. avium growth in the lung and that this macrophage function is in part IFN-γ dependent. In this study, normal BALB/c and BALB/c SCID mice received M. avium intratracheally while on mechanical ventilation. After 30 days, M. avium numbers increased in a concentration-dependent manner in SCID mice compared with normal BALB/c mice. Airway delivery of IFN-γ-activated BALB/c AMs or J774A.1 macrophages overexpressing IFN-γ into the lungs of SCID mice resulted in a significant decrease in M. avium growth (P < 0.01, both comparisons) and limited dissemination to other organs. In addition, airway delivery of IFN-γ activated AMs and macrophages overexpressing IFN-γ increased the levels of IFN-γ and TNF-α in SCID mice. A similar protective effect against M. avium infection using J774A.1 macrophages overexpressing IFN-γ was observed in IFN-γ knockout mice. These data suggest that administration of IFN-γ activated AMs or macrophages overexpressing IFN-γ may partially restore local alveolar host defense against infections like M. avium, even in the presence of ongoing systemic immunosuppression.


Assuntos
Interferon gama/administração & dosagem , Pulmão/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Camundongos SCID/imunologia , Infecções por Mycobacterium/imunologia , Mycobacterium avium/efeitos dos fármacos , Animais , Tolerância Imunológica/efeitos dos fármacos , Interferon gama/biossíntese , Interferon gama/genética , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Mycobacterium/metabolismo , Infecções por Mycobacterium/microbiologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
7.
Sci Rep ; 6: 35387, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27759053

RESUMO

Percutaneous coronary intervention is widely adopted to treat patients with coronary artery disease. However, restenosis remains an unsolved clinical problem after vascular interventions. The role of the systemic and local immune response in the development of restenosis is not fully understood. Hence, the aim of the current study was to investigate the role of the human immune system on subsequent neointima formation elicited by vascular injury in a humanized mouse model. Immunodeficient NOD.Cg-PrkdcscidIL2rgtm1Wjl(NSG) mice were reconstituted with human (h)PBMCs immediately after both carotid wire and femoral cuff injury were induced in order to identify how differences in the severity of injury influenced endothelial regeneration, neointima formation, and homing of human inflammatory and progenitor cells. In contrast to non-reconstituted mice, hPBMC reconstitution reduced neointima formation after femoral cuff injury whereas hPBMCs promoted neointima formation after carotid wire injury 4 weeks after induction of injury. Neointimal endothelium and smooth muscle cells in the injured arteries were of mouse origin. Our results indicate that the immune system may differentially respond to arterial injury depending on the severity of injury, which may also be influenced by the intrinsic properties of the arteries themselves, resulting in either minimal or aggravated neointima formation.


Assuntos
Lesões das Artérias Carótidas/imunologia , Artéria Femoral/imunologia , Oclusão de Enxerto Vascular/imunologia , Leucócitos Mononucleares/imunologia , Lesões do Sistema Vascular/imunologia , Animais , Lesões das Artérias Carótidas/parasitologia , Lesões das Artérias Carótidas/terapia , Modelos Animais de Doenças , Artéria Femoral/lesões , Artéria Femoral/transplante , Oclusão de Enxerto Vascular/fisiopatologia , Humanos , Leucócitos Mononucleares/transplante , Camundongos , Camundongos SCID/imunologia , Camundongos SCID/lesões , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/patologia , Neointima/imunologia , Neointima/fisiopatologia , Lesões do Sistema Vascular/fisiopatologia , Lesões do Sistema Vascular/terapia
8.
Indian J Med Res ; 144(6): 852-864, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28474622

RESUMO

BACKGROUND & OBJECTIVES: Administration of ex vivo-expanded human bone marrow-derived mesenchymal stromal cells (hBMMSC) obtained from single donors has shown therapeutic benefits in both preclinical and clinical studies. In this study, the safety, toxicity and biodistribution profiles of a pooled hBMMSC population, produced from three healthy donors were assessed in rodent and non-rodents. METHODS: The pooled hBMMSC population was characterized by their expression of various cell surface markers, differentiation potential and immunomodulatory activity. To establish in vivo safety of the pooled cells, these were administered by various injection routes into rodents and non-rodents to determine overall toxicity, biodistribution and tumorigenic potential in a series of preclinical studies. RESULTS: Single injections of hBMMSC at various doses through intravenous or intramuscular routes did not cause toxicity in rats and rabbits. In addition, repeat administration of hBMMSC was also well tolerated by rats, and no prenatal toxicity was observed by multiple administration in the same animal species. Ex vivo-expanded and cryopreserved hBMMSCs did not induce tumour formation in severe combined immunodeficient (SCID) mice. INTERPRETATION & CONCLUSIONS: Our results showed that the pooled hBMMSC population was non-toxic, non-teratogenic and non-tumorigenic in animals. Further studies need to be done to find out if it can be safely administered in human patients.


Assuntos
Células da Medula Óssea/citologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Humanos , Masculino , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos SCID/imunologia , Osteogênese/genética , Osteogênese/imunologia , Coelhos , Ratos
9.
Lab Anim ; 49(4): 302-10, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25667226

RESUMO

Sentinel exposure to soiled bedding is frequently used for health monitoring of mice housed in individually ventilated cage systems (IVCS). Despite its advantages, the use of soiled bedding sentinels (SBSs) is far for being a reliable method. Two studies were conducted to evaluate the sensitivity of immunodeficient SBSs NOD.CB17-Prkdc(scid)/NCrHsd (NOD SCID) against two immunocompetent outbred strains, Hsd:ICR (CD-1) and RjOr1:Swiss (Swiss) to pinworm detection in IVCS-housing. Four different diagnostic methods were used: perianal tape test, fecal flotation, plate method and histology. Positivity was considered if at least one of the techniques used was positive. In the first study NOD SCID were more sensitive than CD-1 SBSs (P < 0.05), and except for the fecal flotation test performed at week 6, all the diagnostic methods were more sensitive with NOD SCID mice (P < 0.05). In the second study differences between the Swiss and NOD SCID mice were less obvious (P = 0.08). When compared separately, the different diagnostic methods, except for the fecal flotation test, were all more sensitive in the NOD SCID mice (P < 0.05). In addition, the anal tape test in the Swiss SBSs was more sensitive at week 7 than at week 15 (P < 0.05). In conclusion, combining various diagnostic techniques and samplings at week 7 post-exposure with non-invasive methods increases the rate of pinworm detection. Immunodeficient SBSs showed higher sensitivity than immunocompetent ones. Thus, use of immunodeficient SBSs is highly recommended in health control protocols.


Assuntos
Monitoramento Ambiental/métodos , Camundongos , Oxiuríase/veterinária , Oxyuroidea/isolamento & purificação , Doenças dos Roedores/diagnóstico , Vigilância de Evento Sentinela/veterinária , Animais , Feminino , Pisos e Cobertura de Pisos , Abrigo para Animais , Camundongos/genética , Camundongos/imunologia , Camundongos Endogâmicos ICR/genética , Camundongos Endogâmicos ICR/imunologia , Camundongos Endogâmicos NOD/genética , Camundongos Endogâmicos NOD/imunologia , Camundongos SCID/genética , Camundongos SCID/imunologia , Oxiuríase/diagnóstico , Oxiuríase/parasitologia , Doenças dos Roedores/parasitologia , Organismos Livres de Patógenos Específicos
10.
Nat Rev Immunol ; 12(11): 786-98, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23059428

RESUMO

Significant advances in our understanding of the in vivo functions of human cells and tissues and the human immune system have resulted from the development of 'humanized' mouse strains that are based on severely immunodeficient mice with mutations in the interleukin-2 receptor common γ-chain locus. These mouse strains support the engraftment of a functional human immune system and permit detailed analyses of human immune biology, development and functions. In this Review, we discuss recent advances in the development and utilization of humanized mice, the lessons learnt, the remaining challenges and the promise of using humanized mice for the in vivo study of human immunology.


Assuntos
Sistema Imunitário/fisiologia , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos SCID , Animais , Modelos Animais de Doenças , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos SCID/genética , Camundongos SCID/imunologia , Camundongos Transgênicos
11.
PLoS One ; 7(4): e35497, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558161

RESUMO

Immunodeficient mice reconstituted with human hematopoietic stem cells enable the in vivo study of human hematopoiesis. In particular, NOD-scid-IL2Rγ(null) engrafted mice have been shown to have reasonable levels of T and B cell repopulation and can mount T-cell dependent responses; however, antigen-specific B-cell responses in this model are generally poor. We explored whether developmental defects in the immunoglobulin gene repertoire might be partly responsible for the low level of antibody responses in this model. Roche 454 sequencing was used to obtain over 685,000 reads from cDNA encoding immunoglobulin heavy (IGH) and light (IGK and IGL) genes isolated from immature, naïve, or total splenic B cells in engrafted NOD-scid-IL2Rγ(null) mice, and compared with over 940,000 reads from peripheral B cells of two healthy volunteers. We find that while naïve B-cell repertoires in humanized mice are chiefly indistinguishable from those in human blood B cells, and display highly correlated patterns of immunoglobulin gene segment use, the complementarity-determining region H3 (CDR-H3) repertoires are nevertheless extremely diverse and are specific for each individual. Despite this diversity, preferential D(H)-J(H) pairings repeatedly occur within the CDR-H3 interval that are strikingly similar across all repertoires examined, implying a genetic constraint imposed on repertoire generation. Moreover, CDR-H3 length, charged amino-acid content, and hydropathy are indistinguishable between humans and humanized mice, with no evidence of global autoimmune signatures. Importantly, however, a statistically greater usage of the inherently autoreactive IGHV4-34 and IGKV4-1 genes was observed in the newly formed immature B cells relative to naïve B or total splenic B cells in the humanized mice, a finding consistent with the deletion of autoreactive B cells in humans. Overall, our results provide evidence that key features of the primary repertoire are shaped by genetic factors intrinsic to human B cells and are principally unaltered by differences between mouse and human stromal microenvironments.


Assuntos
Anticorpos Monoclonais Humanizados/genética , Linfócitos B/imunologia , Variação Genética , Hematopoese/imunologia , Camundongos Endogâmicos NOD/imunologia , Camundongos SCID/imunologia , Animais , Anticorpos Monoclonais Humanizados/imunologia , Sequência de Bases , Biologia Computacional , Primers do DNA/genética , DNA Complementar/genética , Citometria de Fluxo , Corantes Fluorescentes , Transplante de Células-Tronco Hematopoéticas , Humanos , Subunidades de Imunoglobulinas/genética , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos , Camundongos Endogâmicos NOD/genética , Camundongos SCID/genética , Dados de Sequência Molecular , Análise de Sequência de DNA , Estatísticas não Paramétricas
12.
Exp Anim ; 60(5): 463-70, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22041283

RESUMO

Pasteurella pneumotropica is an opportunistic pathogen in rodents. Natural infection in immunodeficient animals suggests that immunodeficiency is a major factor in P. pneumotropica pathogenesis. To understand this process, we performed clinical, pathological and bacteriological studies of immunodeficient NOD/ShiJic-scid/Jcl and immunocompetent Crlj:CD1 (ICR) mice experimentally infected with P. pneumotropica ATCC 35149. From 14 days postinoculation, some of P. pneumotropica-infected NOD/ShiJic-scid/Jcl mice developed clinical signs of weight loss. Three of 10 P. pneumotropica-infected NOD/ShiJic-scid/Jcl mice developed clinical signs of depression, ruffled coat, and weight loss and died at 27, 34, and 59 days postinoculation. At 35 days postinoculation, almost all P. pneumotropica-infected NOD/ShiJic-scid/Jcl mice had lung abscesses. The bacteria were isolated from the upper and lower respiratory tracts, including the lungs, and blood. In contrast, P. pneumotropica-infected ICR mice exhibited no clinical signs or lesions. The bacteria were isolated from the upper, but not the lower respiratory tracts. We developed an animal model for understanding host interactions with P. pneumotropica.


Assuntos
Imunocompetência , Hospedeiro Imunocomprometido , Camundongos Endogâmicos ICR/imunologia , Camundongos Endogâmicos ICR/microbiologia , Camundongos Endogâmicos NOD/imunologia , Camundongos Endogâmicos NOD/microbiologia , Camundongos SCID/imunologia , Camundongos SCID/microbiologia , Infecções por Pasteurella/imunologia , Infecções por Pasteurella/microbiologia , Pasteurella pneumotropica/patogenicidade , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Camundongos , Infecções por Pasteurella/patologia , Infecções por Pasteurella/fisiopatologia , Pasteurella pneumotropica/isolamento & purificação , Sistema Respiratório/microbiologia , Sistema Respiratório/patologia , Virulência
13.
Biol Reprod ; 84(4): 682-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21148106

RESUMO

Humanized mice, which refers to immunodeficient mice repopulated with the human immune system, are powerful tools for study in the field of immunology. It has been difficult, however, to generate these transgenic (Tg) mice directly from such strains as the NOD/SCID mouse. In this study, we describe a method developed by us for the generation of Tg mice on an NOD/SCID background. First, we obtained fertilized eggs efficiently by means of in vitro fertilization (IVF); then, we attempted to generate CAG-EGFP Tg mice on an NOD/SCID background, finding that delayed timing of the microinjection after the IVF improved the time to development of the two-cell-stage embryos and the obtainment of newborns. We successfully generated Tg mice and confirmed the germ-line transmission in the offspring. In conclusion, we established a novel system for directly generating transgenic mice on an NOD/SCID background. This novel system is expected to allow improved efficiency of the generation of humanized mice.


Assuntos
Técnicas de Transferência de Genes , Camundongos Endogâmicos NOD/genética , Camundongos Endogâmicos NOD/imunologia , Camundongos SCID/genética , Camundongos SCID/imunologia , Camundongos Transgênicos/genética , Camundongos Transgênicos/imunologia , Animais , Sequência de Bases , Primers do DNA/genética , Feminino , Fertilização In Vitro , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Microinjeções/métodos , Gravidez
14.
J Clin Invest ; 120(6): 2218-29, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20458139

RESUMO

IgE-mediated hypersensitivity is central to the pathogenesis of asthma and other allergic diseases. Although neutralization of serum IgE with IgE-specific antibodies is in general an efficacious treatment for allergic asthma, one limitation of this approach is its lack of effect on IgE production. Here, we have developed a strategy to disrupt IgE production by generating monoclonal antibodies that target a segment of membrane IgE on human IgE-switched B cells that is not present in serum IgE. This segment is known as the M1' domain, and using genetically modified mice that contain the human M1' domain inserted into the mouse IgE locus, we demonstrated that M1'-specific antibodies reduced serum IgE and IgE-producing plasma cells in vivo, without affecting other immunoglobulin isotypes. M1'-specific antibodies were effective when delivered prophylactically and therapeutically in mouse models of immunization, allergic asthma, and Nippostrongylus brasiliensis infection, likely by inducing apoptosis of IgE-producing B cells. In addition, we generated a humanized M1'-specific antibody that was active on primary human cells in vivo, as determined by its reduction of serum IgE levels and IgE plasma cell numbers in a human PBMC-SCID mouse model. Thus, targeting of human IgE-producing B cells with apoptosis-inducing M1'-specific antibodies may be a novel treatment for asthma and allergy.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos/imunologia , Linfócitos B/efeitos dos fármacos , Hipersensibilidade Imediata/imunologia , Camundongos SCID/imunologia , Animais , Anticorpos Monoclonais/imunologia , Asma/imunologia , Linfócitos B/imunologia , Humanos , Hipersensibilidade/imunologia , Imunização , Camundongos , Camundongos Transgênicos , Nippostrongylus/efeitos dos fármacos , Nippostrongylus/imunologia
15.
Curr Opin Endocrinol Diabetes Obes ; 17(2): 120-5, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20150806

RESUMO

PURPOSE OF REVIEW: Update on humanized mouse models and their use in biomedical research. RECENT FINDINGS: The recent description of immunodeficient mice bearing a mutated IL-2 receptor gamma chain (IL2rgamma) facilitated greatly the engraftment and function of human hematolymphoid cells and other cells and tissues. These mice permit the development of human immune systems, including functional T and B cells, following engraftment of hematopoietic stem cells (HSCs). The engrafted functional human immune systems are capable of T and B cell-dependent immune responses, antibody production, antiviral responses, and allograft rejection. Immunodeficient IL2rgamma(null) mice also support heightened engraftment of primary human cancers and malignant progenitor cells, permitting in-vivo investigation of pathogenesis and function. In addition, human-specific infectious agents for which animal models were previously unavailable can now be studied in vivo using these new-generation humanized mice. SUMMARY: Immunodeficient mice bearing an IL2rgamma(null) mutated gene can be engrafted with functional human cells and tissues, including human immune systems, following engraftment with human hematolymphoid cells. These mice are now used as in-vivo models to study human hematopoiesis, immunity, regeneration, stem cell function, cancer, and human-specific infectious agents without putting patients at risk.


Assuntos
Modelos Animais de Doenças , Doença , Camundongos , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Baseada em Transplante de Células e Tecidos/tendências , Doenças Transmissíveis/genética , Doenças Transmissíveis/imunologia , Doenças Transmissíveis/patologia , Doença/genética , Técnicas de Transferência de Genes/tendências , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/fisiologia , Subunidade gama Comum de Receptores de Interleucina/genética , Camundongos SCID/imunologia , Camundongos Transgênicos/imunologia
16.
Am J Physiol Regul Integr Comp Physiol ; 298(4): R1089-97, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20147609

RESUMO

Activation of the immune system by ANG II contributes to the pathogenesis of hypertension, and pharmacological suppression of lymphocyte responses can ameliorate hypertensive end-organ damage. Therefore, to examine the mechanisms through which lymphocytes mediate blood pressure elevation, we studied ANG II-dependent hypertension in scid mice lacking lymphocyte responses and wild-type controls. Scid mice had a blunted hypertensive response to chronic ANG II infusion and accordingly developed less cardiac hypertrophy. Moreover, lymphocyte deficiency led to significant reductions in heart and kidney injury following 4 wk of angiotensin. The muted hypertensive response in the scid mice was associated with increased sodium excretion, urine volumes, and weight loss beginning on day 5 of angiotensin infusion. To explore the mechanisms underlying alterations in blood pressure and renal sodium handling, we measured gene expression for vasoactive mediators in the kidney after 4 wk of ANG II administration. Scid mice and controls had similar renal expression for interferon-gamma, interleukin-1beta, and interleukin-6. By contrast, lymphocyte deficiency (i.e., scid mice) during ANG II infusion led to upregulation of tumor necrosis factor-alpha, endothelial nitric oxide synthase (eNOS), and cyclooxygenase-2 (COX-2) in the kidney. In turn, this enhanced eNOS and COX-2 expression in the scid kidneys was associated with exaggerated renal generation of nitric oxide, prostaglandin E(2), and prostacyclin, all of which promote natriuresis. Thus, the absence of lymphocyte activity protects from hypertension by allowing blood pressure-induced sodium excretion, possibly via stimulation of eNOS- and COX-2-dependent pathways.


Assuntos
Angiotensina II/farmacologia , Hipertensão/induzido quimicamente , Linfócitos/fisiologia , Angiotensina II/efeitos adversos , Animais , Peso Corporal/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/imunologia , Cardiomegalia/fisiopatologia , Cruzamentos Genéticos , Progressão da Doença , Coração/efeitos dos fármacos , Coração/fisiopatologia , Hemodinâmica/fisiologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/lesões , Rim/fisiopatologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos SCID/imunologia , Tamanho do Órgão/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética
18.
Biomed Res ; 30(5): 315-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19887729

RESUMO

SCID mice are a model of human severe combined immunodeficiency disease and are deficient in B cell function in addition to T cell function. Tumors from other species are easily transplanted into SCID mice and will grow without being rejected. We previously reported that the chemokine BRAK/CXCL14 is expressed in normal cells but its expression is down regulated in an in vitro cancer progression model, suggesting that it has the potential for antitumor activity. Here we report that the growth of BRAK/CXCL14 expression vector-transfected oral cancer cells was completely (100%) suppressed in SCID mouse xenografts even though mock-vector introduced control tumor cells grew well with 100% of animals developing tumors. In addition, suppression of xenografts was much faster and the rate was much higher in SCID mice than in T cell function-deficient nude mice. These data indicate the possibility that BRAK expression inhibits tumor cell establishment by regulating interactions between tumor stem cells and NK cells and/or suppressing formation of tumor microvessels.


Assuntos
Quimiocinas CXC/imunologia , Camundongos SCID , Neoplasias Bucais/imunologia , Transplante Heterólogo/imunologia , Animais , Quimiocinas CXC/genética , Feminino , Humanos , Camundongos , Camundongos Nus/imunologia , Camundongos SCID/imunologia , Neoplasias Bucais/patologia , Transplante de Neoplasias , Transplante Heterólogo/patologia
19.
PLoS One ; 4(10): e7251, 2009 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-19802382

RESUMO

BACKGROUND: The lack of a suitable animal model to study viral and immunological mechanisms of human dengue disease has been a deterrent to dengue research. METHODOLOGY/PRINCIPAL FINDINGS: We sought to establish an animal model for dengue virus (DENV) infection and immunity using non-obese diabetic/severe combined immunodeficiency interleukin-2 receptor gamma-chain knockout (NOD-scid IL2rgamma(null)) mice engrafted with human hematopoietic stem cells. Human CD45(+) cells in the bone marrow of engrafted mice were susceptible to in vitro infection using low passage clinical and established strains of DENV. Engrafted mice were infected with DENV type 2 by different routes and at multiple time points post infection, we detected DENV antigen and RNA in the sera, bone marrow, spleen and liver of infected engrafted mice. Anti-dengue IgM antibodies directed against the envelope protein of DENV peaked in the sera of mice at 1 week post infection. Human T cells that developed following engraftment of HLA-A2 transgenic NOD-scid IL2rgamma(null) mice with HLA-A2(+) human cord blood hematopoietic stem cells, were able to secrete IFN-gamma, IL-2 and TNF-alpha in response to stimulation with three previously identified A2 restricted dengue peptides NS4b 2353((111-119)), NS4b 2423((181-189)), and NS4a 2148((56-64)). CONCLUSIONS/SIGNIFICANCE: This is the first study to demonstrate infection of human cells and functional DENV-specific T cell responses in DENV-infected humanized mice. Overall, these mice should be a valuable tool to study the role of prior immunity on subsequent DENV infections.


Assuntos
Vírus da Dengue/imunologia , Sangue Fetal/virologia , Antígeno HLA-A2/imunologia , Hematopoese , Células-Tronco Hematopoéticas/virologia , Interleucina-2/imunologia , Camundongos SCID/imunologia , Animais , Dengue/imunologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Peptídeos/química , RNA/metabolismo
20.
Vet Pathol ; 46(5): 1003-14, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19429977

RESUMO

Infection with human T-cell leukemia virus type 1 (HTLV-1) leads sometimes to the development of adult T-cell lymphoma/leukemia (ATL), which is invariably fatal and often associated with humoral hypercalcemia of malignancy. The transformation of infected CD4 T cells and the pathogenesis of leukemia have been studied with great limitation in tissue culture and patients. To better understand the pathogenesis and perform preclinical drug studies, animal models of ATL are urgently needed. In mice, inoculation of HTLV-1 cell lines mostly leads to development of localized lymphomas. To develop an ATL animal model with leukemic spread of ATL cells, mouse strains with different well-defined immune deficiencies were inoculated intraperitoneally with different HTLV-1-infected cell lines (ACH.2, C8166, MT-2, MET-1). Inoculation of MET-1 cells into NOD/SCID mice provided the best model system for slowly developing T-cell leukemia with multiple organ involvement. In leukemic mice, an increase in serum calcium levels correlated with expression of receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells ligand on leukemic cells and secretion of parathyroid hormone-related protein and interleukin-6. In contrast to the other cell lines that did not spread systemically, MET-1 expressed both the adhesion molecules CD11a (LFA-1alpha) and CD49d (VLA-4alpha) and produced or induced expression of matrix metalloproteinases 1, 2, 3, and 9, thus underlining the importance of these molecules in the spread of adult T-cell leukemia cells. The MET-1/NOD/SCID model will be useful for developing interventions against invasion and spread of leukemic cells and subsequent humoral hypercalcemia of malignancy.


Assuntos
Modelos Animais de Doenças , Hipercalcemia/etiologia , Leucemia-Linfoma de Células T do Adulto/imunologia , Camundongos Endogâmicos NOD/imunologia , Camundongos SCID/imunologia , Animais , Basigina/imunologia , Western Blotting/veterinária , Antígeno CD11a/imunologia , Linhagem Celular , Feminino , Humanos , Hipercalcemia/imunologia , Imuno-Histoquímica/veterinária , Integrina alfa4/imunologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Camundongos Knockout , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/imunologia , Ligante RANK/imunologia , RNA/química , RNA/genética , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...