Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38370707

RESUMEN

Aging studies in mammalian models often depend on natural lifespan data as a primary outcome. Tools for lifespan prediction could accelerate these studies and reduce the need for veterinary intervention. Here, we leveraged large-scale longitudinal frailty and lifespan data on two genetically distinct mouse cohorts to evaluate noninvasive strategies to predict life expectancy in mice. We applied a modified frailty assessment, the Fragility Index, derived from existing frailty indices with additional deficits selected by veterinarians. We developed an ensemble machine learning classifier to predict imminent mortality (95% proportion of life lived [95PLL]). Our algorithm represented improvement over previous predictive criteria but fell short of the level of reliability that would be needed to make advanced prediction of lifespan and thus accelerate lifespan studies. Highly sensitive and specific frailty-based predictive endpoint criteria for aged mice remain elusive. While frailty-based prediction falls short as a surrogate for lifespan, it did demonstrate significant predictive power and as such must contain information that could be used to inform the conclusion of aging experiments. We propose a frailty-based measure of healthspan as an alternative target for aging research and demonstrate that lifespan and healthspan criteria reveal distinct aspects of aging in mice.

2.
PLoS One ; 18(6): e0287204, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37363910

RESUMEN

Tarsal joint abnormalities have been observed in aged male mice on a C57BL background. This joint disease consists of calcaneal displacement, inflammation, and proliferation of cartilage and connective tissue, that can progress to ankylosis of the joint. While tarsal pathology has been described previously in C57BL/6N substrains, as well as in STR/ort and B10.BR strain, no current literature describes this disease occurring in C57BL/6J mice. More importantly the behavioral features that may result from such a change to the joint have yet to be evaluated. This condition was observed in older male mice of the C57BL/6J lineage, around the age of 20 weeks or older, at a frequency of 1% of the population. To assess potential phenotypic sequela, this study sought to evaluate body weight, frailty assessment, home cage wheel running, dynamic weight bearing, and mechanical allodynia with and without the presence of pain relief with morphine. Overall mice with tarsal injuries had significantly higher frailty scores (p< 0.05) and weighed less (p<0.01) compared to unaffected mice. Affected mice had greater overall touch sensitivity (p<0.05) and they placed more weight on their forelimbs (p<0.01) compared to their hind limbs. Lastly, when housed with a running wheel, affected mice ran for a shorter length of time (p<0.01) but tended to run a greater distance within the time they did run (p<0.01) compared to unaffected mice. When tested just after being given morphine, the affected mice performed more similarly to unaffected mice, suggesting there is a pain sensation to this disease process. This highlights the importance of further characterizing inbred mouse mutations, as they may impact research programs or specific study goals.


Asunto(s)
Fragilidad , Actividad Motora , Ratones , Masculino , Animales , Ratones Endogámicos C57BL , Morfina , Dolor
3.
PLoS One ; 15(6): e0230162, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32542000

RESUMEN

Dislocation in hindlimb tarsals are being observed at a low, but persistent frequency in group-housed adult male mice from C57BL/6N substrains. Clinical signs included a sudden onset of mild to severe unilateral or bilateral tarsal abduction, swelling, abnormal hindlimb morphology and lameness. Contraction of digits and gait abnormalities were noted in multiple cases. Radiographical and histological examination revealed caudal dislocation of the calcaneus and partial dislocation of the calcaneoquartal (calcaneus-tarsal bone IV) joint. The detection, frequency, and cause of this pathology in five large mouse production and phenotyping centres (MRC Harwell, UK; The Jackson Laboratory, USA; The Centre for Phenogenomics, Canada; German Mouse Clinic, Germany; Baylor College of Medicine, USA) are discussed.


Asunto(s)
Crianza de Animales Domésticos/instrumentación , Internacionalidad , Huesos Tarsianos/lesiones , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Huesos Tarsianos/diagnóstico por imagen , Tomografía Computarizada por Rayos X
4.
BMC Res Notes ; 10(1): 573, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29116018

RESUMEN

OBJECTIVE: Gain-of-function (GOF) mutations in RHBDF2 cause tylosis. Patients present with hyperproliferative skin, and keratinocytes from tylosis patients' skin show an enhanced wound-healing phenotype. The curly bare mouse model of tylosis, carrying a GOF mutation in the Rhbdf2 gene (Rhbdf2 cub ), presents with epidermal hyperplasia and shows accelerated cutaneous wound-healing phenotype through enhanced secretion of the epidermal growth factor receptor family ligand amphiregulin. Despite these advances in our understanding of tylosis, key questions remain. For instance, it is not known whether the disease is skin-specific, whether the immune system or the surrounding microenvironment plays a role, and whether mouse genetic background influences the hyperproliferative-skin and wound-healing phenotypes observed in Rhbdf2 cub mice. RESULTS: We performed bone marrow transfers and reciprocal skin transplants and found that bone marrow transfer from C57BL/6 (B6)-Rhbdf2 cub/cub donor mice to B6 wildtype recipient mice failed to transfer the hyperproliferative-skin and wound-healing phenotypes in B6 mice. Furthermore, skin grafts from B6 mice to the dorsal skin of B6-Rhbdf2 cub/cub mice maintained the phenotype of the donor mice. To test the influence of mouse genetic background, we backcrossed Rhbdf2 cub onto the MRL/MpJ strain and found that the hyperproliferative-skin and wound-healing phenotypes caused by the Rhbdf2 cub mutation persisted on the MRL/MpJ strain.


Asunto(s)
Proteínas Portadoras/fisiología , Queratinocitos , Queratodermia Palmar y Plantar Difusa/genética , Trasplante de Piel , Cicatrización de Heridas/genética , Animales , Trasplante de Médula Ósea , Proliferación Celular/genética , Modelos Animales de Enfermedad , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Fenotipo
5.
Comp Med ; 67(4): 335-343, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28830580

RESUMEN

Segmented filamentous bacterium (SFB) a gram-positive, anaerobic, and intestinal commensal organism directly influences the development of Th17 helper cells in the small intestine of mice. In NOD mice, SFB colonization interferes with the development of type 1 diabetes (T1D), a T-cell-mediated autoimmune disease, suggesting that SFB may influence Th17 cells to inhibit Th1 populations associated with the anti-ß-cell immune response. This effect is a serious concern for investigators who use NOD mice for diabetes research because the expected incidence of disease decreases markedly when they are colonized by SFB. A room housing mice for T1D studies at The Jackson Laboratory was determined by fecal PCR testing to have widespread SFB colonization of multiple NOD strains after a steady decline in the incidence of T1D was noted. Rederivation of all NOD-related mouse strains was not feasible; therefore an alternative treatment using antibiotics to eliminate SFB from colonized mice was undertaken. After antibiotic treatment, soiled bedding from NOD mouse strains housed in SFB-free high-health-status production barrier rooms was used to reintroduce the gastrointestinal microbiota. Over the past 16 mo since treating the mice and disinfecting the mouse room, regular PCR testing has shown that no additional SFB colonization of mice has occurred, and the expected incidence of T1D has been reestablished in the offspring of treated mice.


Asunto(s)
Ampicilina/farmacología , Antibacterianos/farmacología , Diabetes Mellitus Tipo 1/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Infecciones por Bacterias Grampositivas/veterinaria , Intestinos/efectos de los fármacos , Crianza de Animales Domésticos/métodos , Animales , Descontaminación/métodos , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Monitoreo del Ambiente/métodos , Heces/microbiología , Predisposición Genética a la Enfermedad , Bacterias Grampositivas/clasificación , Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Interacciones Huésped-Patógeno , Intestinos/inmunología , Intestinos/microbiología , Ratones Endogámicos NOD , Fenotipo , Células TH1/inmunología , Células TH1/microbiología , Células Th17/inmunología , Células Th17/microbiología , Factores de Tiempo
6.
Endocrinology ; 158(9): 2707-2712, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28633442

RESUMEN

Previous studies with continuous glucose monitoring in mice have been limited to several days or weeks, with the mouse's physical attachment to the equipment affecting behavior and measurements. In the current study, we measured blood glucose and body temperature at 10-second intervals for 12 weeks in a cohort of NOD/ShiLtJ female mice using wireless telemetry. This allowed us to obtain a high-resolution profile of the circadian rhythm of these two parameters and the onset of hyperglycemic development in real time. The most striking observations were the elevated nocturnal concentrations of glucose into the diabetic range days before elevations in diurnal glucose (when glucose concentrations are historically measured) and the strong, negative correlation between elevated blood glucose concentrations and body temperature with a steady decline of the body temperature with diabetes development. Taken together, this technological advancement provides improved resolution in the study of the disease trajectory of diabetes in mouse models, including relevant translatability to the current technologies of continuous glucose monitoring now regularly used in patients.


Asunto(s)
Glucemia/análisis , Temperatura Corporal/fisiología , Ritmo Circadiano/fisiología , Telemetría/métodos , Tecnología Inalámbrica , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Progresión de la Enfermedad , Femenino , Prueba de Tolerancia a la Glucosa , Hiperglucemia/metabolismo , Hiperglucemia/patología , Ratones , Ratones Endogámicos NOD
7.
Cold Spring Harb Protoc ; 2014(7): 737-40, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24987138

RESUMEN

Multiple sites can be used for the engraftment of primary human cells and tissues into murine hosts. For example, leukemias are usually best engrafted intravenously, but they can also be engrafted directly into the bone marrow cavity. Some solid tumors such as colon tumors grow successfully following subcutaneous engraftment, sometimes requiring provision of a Matrigel artificial basement membrane. In certain cases (e.g., human bladder cancer and ductal breast carcinoma), the use of the autochthonous site (bladder and mammary duct, respectively) is often most efficient, whereas the tumors can grow poorly when transplanted subcutaneously or heterochthonously. Here, we present a protocol for the surgical implantation of tissues under the kidney capsule. The kidney is especially suited for the transplantation of normal as well as malignant cells and tissues. It is very accessible, and transplanted tissues are well contained under the renal capsule in a highly vascularized site. Furthermore, the retroperitoneal location of the kidney, together with its separation from other organs, is advantageous both for imaging and biopsy.


Asunto(s)
Riñón/cirugía , Trasplante Heterólogo/métodos , Animales , Cápsula Glomerular/cirugía , Humanos , Ratones , Trasplante de Neoplasias
8.
Cold Spring Harb Protoc ; 2014(7): 694-708, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24987146

RESUMEN

Since the discovery of the "nude" mouse more than 40 years ago, investigators have attempted to model human tumor growth in immunodeficient mice. Here, we summarize how the field has advanced over the ensuing years owing to improvements in the murine recipients of human tumors. These improvements include the discovery of the scid mutation and development of targeted mutations in the recombination-activating genes 1 and 2 (Rag1(null), Rag2(null)) that severely cripple the adaptive immune response of the murine host. More recently, mice deficient in adaptive immunity have been crossed with mice bearing targeted mutations designed to weaken the innate immune system, ultimately leading to the development of immunodeficient mice bearing a targeted mutation in the gene encoding the interleukin 2 (IL2) receptor common γ chain (IL2rg(null), also known in humans as cytokine receptor common subunit γ). The IL2rg(null) mutation has been used to develop several immunodeficient strains of mice, including the NOD-scid IL2rg(null) (NSG) strain. Using NSG mice as human xenograft recipients, it is now possible to grow almost all types of primary human tumors in vivo, including most solid tumors and hematological malignancies that maintain characteristics of the primary tumor in the patient. Programs to optimize patient-specific therapy using patient-derived xenograft tumor growth in NSG mice have been established at several institutions, including The Jackson Laboratory. Moreover, NSG mice can be engrafted with functional human immune systems, permitting for the first time the potential to study primary human tumors in vivo in the presence of a human immune system.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias/patología , Neoplasias/terapia , Animales , Humanos , Sistema Inmunológico/patología , Ratones , Ratones SCID , Neoplasias/inmunología , Investigación/tendencias , Trasplante Heterólogo/tendencias
9.
PLoS One ; 8(1): e53426, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23301070

RESUMEN

"Hairpatches" (Hpt) is a naturally occurring, autosomal semi-dominant mouse mutation. Hpt/Hpt homozygotes die in utero, while Hpt/+ heterozygotes exhibit progressive renal failure accompanied by patchy alopecia. This mutation is a model for the rare human disorder "glomerulonephritis with sparse hair and telangiectases" (OMIM 137940). Fine mapping localized the Hpt locus to a 6.7 Mb region of Chromosome 4 containing 62 known genes. Quantitative real time PCR revealed differential expression for only one gene in the interval, T-cell acute lymphocytic leukemia 1 (Tal1), which was highly upregulated in the kidney and skin of Hpt/+ mice. Southern blot analysis of Hpt mutant DNA indicated a new EcoRI site in the Tal1 gene. High throughput sequencing identified an endogenous retroviral class II intracisternal A particle insertion in Tal1 intron 4. Our data suggests that the IAP insertion in Tal1 underlies the histopathological changes in the kidney by three weeks of age, and that glomerulosclerosis is a consequence of an initial developmental defect, progressing in severity over time. The Hairpatches mouse model allows an investigation into the effects of Tal1, a transcription factor characterized by complex regulation patterns, and its effects on renal disease.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Elementos Transponibles de ADN/genética , Enfermedades Renales/metabolismo , Mutación , Proteínas Proto-Oncogénicas/genética , Alopecia/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Mapeo Cromosómico , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Ligamiento Genético , Enfermedades Renales/virología , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Tiempo , Factores de Transcripción , Regulación hacia Arriba
10.
Toxicol Pathol ; 41(6): 880-92, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23129576

RESUMEN

Dilated cardiomyopathy (DCM) in A/J mice homozygous for the spontaneous thrombocytopenia and cardiomyopathy (trac) mutation results from a single base pair change in the Abcg5 gene. A similar mutation in humans causes sitosterolemia with high plant sterol levels, hypercholesterolemia, and early onset atherosclerosis. Analyses of CD3+ and Mac-3+ cells and stainable collagen in hearts showed inflammation and myocyte degeneration in A/J-trac/trac mice beginning postweaning and progressed to marked dilative and fibrosing cardiomyopathy by 140 days. Transmission electron microscopy (TEM) demonstrated myocyte vacuoles consistent with swollen endoplasmic reticulum (ER). Myocytes with cytoplasmic glycogen and irregular actinomyosin filament bundles formed mature intercalated disks with normal myocytes suggesting myocyte repair. A/J-trac/trac mice fed lifelong phytosterol-free diets did not develop cardiomyopathy. BALB/cByJ-trac/trac mice had lesser inflammatory infiltrates and later onset DCM. BALB/cByJ-trac/trac mice changed from normal to phytosterol-free diets had lesser T cell infiltrates but persistent monocyte infiltrates and equivalent fibrosis to mice on normal diets. B- and T-cell-deficient BALB/cBy-Rag1(null) trac/trac mice fed normal diets did not develop inflammatory infiltrates or DCM. We conclude that the trac/trac mouse has many features of inflammatory DCM and that the reversibility of myocardial T cell infiltration provides a novel model for investigating the progression of myocardial fibrosis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/deficiencia , Cardiomiopatía Dilatada/metabolismo , Inflamación/metabolismo , Lipoproteínas/deficiencia , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Histocitoquímica , Inflamación/genética , Inflamación/patología , Lipoproteínas/genética , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica , Monocitos/metabolismo , Monocitos/patología , Miocardio/química , Miocardio/metabolismo , Miocardio/patología , Miofibrillas/metabolismo , Miofibrillas/patología , Fitosteroles/farmacología , Linfocitos T/metabolismo , Linfocitos T/patología
11.
J Vis Exp ; (69)2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23207870

RESUMEN

Glioma is the one of the most lethal forms of human cancer. The most effective glioma therapy to date-surgery followed by radiation treatment-offers patients only modest benefits, as most patients do not survive more than five years following diagnosis due to glioma relapse (1,2). The discovery of cancer stem cells in human brain tumors holds promise for having an enormous impact on the development of novel therapeutic strategies for glioma (3). Cancer stem cells are defined by their ability both to self-renew and to differentiate, and are thought to be the only cells in a tumor that have the capacity to initiate new tumors (4). Glioma relapse following radiation therapy is thought to arise from resistance of glioma stem cells (GSCs) to therapy (5-10). In vivo, GSCs are shown to reside in a perivascular niche that is important for maintaining their stem cell-like characteristics (11-14). Central to the organization of the GSC niche are vascular endothelial cells (12). Existing evidence suggests that GSCs and their interaction with the vascular endothelial cells are important for tumor development, and identify GSCs and their interaction with endothelial cells as important therapeutic targets for glioma. The presence of GSCs is determined experimentally by their capability to initiate new tumors upon orthotopic transplantation (15). This is typically achieved by injecting a specific number of GBM cells isolated from human tumors into the brains of severely immuno-deficient mice, or of mouse GBM cells into the brains of congenic host mice. Assays for tumor growth are then performed following sufficient time to allow GSCs among the injected GBM cells to give rise to new tumors-typically several weeks or months. Hence, existing assays do not allow examination of the important pathological process of tumor initiation from single GSCs in vivo. Consequently, essential insights into the specific roles of GSCs and their interaction with the vascular endothelial cells in the early stages of tumor initiation are lacking. Such insights are critical for developing novel therapeutic strategies for glioma, and will have great implications for preventing glioma relapse in patients. Here we have adapted the PoRTS cranial window procedure (16)and in vivo two-photon microscopy to allow visualization of tumor initiation from injected GBM cells in the brain of a live mouse. Our technique will pave the way for future efforts to elucidate the key signaling mechanisms between GSCs and vascular endothelial cells during glioma initiation.


Asunto(s)
Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Glioma/patología , Células Madre Neoplásicas/patología , Cráneo/cirugía , Animales , Neoplasias Encefálicas/irrigación sanguínea , Glioma/irrigación sanguínea , Ratones
12.
Cancer Genet ; 205(5): 232-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22682622

RESUMEN

Although researchers have yet to establish a link between muscular dystrophy (MD) and sarcomas in human patients, literature suggests that the MD genes dystrophin and dysferlin act as tumor suppressor genes in mouse models of MD. For instance, dystrophin-deficient mdx and dysferlin-deficient A/J mice, models of human Duchenne MD and limb-girdle MD type 2B, respectively, develop mixed sarcomas with variable penetrance and latency. To further establish the correlation between MD and sarcoma development, and to test whether a combined deletion of dystrophin and dysferlin exacerbates MD and augments the incidence of sarcomas, we generated dystrophin and dysferlin double mutant mice (STOCK-Dysf(prmd)Dmd(mdx-5Cv)). Not surprisingly, the double mutant mice develop severe MD symptoms and, moreover, develop rhabdomyosarcoma (RMS) at an average age of 12 months, with an incidence of >90%. Histological and immunohistochemical analyses, using a panel of antibodies against skeletal muscle cell proteins, electron microscopy, cytogenetics, and molecular analysis reveal that the double mutant mice develop RMS. The present finding bolsters the correlation between MD and sarcomas, and provides a model not only to examine the cellular origins but also to identify mechanisms and signal transduction pathways triggering development of RMS.


Asunto(s)
Modelos Animales de Enfermedad , Distrofina/genética , Proteínas de la Membrana/genética , Rabdomiosarcoma/genética , Animales , Células Cultivadas , Disferlina , Femenino , Marcadores Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Mutación , Cariotipificación Espectral
13.
Curr Protoc Mouse Biol ; 2(3): 263-71, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26069014

RESUMEN

Personnel performing surgery on mice commonly lack formal medical training and most training in mouse surgery focuses on the mechanical skills required for a procedure with less emphasis on perioperative techniques. Consequently, a basic concept that underpins successful surgery, aseptic technique, may not be fully understood or incorporated into the training process. This unit provides a framework in which to plan and carry out surgical procedures in mice using appropriate aseptic technique. Curr. Protoc. Mouse Biol. 2:263-271 © 2012 by John Wiley & Sons, Inc.

14.
Blood ; 115(6): 1267-76, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19846887

RESUMEN

The spontaneous mouse mutation "thrombocytopenia and cardiomyopathy" (trac) causes macrothrombocytopenia, prolonged bleeding times, anemia, leukopenia, infertility, cardiomyopathy, and shortened life span. Homozygotes show a 20-fold decrease in platelet numbers and a 3-fold increase in platelet size with structural alterations and functional impairments in activation and aggregation. Megakaryocytes in trac/trac mice are present in increased numbers, have poorly developed demarcation membrane systems, and have decreased polyploidy. The thrombocytopenia is not intrinsic to defects at the level of hematopoietic progenitor cells but is associated with a microenvironmental abnormality. The trac mutation maps to mouse chromosome 17, syntenic with human chromosome 2p21-22. A G to A mutation in exon 10 of the adenosine triphosphate (ATP)-binding cassette subfamily G, member 5 (Abcg5) gene, alters a tryptophan codon (UGG) to a premature stop codon (UAG). Crosses with mice doubly transgenic for the human ABCG5 and ABCG8 genes rescued platelet counts and volumes. ABCG5 and ABCG8 form a functional complex that limits dietary phytosterol accumulation. Phytosterolemia in trac/trac mice confirmed a functional defect in the ABCG5/ABCG8 transport system. The trac mutation provides a new clinically significant animal model for human phytosterolemia and provides a new means for studying the role of phytosterols in hematologic diseases and testing therapeutic interventions.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Cardiomiopatías/genética , Modelos Animales de Enfermedad , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas/fisiología , Mutación/genética , Fitosteroles/metabolismo , Sitoesteroles/metabolismo , Trombocitopenia/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Tiempo de Sangría , Cardiomiopatías/patología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Cruzamientos Genéticos , Femenino , Feto/citología , Feto/metabolismo , Errores Innatos del Metabolismo Lipídico/patología , Lipoproteínas/genética , Hígado/citología , Hígado/metabolismo , Masculino , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Trombocitopenia/patología
15.
Nat Biotechnol ; 25(11): 1315-21, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17952057

RESUMEN

Acute myelogenous leukemia (AML) is the most common adult leukemia, characterized by the clonal expansion of immature myeloblasts initiating from rare leukemic stem (LS) cells. To understand the functional properties of human LS cells, we developed a primary human AML xenotransplantation model using newborn nonobese diabetic/severe combined immunodeficient/interleukin (NOD/SCID/IL)2r gamma(null) mice carrying a complete null mutation of the cytokine gamma c upon the SCID background. Using this model, we demonstrated that LS cells exclusively recapitulate AML and retain self-renewal capacity in vivo. They home to and engraft within the osteoblast-rich area of the bone marrow, where AML cells are protected from chemotherapy-induced apoptosis. Quiescence of human LS cells may be a mechanism underlying resistance to cell cycle-dependent cytotoxic therapy. Global transcriptional profiling identified LS cell-specific transcripts that are stable through serial transplantation. These results indicate the potential utility of this AML xenograft model in the development of novel therapeutic strategies targeted at LS cells.


Asunto(s)
División Celular , Resistencia a Antineoplásicos , Leucemia Mieloide Aguda/metabolismo , Modelos Biológicos , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Animales , Apoptosis , Médula Ósea , Perfilación de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Mutantes , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Osteoblastos
16.
Ann N Y Acad Sci ; 1103: 77-89, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17332083

RESUMEN

There are many rodent models of autoimmune diabetes that have been used to study the pathogenesis of human type 1 diabetes (T1D), including the non-obese diabetic (NOD) mouse, the biobreeding (BB) rat, and the transgenic mouse models. However, mice and rats are not humans, and these rodent models do not completely recapitulate the autoimmune pathogenesis of the human disease. In addition, many of the reagents, tools, and therapeutics proposed for use in humans may be species specific and cannot be investigated in rodents. Researchers have used nonhuman primates to more closely mimic the human immune system and, to study species-specific therapeutics, but these studies are associated with additional ethical and economic constraints and, to date, no model of autoimmune diabetes in this species has been described. New animal models are needed that will permit the in vivo investigation of human immune systems and analyses of the pathogenesis of human T1D without putting individuals at risk. To fill this need, we are developing humanized mouse models for the in vivo study of T1D. These models are based on our newly generated stock of NOD-scid IL2rgamma(null) mice, which engraft at higher levels with human hematolymphoid cells and exhibit enhanced function of the engrafted human immune systems compared with previous humanized mouse models. Overall, development of these new generations of humanized mice should facilitate in vivo studies of the human immune system as well as permit the investigation of the pathogenesis and effector phases of human T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Reproducibilidad de los Resultados
17.
FASEB J ; 20(7): 950-2, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16585061

RESUMEN

To obtain insights into the cardiomyogenic potential of hematopoietic tissue, we intravenously (i.v.) injected purified hematopoietic stem/progenitor cells into newborn recipients that may fully potentiate the developmental plasticity of stem cells. Transplantation of mouse bone marrow (BM) lineage antigen-negative (Lin-) cells resulted in the generation of the cells that displayed cardiomyocyte-specific antigenic profiles and contractile function when transplanted into syngeneic newborn recipients. To clarify the mechanism underlying the cardiomyogenic potential, green fluorescent protein (GFP)-labeled BM Lin-ScaI+ hematopoietic progenitors were transplanted into neonatal mice constitutively expressing cyan fluorescence protein (CFP). Lambda image acquisition and linear unmixing analysis using confocal microscopy successfully separated GFP and CFP, and revealed that donor GFP+ cardiomyocytes coexpressed host-derived CFP. We further reconstituted human hemopoietic- and immune systems in mice by injecting human cord blood (CB)-derived Lin-CD34+CD38- hematopoietic stem cells (HSCs) into neonatal T cell(-)B cell(-)NK cell- immune-deficient NOD/SCID/IL2rgamma(null) mice. Fluoroescence in situ hybridization analysis of recipient cardiac tissues demonstrated that human and murine chromosomes were colocalized in the same cardiomyocytes, indicating that cell fusion occurred between human hematopoietic progeny and mouse cardiomyocytes. These syngeneic- and xenogeneic neonatal transplantations provide compelling evidence that hematopoietic stem/progenitor cells contribute to the postnatal generation of cardiomyocytes through cell fusion, not through transdifferentiation.


Asunto(s)
Células Madre Hematopoyéticas/citología , Miocitos Cardíacos/citología , Animales , Fusión Celular , Separación Celular , Células Cultivadas , Cromosomas Humanos , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Subunidad gamma Común de Receptores de Interleucina , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Receptores de Interleucina/genética
18.
Invest Ophthalmol Vis Sci ; 47(3): 1201-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16505059

RESUMEN

PURPOSE: Viable motheaten mutant mice (abbreviated allele symbol me(v)) are deficient in Src-homology 2-domain phosphatase (SHP)-1, a critical negative regulator of signal transduction in hematopoietic cells. These mice exhibit immune dysfunction, hyperproliferation of myeloid cells, and regenerative anemia. This study focused on the role of SHP-1 in retinal homeostasis. METHODS: Ophthalmoscopy, histology, transmission electron microscopy (TEM), electroretinography (ERG), immunohistochemistry, Western blot, bone marrow transplantation, and genetic crosses were performed for phenotypic characterization and functional studies of retinal degeneration (RD) in me(v)/me(v) mice. RESULTS: Fundus examinations of me(v)/me(v) mice revealed numerous, small white spots. Histologic examination demonstrated photoreceptor loss beginning at 3 weeks of age, and TEM revealed disorganization and reduction in the number of outer segments, as well as the presence of phagocytic cells in the subretinal space. Rod- and cone-mediated ERGs were abnormal. SHP-1 protein was expressed in mouse and human retinal lysates and was localized to the outer nuclear layer of the retina in me(v)/me(v) and control mice. Autoantibodies are not necessary for RD, as B-cell-deficient me(v)/me(v) Igh-6(tm1Cgn) mice had no attenuation of photoreceptor cell loss compared with age-matched me(v)/me(v) mice. Histologic examination of lungs and retinas from normal recipients of me(v)/me(v) marrow revealed the classic acidophilic macrophage pneumonia of me(v)/me(v) mice, but no retinal degeneration. CONCLUSIONS: me(v)/me(v) mice exhibit normal retinal development with the onset of RD at 3 weeks of age and a rapidly progressive loss of photoreceptors. These findings support the hypothesis that SHP-1 plays a critical role in retinal homeostasis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas Tirosina Fosfatasas/deficiencia , Degeneración Retiniana/enzimología , Animales , Apoptosis , Western Blotting , Trasplante de Médula Ósea , Cruzamientos Genéticos , Electrorretinografía , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Oftalmoscopía , Células Fotorreceptoras de Vertebrados/enzimología , Células Fotorreceptoras de Vertebrados/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Degeneración Retiniana/fisiopatología
19.
J Exp Med ; 203(2): 275-80, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16476768

RESUMEN

Rheumatoid arthritis (RA) is a complex autoimmune disease with a poorly understood pathogenesis. The disease is associated with polyclonal B cell activation and the production of autoantibodies (autoAbs), but there is a longstanding controversy as to whether such Abs contribute to, or are secondary to, the pathogenesis of RA. To address the potential pathogenicity of human RA-associated Abs, we developed a passive transfer model involving mice deficient in the low-affinity inhibitory Fc receptor, FcgammaRIIB. We report that plasma or serum from patients with active RA can induce inflammation and histological lesions in FcgammaRIIB-/- mice consistent with arthritis, and that this pathogenic activity is caused by the immunoglobulin G-rich fraction. Our results suggest that humoral autoimmunity can contribute directly to autoimmune arthritis, and that FcgammaRIIB-/- mice are a promising model to evaluate the arthritogenic potential of human autoAbs.


Asunto(s)
Artritis Experimental/inmunología , Autoanticuerpos/toxicidad , Sitios de Unión de Anticuerpos , Receptores de IgG/deficiencia , Receptores de IgG/genética , Animales , Artritis Experimental/genética , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Sitios de Unión de Anticuerpos/genética , Femenino , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Invest Ophthalmol Vis Sci ; 46(11): 4097-106, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16249486

RESUMEN

PURPOSE: The present study was conducted to investigate healing of alkali-burned corneas in MRL/MpJ (MRL) mice. METHODS: Gross, clinical, and histologic criteria were used to compare healing of alkali-burned corneas in MRL and control C57BL/6J (B6) mice. Effects of neutrophil depletion of B6 mice and allogeneic reconstitution of B6 mice with MRL bone marrow on wound healing were evaluated. Gene expression patterns in normal and wounded corneas were surveyed with array-based quantitative real-time RT-PCR (AQPCR). RESULTS: MRL mice showed accelerated reepithelialization and decreased corneal opacity compared with B6 mice after alkali wounding. Marked inflammatory cell infiltration and fibrosis were evident in the corneas and anterior chambers of B6 mice. MRL mice showed less severe lesions, except for stromal edema. Rapid reepithelialization and reduced keratitis/iritis were also observed in neutrophil-depleted B6 mice, but not in B6 mice reconstituted with MRL bone marrow. AQPCR showed transcriptional changes of fewer genes associated with inflammation and corneal tissue homeostasis in alkali-burned corneas from MRL mice. Increased expression of an anti-inflammatory gene, Socs1, and a gene associated with healing, Mmp1a, were evident in MRL corneas. CONCLUSIONS: Alkali-burned corneas heal faster and more completely in MRL mice than in B6 mice, by means of rapid reepithelialization, reduced inflammation, and reduced fibrosis. Reduced inflammation, including decreased neutrophil infiltrates and the lack of a robust proinflammatory gene expression signature correlates with the rapid healing. However, the rapid-healing phenotype is not intrinsic to MRL hematopoietic progenitor cells.


Asunto(s)
Quemaduras Químicas/metabolismo , Enfermedades de la Córnea/metabolismo , Quemaduras Oculares/inducido químicamente , Cicatrización de Heridas/fisiología , Animales , Trasplante de Médula Ósea , Quemaduras Químicas/inmunología , Quemaduras Químicas/patología , Quemaduras Químicas/cirugía , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Colagenasas/genética , Colagenasas/metabolismo , Enfermedades de la Córnea/inmunología , Enfermedades de la Córnea/patología , Enfermedades de la Córnea/cirugía , Modelos Animales de Enfermedad , Epitelio Corneal/fisiología , Citometría de Flujo , Regulación de la Expresión Génica/fisiología , Masculino , Metaloproteinasa 1 de la Matriz , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Neutrófilos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Hidróxido de Sodio , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA