Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Regen Ther ; 24: 451-458, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37772130

RESUMEN

Introduction: Duchenne muscular dystrophy (DMD) is a hereditary neuromuscular disorder caused by mutation in the dystrophin gene (DMD) on the X chromosome. Female DMD carriers occasionally exhibit symptoms such as muscle weakness and heart failure. Here, we investigated the characteristics and representativeness of female DMD carrier (DMD-XKOXWT) pigs as a suitable disease model. Methods: In vitro fertilization using sperm from a DMD-XKOY↔XWTXWT chimeric boar yielded DMD-XKOXWT females, which were used to generate F2 and F3 progeny, including DMD-XKOXWT females. F1-F3 piglets were genotyped and subjected to biochemical analysis for blood creatine kinase (CK), aspartate aminotransferase, and lactate dehydrogenase. Skeletal muscle and myocardial tissue were analyzed for the expression of dystrophin and utrophin, as well as for lymphocyte and macrophage infiltration. Results: DMD-XKOXWT pigs exhibited various characteristics common to human DMD carrier patients, namely, asymptomatic hyperCKemia, dystrophin expression patterns in the skeletal and cardiac muscles, histopathological features of skeletal muscle degeneration, myocardial lesions in adulthood, and sporadic death. Pathological abnormalities observed in the skeletal muscles in DMD-XKOXWT pigs point to a frequent incidence of pathological abnormalities in the musculoskeletal tissues of latent DMD carriers. Our findings suggest a higher risk of myocardial abnormalities in DMD carrier women than previously believed. Conclusions: We demonstrated that DMD-XKOXWT pigs could serve as a suitable large animal model for understanding the pathogenic mechanism in DMD carriers and developing therapies for female DMD carriers.

2.
Mol Ther Nucleic Acids ; 33: 444-453, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37588685

RESUMEN

Mammalian artificial chromosomes have enabled the introduction of extremely large amounts of genetic information into animal cells in an autonomously replicating, nonintegrating format. However, the evaluation of human artificial chromosomes (HACs) as novel tools for curing intractable hereditary disorders has been hindered by the limited efficacy of the delivery system. We generated dystrophin gene knockout (DMD-KO) pigs harboring the HAC bearing the entire human DMD via a somatic cell cloning procedure (DYS-HAC-cloned pig). Restored human dystrophin expression was confirmed by immunofluorescence staining in the skeletal muscle of the DYS-HAC-cloned pigs. Viability at the first month postpartum of the DYS-HAC-cloned pigs, including motor function in the hind leg and serum creatinine kinase level, was improved significantly when compared with that in the original DMD-KO pigs. However, decrease in systemic retention of the DYS-HAC vector and limited production of the DMD protein might have caused severe respiratory impairment with general prostration by 3 months postpartum. The results demonstrate that the use of transchromosomic cloned pigs permitted a straightforward estimation of the efficacy of the DYS-HAC carried in affected tissues/organs in a large-animal disease model, providing novel insights into the therapeutic application of exogenous mammalian artificial chromosomes.

3.
Lab Invest ; 103(2): 100013, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-37039150

RESUMEN

Amyotrophic lateral sclerosis (ALS) causes progressive degeneration of the motor neurons. In this study, we delivered the genetic construct including the whole locus of human mutant superoxide dismutase 1 (SOD1) with the promoter region of human SOD1 into porcine zygotes using intracytoplasmic sperm injection-mediated gene transfer, and we thereby generated a pig model of human mutant SOD1-mediated familial ALS. The established ALS pig model exhibited an initial abnormality of motor neurons with accumulated misfolded SOD1. The ALS pig model, with a body size similar to that of human beings, will provide opportunities for cell and gene therapy platforms in preclinical translational research.


Asunto(s)
Esclerosis Amiotrófica Lateral , Superóxido Dismutasa-1 , Animales , Humanos , Masculino , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Modelos Animales de Enfermedad , Neuronas Motoras/patología , Mutación , Semen , Superóxido Dismutasa-1/genética , Porcinos
4.
J Inherit Metab Dis ; 45(6): 1059-1069, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35866457

RESUMEN

Patients with urea cycle disorders intermittently develop episodes of decompensation with hyperammonemia. Although such an episode is often associated with starvation and catabolism, its molecular basis is not fully understood. First, we attempted to elucidate the mechanism of such starvation-associated hyperammonemia. Using a mouse embryonic fibroblast (MEF) culture system, we found that glucose starvation increases ammonia production, and that this increase is associated with enhanced glutaminolysis. These results led us to focus on α-ketoglutarate (AKG), a glutamate dehydrogenase inhibitor, and a major anaplerotic metabolite. Hence, we sought to determine the effect of dimethyl α-ketoglutarate (DKG), a cell-permeable AKG analog, on MEFs and found that DKG mitigates ammonia production primarily by reducing flux through glutamate dehydrogenase. We also verified that DKG reduces ammonia in an NH4 Cl-challenged hyperammonemia mouse model and observed that DKG administration reduces plasma ammonia concentration to 22.8% of the mean value for control mice that received only NH4 Cl. In addition, we detected increases in ornithine concentration and in the ratio of ornithine to arginine following DKG treatment. We subsequently administered DKG intravenously to a newborn pig with hyperammonemia due to ornithine transcarbamylase deficiency and found that blood ammonia concentration declined significantly over time. We determined that this effect is associated with facilitated reductive amination and glutamine synthesis. Our present data indicate that energy starvation triggers hyperammonemia through enhanced glutaminolysis and that DKG reduces ammonia accumulation via pleiotropic mechanisms both in vitro and in vivo. Thus, cell-permeable forms of AKG are feasible candidates for a novel hyperammonemia treatment.


Asunto(s)
Hiperamonemia , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa , Ratones , Animales , Porcinos , Hiperamonemia/tratamiento farmacológico , Hiperamonemia/metabolismo , Glutamina/metabolismo , Amoníaco , Glutamato Deshidrogenasa , Fibroblastos/metabolismo , Ornitina
5.
Regen Ther ; 21: 62-72, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35765545

RESUMEN

Introduction: Previously, we performed gene knockout (KO) of interleukin-2 receptor gamma (IL2RG) in porcine fetal fibroblasts using zinc finger nuclease-encoding mRNAs, subsequently generating IL2RG KO pigs using these cells through somatic cell nuclear transfer. The IL2RG KO pigs lacked a thymus and were deficient in T lymphocytes and natural killer cells, similar to human X-linked severe combined immunodeficiency (SCID) patients. The present study aimed to evaluate whether pigs can support the growth of xenografted human cells and have the potential to be an effective animal model. Methods: The IL2RG XKOY pigs used in this study were obtained by mating IL2RG XKOX females with wild-type boars. This permitted the routine production of IL2RG KO pigs via natural breeding without complicated somatic cell cloning procedures; therefore, a sufficient number of pigs could be prepared. We transplanted human HeLa S3 cells expressing the tandem dimer tomato into the ears and pancreas of IL2RG KO pigs. Additionally, a newly developed method for the aseptic rearing of SCID pigs was used in case of necessity. Results: Tumors from the transplanted cells quickly developed in all pigs and were verified by histology and immunohistochemistry. We also transplanted these cells into the pancreas of designated pathogen-free pigs housed in novel biocontainment facilities, and large tumors were confirmed. Conclusions: IL2RG KO pigs have the potential to become useful animal models in a variety of translational biology fields.

6.
Lab Invest ; 102(5): 560-569, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34980882

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, manifesting as the progressive development of fluid-filled renal cysts. In approximately half of all patients with ADPKD, end-stage renal disease results in decreased renal function. In this study, we used CRISPR-Cas9 and somatic cell cloning to produce pigs with the unique mutation c.152_153insG (PKD1insG/+). Pathological analysis of founder cloned animals and progeny revealed that PKD1insG/+ pigs developed many pathological conditions similar to those of patients with heterozygous mutations in PKD1. Pathological similarities included the formation of macroscopic renal cysts at the neonatal stage, number and cystogenic dynamics of the renal cysts formed, interstitial fibrosis of the renal tissue, and presence of a premature asymptomatic stage. Our findings demonstrate that PKD1insG/+ pigs recapitulate the characteristic symptoms of ADPKD.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Animales , Femenino , Heterocigoto , Humanos , Riñón/patología , Masculino , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Porcinos , Canales Catiónicos TRPP/genética
7.
Development ; 148(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34874452

RESUMEN

Despite four decades of effort, robust propagation of pluripotent stem cells from livestock animals remains challenging. The requirements for self-renewal are unclear and the relationship of cultured stem cells to pluripotent cells resident in the embryo uncertain. Here, we avoided using feeder cells or serum factors to provide a defined culture microenvironment. We show that the combination of activin A, fibroblast growth factor and the Wnt inhibitor XAV939 (AFX) supports establishment and continuous expansion of pluripotent stem cell lines from porcine, ovine and bovine embryos. Germ layer differentiation was evident in teratomas and readily induced in vitro. Global transcriptome analyses highlighted commonality in transcription factor expression across the three species, while global comparison with porcine embryo stages showed proximity to bilaminar disc epiblast. Clonal genetic manipulation and gene targeting were exemplified in porcine stem cells. We further demonstrated that genetically modified AFX stem cells gave rise to cloned porcine foetuses by nuclear transfer. In summary, for major livestock mammals, pluripotent stem cells related to the formative embryonic disc are reliably established using a common and defined signalling environment. This article has an associated 'The people behind the papers' interview.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/metabolismo , Estratos Germinativos/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Bovinos , Embrión de Mamíferos/citología , Estratos Germinativos/citología , Ganado , Células Madre Pluripotentes/citología , Ovinos , Especificidad de la Especie , Porcinos
8.
J Clin Med ; 10(15)2021 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-34362010

RESUMEN

To develop novel medical technologies, pig disease models are invaluable especially in the final stages of translational research. Recently, we established a genetically engineered ornithine transcarbamylase-deficient (OTCD) pig strain. Here, we report its characterization and treatment responsiveness. OTCD pigs were obtained by mating an OTCD carrier female (OTC-Xc.186_190delXWT) with a wild-type male. Due to the X-linked recessive mode of inheritance, the disease phenotype emerged only in males. Medication with nitrogen-scavenging agents was based on a clinical protocol. OTCD pigs were born smaller than their wild-type and carrier littermates, showing anemia and faltering. Biochemically, high levels of urinary orotic acid and loss of OTC activity were observed. The natural life course of OTCD pigs was characterized by a decrease in arterial percentage saturation of oxygen and body temperature, as well as an increase in blood ammonia levels; the pigs died in 24.0 ± 5.0 h (mean ± SD, n = 6). The established standard medication composed with nitrogen-scavenging agents and transfusion nearly doubled the survival time to 42.4 ± 13.7 h (n = 6). Our OTCD pig model appropriately mimicked the human pathology. Along with established protocols in handling and medication, this is a first step in developing a large animal disease model that is useful for translational research into novel medical technologies, such as cell transplantation and gene therapy, as well as in relation to urea cycle disorder.

9.
World J Diabetes ; 12(4): 306-330, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33889282

RESUMEN

Diabetes is among the top 10 causes of death in adults and caused approximately four million deaths worldwide in 2017. The incidence and prevalence of diabetes is predicted to increase. To alleviate this potentially severe situation, safer and more effective therapeutics are urgently required. Mice have long been the mainstay as preclinical models for basic research on diabetes, although they are not ideally suited for translating basic knowledge into clinical applications. To validate and optimize novel therapeutics for safe application in humans, an appropriate large animal model is needed. Large animals, especially pigs, are well suited for biomedical research and share many similarities with humans, including body size, anatomical features, physiology, and pathophysiology. Moreover, pigs already play an important role in translational studies, including clinical trials for xenotransplantation. Progress in genetic engineering over the past few decades has facilitated the development of transgenic animals, including porcine models of diabetes. This article discusses features that attest to the attractiveness of genetically modified porcine models of diabetes for testing novel treatment strategies using recent technical advances.

10.
Artículo en Inglés | MEDLINE | ID: mdl-33257422

RESUMEN

INTRODUCTION: Pancreatic duodenum homeobox 1 (Pdx1) expression is crucial for pancreatic organogenesis and is a key regulator of insulin gene expression. Hairy and enhancer of split 1 (Hes1) controls tissue morphogenesis by maintaining undifferentiated cells. Hes1 encodes a basic helix loop helix (bHLH) transcriptional repressor and functionally antagonizes positive bHLH genes, such as the endocrine determination gene neurogenin-3. Here, we generated a new pig model for diabetes by genetic engineering Pdx1 and Hes1 genes. RESEARCH DESIGN AND METHODS: A transgenic (Tg) chimera pig with germ cells carrying a construct expressing Hes1 under the control of the Pdx1 promoter was used to mate with wild-type gilts to obtain Tg piglets. RESULTS: The Tg pigs showed perinatal death; however, this phenotype could be rescued by insulin treatment. The duodenal and splenic lobes of the Tg pigs were slender and did not fully develop, whereas the connective lobe was absent. ß cells were not detected, even in the adult pancreas, although other endocrine cells were detected, and exocrine cells functioned normally. The pigs showed no irregularities in any organs, except diabetes-associated pathological alterations, such as retinopathy and renal damage. CONCLUSION: Pdx1-Hes1 Tg pigs were an attractive model for the analysis of pancreatic development and testing of novel treatment strategies for diabetes.


Asunto(s)
Diabetes Mellitus , Células Secretoras de Insulina , Animales , Femenino , Ingeniería Genética , Proteínas de Homeodominio/genética , Embarazo , Porcinos , Transactivadores/genética
11.
Regen Ther ; 14: 215-221, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32435673

RESUMEN

INTRODUCTION: Regenerative therapy using chondrocyte sheets is effective for osteoarthritis. The clinical application of chondrocyte sheet therapy is expected to be further advanced by the use of a feasible cryopreservation technique. Previously, we developed a chondrocyte sheet vitrification method; however, it was too complex to be used for routine clinical application. Here, we aimed to develop a prototype method for vitrifying chondrocyte sheets for clinical practice. METHODS: We developed a "circulating vitrification bag" as a container to process cell sheets for vitrification in an efficient and sanitary fashion. Moreover, we invented the "vitrification storage box", which is useful for the vitrification of cell sheets, long-term preservation, and transportation. These devices were used to vitrify rabbit chondrocyte sheets, which were then assessed for their structural characteristics and the viability of the component cells after rewarming. RESULTS: In all cell sheet samples (n = 7) vitrified by the circulating vitrification bag method, the integrity of the sheet structure was maintained, and the cell survival rate was similar to that of non-vitrified samples (91.0 ± 2.9% vs. 90.0 ± 3.0%). Proteoglycan and type II collagen, which are major components of cartilage, were densely and evenly distributed throughout the chondrocyte sheet subjected to vitrification similarly to that observed in the non-vitrified sheet. After long-term storage using the vitrification storage box, the cell sheets maintained normal structure and cell viability (survival rate: 81.2 ± 1.0% vs. 84.3 ± 1.8%) compared to the non-vitrified sheet. CONCLUSION: Our results indicate that the circulating vitrification bag method is an effective approach for realizing the clinical application of vitrified chondrocyte sheets. The vitrification storage box is also useful for the long-term preservation of vitrified cell sheets, further enhancing the feasibility of the clinical application of cryopreserved chondrocyte sheets.

12.
Reprod Med Biol ; 19(2): 142-150, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32273819

RESUMEN

PURPOSE: This study aims to demonstrate vitrification methods that provide reliable cryopreservation for embryos with compromised cryotolerance. METHODS: Two-cell stage mouse embryos and in vitro produced porcine embryos were vitrified using the hollow fiber vitrification (HFV) and Cryotop (CT) methods. The performance of these two methods was compared by the viability of the vitrified-rewarmed embryos. RESULTS: Regardless of the method used, 100% of the mouse 2-cell embryos developed successfully after vitrification-rewarming into the blastocyst stage, whereas vitrification tests using porcine morulae with the HFV method produced significantly better results. The developmental rates of vitrified porcine morula into the blastocyst stage, as well as blastocyst cell number, were 90.3% and 112.3 ± 6.9 in the HFV group compared with 63.4% and 89.5 ± 8.1 in the CT group (P < .05). Vitrification tests using 4- to 8-cell porcine embryos resulted in development into the blastocyst stage (45.5%) in the HFV group alone, demonstrating its better efficacy. The HFV method did not impair embryo viability, even after spontaneous rewarming at room temperature for vitrified embryos, which is generally considered a contraindication. CONCLUSION: Vitrification test using embryos with compromised cryotolerance allows for more precise determining of effective cryopreservation methods and devices.

13.
Lab Invest ; 100(6): 900, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32203148

RESUMEN

This article was originally published under Nature Research's License to Publish, but has now been made available under a [CC BY 4.0] license. The PDF and HTML versions of the article have been modified accordingly.

14.
Lab Invest ; 100(6): 887-899, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32060408

RESUMEN

Genetic cardiomyopathy is a group of intractable cardiovascular disorders involving heterogeneous genetic contribution. This heterogeneity has hindered the development of life-saving therapies for this serious disease. Genetic mutations in dystrophin and its associated glycoproteins cause cardiomuscular dysfunction. Large animal models incorporating these genetic defects are crucial for developing effective medical treatments, such as tissue regeneration and gene therapy. In the present study, we knocked out the δ-sarcoglycan (δ-SG) gene (SGCD) in domestic pig by using a combination of efficient de novo gene editing and somatic cell nuclear transfer. Loss of δ-SG expression in the SGCD knockout pigs caused a concomitant reduction in the levels of α-, ß-, and γ-SG in the cardiac and skeletal sarcolemma, resulting in systolic dysfunction, myocardial tissue degeneration, and sudden death. These animals exhibited symptoms resembling human genetic cardiomyopathy and are thus promising for use in preclinical studies of next-generation therapies.


Asunto(s)
Cardiomiopatías , Sarcoglicanos , Animales , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Femenino , Mutación del Sistema de Lectura/genética , Técnicas de Inactivación de Genes , Masculino , Miocardio/química , Miocardio/metabolismo , Miocardio/patología , Sarcoglicanos/deficiencia , Sarcoglicanos/genética , Porcinos
15.
Stem Cell Reports ; 14(1): 21-33, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31883918

RESUMEN

We have previously established a concept of developing exogenic pancreas in a genetically modified pig fetus with an apancreatic trait, thereby proposing the possibility of in vivo generation of functional human organs in xenogenic large animals. In this study, we aimed to demonstrate a further proof-of-concept of the compensation for disabled organogeneses in pig, including pancreatogenesis, nephrogenesis, hepatogenesis, and vasculogenesis. These dysorganogenetic phenotypes could be efficiently induced via genome editing of the cloned pigs. Induced dysorganogenetic traits could also be compensated by allogenic blastocyst complementation, thereby proving the extended concept of organ regeneration from exogenous pluripotent cells in empty niches during various organogeneses. These results suggest that the feasibility of blastocyst complementation using genome-edited cloned embryos permits experimentation toward the in vivo organ generation in pigs from xenogenic pluripotent cells.


Asunto(s)
Blastocisto/citología , Blastocisto/metabolismo , Diferenciación Celular , Organogénesis , Animales , Animales Modificados Genéticamente , Biomarcadores , Diferenciación Celular/genética , Clonación de Organismos , Feto , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Proteínas de Homeodominio , Organogénesis/genética , Páncreas/embriología , Fenotipo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Regeneración , Porcinos , Transactivadores/deficiencia
16.
Sci Rep ; 9(1): 8016, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142767

RESUMEN

To combat organ shortage in transplantation medicine, a novel strategy has been proposed to generate human organs from exogenous pluripotent stem cells utilizing the developmental mechanisms of pig embryos/foetuses. Genetically modified pigs missing specific organs are key elements in this strategy. In this study, we demonstrate the feasibility of using a genome-editing approach to generate anephrogenic foetuses in a genetically engineered pig model. SALL1 knockout (KO) was successfully induced by injecting genome-editing molecules into the cytoplasm of pig zygotes, which generated the anephrogenic phenotype. Extinguished SALL1 expression and marked dysgenesis of nephron structures were observed in the rudimentary kidney tissue of SALL1-KO foetuses. Biallelic KO mutations of the target gene induced nephrogenic defects; however, biallelic mutations involving small in-frame deletions did not induce the anephrogenic phenotype. Through production of F1 progeny from mutant founder pigs, we identified mutations that could reliably induce the anephrogenic phenotype and hence established a line of fertile SALL1-mutant pigs. Our study lays important technical groundwork for the realization of human kidney regeneration through the use of an empty developmental niche in pig foetuses.


Asunto(s)
Animales Modificados Genéticamente , Edición Génica/métodos , Nefronas/crecimiento & desarrollo , Ingeniería de Tejidos/métodos , Factores de Transcripción/genética , Aloinjertos/provisión & distribución , Animales , Sistemas CRISPR-Cas/genética , Estudios de Factibilidad , Femenino , Desarrollo Fetal/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Trasplante de Riñón , Masculino , Mutación , Células Madre Pluripotentes/fisiología , Regeneración/fisiología , Sus scrofa , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Cigoto/crecimiento & desarrollo
17.
Proc Natl Acad Sci U S A ; 115(4): 708-713, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311328

RESUMEN

Genetically engineered pigs play an indispensable role in the study of rare monogenic diseases. Pigs harboring a gene responsible for a specific disease can be efficiently generated via somatic cell cloning. The generation of somatic cell-cloned pigs from male cells with mutation(s) in an X chromosomal gene is a reliable and straightforward method for reproducing X-linked genetic diseases (XLGDs) in pigs. However, the severe symptoms of XLGDs are often accompanied by impaired growth and reproductive disorders, which hinder the reproduction of these valuable model animals. Here, we generated unique chimeric boars composed of mutant cells harboring a lethal XLGD and normal cells. The chimeric boars exhibited the cured phenotype with fertility while carrying and transmitting the genotype of the XLGD. This unique reproduction system permits routine production of XLGD model pigs through the male-based breeding, thereby opening an avenue for translational research using disease model pigs.


Asunto(s)
Técnicas de Cultivo de Embriones/métodos , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Reproducción/genética , Animales , Animales Modificados Genéticamente/genética , Cruzamiento , Quimera , Clonación de Organismos/métodos , Modelos Animales de Enfermedad , Fertilidad , Técnicas de Inactivación de Genes/métodos , Ingeniería Genética/métodos , Masculino , Técnicas de Transferencia Nuclear , Porcinos/genética
18.
J Reprod Dev ; 62(2): 219-23, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26875691

RESUMEN

A novel hollow fiber vitrification (HFV) method was applied to materials that have previously been difficult to cryopreserve, thereby expanding the potential application of this method. The results showed that zona-free porcine morulae and their isolated blastomeres remained viable even after vitrification. The rate of development to blastocysts after vitrification was similar for zona-free and zona-intact morulae (21/23, 91.3% for both). Vitrified blastomeres had a developmental potential equal to that of non-vitrified blastomeres (blastocyst formation rate after reaggregation: 16/17, 94.1% for both). The HFV method was also effective for the cryopreservation of in vitro matured/fertilized bovine embryos at the 2- to 4-cell, 8- to 16-cell and morula stages. The blastocyst formation rates of vitrified embryos (66.1-82.5%) were similar to those of non-vitrified embryos (74.5-82.5%). These results indicate that this novel HFV method is an effective tool for embryo cryopreservation that can enhance current practices in reproductive biology.


Asunto(s)
Criopreservación/métodos , Vitrificación , Animales , Blastocisto/citología , Blastómeros/citología , Blastómeros/ultraestructura , Bovinos , Células del Cúmulo/citología , Transferencia de Embrión , Desarrollo Embrionario , Femenino , Fertilización In Vitro , Mórula/citología , Oocitos/citología , Porcinos , Temperatura , Factores de Tiempo
19.
Proc Natl Acad Sci U S A ; 112(42): 12980-5, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26392557

RESUMEN

There have been several recent attempts to generate, de novo, a functional whole kidney from stem cells using the organogenic niche or blastocyst complementation methods. However, none of these attempts succeeded in constructing a urinary excretion pathway for the stem cell-generated embryonic kidney. First, we transplanted metanephroi from cloned pig fetuses into gilts; the metanephroi grew to about 3 cm and produced urine, although hydronephrosis eventually was observed because of the lack of an excretion pathway. Second, we demonstrated the construction of urine excretion pathways in rats. Rat metanephroi or metanephroi with bladders (developed from cloacas) were transplanted into host rats. Histopathologic analysis showed that tubular lumina dilation and interstitial fibrosis were reduced in kidneys developed from cloacal transplants compared with metanephroi transplantation. Then we connected the host animal's ureter to the cloacal-developed bladder, a technique we called the "stepwise peristaltic ureter" (SWPU) system. The application of the SWPU system avoided hydronephrosis and permitted the cloacas to differentiate well, with cloacal urine being excreted persistently through the recipient ureter. Finally, we demonstrated a viable preclinical application of the SWPU system in cloned pigs. The SWPU system also inhibited hydronephrosis in the pig study. To our knowledge, this is the first report showing that the SWPU system may resolve two important problems in the generation of kidneys from stem cells: construction of a urine excretion pathway and continued growth of the newly generated kidney.


Asunto(s)
Células Madre Embrionarias/citología , Riñón/fisiología , Orina , Animales , Riñón/embriología , Masculino , Ratas , Ratas Endogámicas Lew , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...