Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Ann Neurol ; 74(2): 297-300, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23913475

RESUMEN

Lafora disease (LD) is a fatal progressive myoclonus epilepsy characterized neuropathologically by aggregates of abnormally structured glycogen and proteins (Lafora bodies [LBs]), and neurodegeneration. Whether LBs could be prevented by inhibiting glycogen synthesis and whether they are pathogenic remain uncertain. We genetically eliminated brain glycogen synthesis in LD mice. This resulted in long-term prevention of LB formation, neurodegeneration, and seizure susceptibility. This study establishes that glycogen synthesis is requisite for LB formation and that LBs are pathogenic. It opens a therapeutic window for potential treatments in LD with known and future small molecule inhibitors of glycogen synthesis.


Asunto(s)
Glucógeno/antagonistas & inhibidores , Glucógeno/biosíntesis , Enfermedad de Lafora/prevención & control , Animales , Modelos Animales de Enfermedad , Fosfatasas de Especificidad Dual/genética , Técnicas de Inactivación de Genes , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Enfermedad de Lafora/patología , Enfermedad de Lafora/fisiopatología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Fosfatasas no Receptoras
2.
J Vis Exp ; (193)2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-37602854

RESUMEN

ARTICLES DISCUSSED: Wilson, W. A. Spectrophotometric methods for the study of eukaryotic glycogen metabolism. Journal of Visualized Experiments. (174), e63046 (2021). Wang, J. J. et al. A non-degradative extraction method for molecular structure characterization of bacterial glycogen particles. Journal of Visualized Experiments. (180), e63016 (2022). Wang, Z., Liu, Q., Wang, L., Gilbert, R. G., Sullivan, M. A. The extraction of liver glycogen molecules for glycogen structure determination. Journal of Visualized Experiments. (180), e63088 (2022). Jensen, R., Ortenblad, N., di Benedetto, C., Qvortrup, K., Nielsen, J. Quantification of subcellular glycogen distribution in skeletal muscle fibers using transmission electron microscopy. Journal of Visualized Experiments. (180), e63347 (2022). Fermont, L., Szydlowski, N., Colleoni, C. Determination of glucan chain length distribution of glycogen using the fluorophore-assisted carbohydrate electrophoresis (FACE) method. Journal of Visualized Experiments. (181), e63392 (2022).


Asunto(s)
Eucariontes , Células Eucariotas , Citoplasma , Colorantes Fluorescentes , Glucógeno
3.
J Neurosci Res ; 89(4): 585-91, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21259334

RESUMEN

Brain glycogen is proposed to function under both physiological and pathological conditions. Pharmacological elevation of this glucose polymer in brain is hypothesized to protect neurons against hypoglycemia-induced cell death. Elevation of brain glycogen levels due to prior hypoglycemia is postulated to contribute to the development of hypoglycemia-associated autonomic failure (HAAF) in insulin-treated diabetic patients. This latter mode of elevating glycogen levels is termed "supercompensation." We tested whether brain glycogen supercompensation occurs in healthy, conscious mice after recovery from insulin-induced acute or recurrent hypoglycemia. Blood glucose levels were lowered to less than 2.2 mmol/liter for 90 min by administration of insulin. Brain glucose levels decreased at least 80% and brain glycogen levels decreased approximately 50% after episodes of either acute or recurrent hypoglycemia. After these hypoglycemic episodes, mice were allowed access to food for 6 or 27 hr. After 6 hr, blood and brain glucose levels were restored but brain glycogen levels were elevated by 25% in mice that had been subjected to either acute or recurrent hypoglycemia compared with saline-treated controls. After a 27-hr recovery period, the concentration of brain glycogen had returned to baseline levels in mice previously subjected to either acute or recurrent hypoglycemia. We conclude that brain glycogen supercompensation occurs in healthy mice, but its functional significance remains to be established.


Asunto(s)
Encéfalo/metabolismo , Glucógeno/metabolismo , Hipoglucemia/metabolismo , Animales , Encéfalo/fisiopatología , Glucosa/metabolismo , Hipoglucemia/inducido químicamente , Hipoglucemia/fisiopatología , Hipoglucemiantes/farmacología , Insulina/farmacología , Masculino , Ratones
4.
Public Health ; 124(6): 326-31, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20483439

RESUMEN

OBJECTIVES: To describe the multidisciplinary investigation and management of a rapidly increasing number of cases of Legionnaires' disease in the North Shropshire area, UK during August 2006. STUDY DESIGN: Epidemiological and environmental investigation of a cluster of cases of Legionnaires' disease. METHODS: Outbreak investigation included: agreeing case definitions; case finding; epidemiological survey; identification and environmental investigation of potential sources; microbiological analysis of clinical and environmental samples; mapping the location of potential sources; and the movement and residence of cases. RESULTS: Three cases of Legionnaires' disease were admitted to a local hospital between 30 and 31 August 2006. Two of these cases were Shropshire residents, with the third living in Wales. A fourth case was also identified which, it was thought, may have been linked to this cluster as the patient had a history of travel to the same area as the two Shropshire residents. Over the next few weeks, three more cases were identified, two of whom were admitted to hospital. Subsequent detailed environmental, epidemiological and microbiological investigation did not support the hypothesis that any of these cases could be linked to a common source. CONCLUSIONS: The results of this investigation strongly suggest that a single source was not responsible for the cluster, and it was concluded that this incident was a pseudo-outbreak. This investigation serves as a reminder that clusters can and do occur, and that an apparent outbreak may be a collection of sporadic cases distinguishable only by rigorous epidemiological, environmental and microbiological investigation.


Asunto(s)
Infecciones Comunitarias Adquiridas/epidemiología , Enfermedad de los Legionarios/epidemiología , Salud Pública/métodos , Adulto , Anciano , Análisis por Conglomerados , Infecciones Comunitarias Adquiridas/microbiología , Brotes de Enfermedades , Humanos , Legionella pneumophila/aislamiento & purificación , Enfermedad de los Legionarios/microbiología , Masculino , Persona de Mediana Edad , Viaje , Reino Unido/epidemiología
5.
Ann Clin Transl Neurol ; 7(11): 2186-2198, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33034425

RESUMEN

OBJECTIVE: Adult polyglucosan body disease (APBD) is an adult-onset neurological variant of glycogen storage disease type IV. APBD is caused by recessive mutations in the glycogen branching enzyme gene, and the consequent accumulation of poorly branched glycogen aggregates called polyglucosan bodies in the nervous system. There are presently no treatments for APBD. Here, we test whether downregulation of glycogen synthesis is therapeutic in a mouse model of the disease. METHODS: We characterized the effects of knocking out two pro-glycogenic proteins in an APBD mouse model. APBD mice were crossed with mice deficient in glycogen synthase (GYS1), or mice deficient in protein phosphatase 1 regulatory subunit 3C (PPP1R3C), a protein involved in the activation of GYS1. Phenotypic and histological parameters were analyzed and glycogen was quantified. RESULTS: APBD mice deficient in GYS1 or PPP1R3C demonstrated improvements in life span, morphology, and behavioral assays of neuromuscular function. Histological analysis revealed a reduction in polyglucosan body accumulation and of astro- and micro-gliosis in the brains of GYS1- and PPP1R3C-deficient APBD mice. Brain glycogen quantification confirmed the reduction in abnormal glycogen accumulation. Analysis of skeletal muscle, heart, and liver found that GYS1 deficiency reduced polyglucosan body accumulation in all three tissues and PPP1R3C knockout reduced skeletal muscle polyglucosan bodies. INTERPRETATION: GYS1 and PPP1R3C are effective therapeutic targets in the APBD mouse model. These findings represent a critical step toward the development of a treatment for APBD and potentially other glycogen storage disease type IV patients.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno/metabolismo , Glucógeno Sintasa/deficiencia , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Enfermedades del Sistema Nervioso/metabolismo , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno/fisiopatología , Enfermedad del Almacenamiento de Glucógeno/terapia , Ratones , Ratones Noqueados , Enfermedades del Sistema Nervioso/fisiopatología , Enfermedades del Sistema Nervioso/terapia
6.
Am J Transplant ; 9(8): 1760-72, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19563344

RESUMEN

The inherent immunosuppressive properties and low immunogenicity of mesenchymal stems cells (MSCs) suggested their therapeutic potential in transplantation. We investigated whether MSCs could prolong allograft survival. Treatment involving infusion of MSCs into BALB/c recipients 24 hours after receiving a heart allograft from a C57BL/6 donor significantly abated rejection and doubled graft mean survival time compared to untreated recipients. Furthermore, combination therapy of MSCs and low-dose Rapamycin (Rapa) achieved long-term heart graft survival (>100 days) with normal histology. The treated recipients readily accepted donor skin grafts but rejected third-party skin grafts, indicating the establishment of tolerance. Tolerant recipients exhibited neither intragraft nor circulating antidonor antibodies, but demonstrated significantly high frequencies of both tolerogenic dendritic cells (Tol-DCs) and CD4(+)CD25(+)Foxp3(+)T cells in the spleens. Infusion of GFP(+)C57BL/6-MSCs in combination with Rapa revealed that the GFP-MSCs accumulated in the lymphoid organs and grafts of tolerant recipients. Thus, engraftment of infused MSCs within the recipient's lymphoid organs and allograft appeared to be instrumental in the induction of allograft-specific tolerance when administered in combination with a subtherapeutic dose of Rapamycin. This study supports the clinical applicability of MSCs in transplantation.


Asunto(s)
Rechazo de Injerto/prevención & control , Trasplante de Corazón/inmunología , Inmunosupresores/uso terapéutico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/patología , Sirolimus/uso terapéutico , Tolerancia al Trasplante/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Movimiento Celular/fisiología , Células Cultivadas , Células Dendríticas/patología , Relación Dosis-Respuesta a Droga , Factores de Transcripción Forkhead/metabolismo , Rechazo de Injerto/inmunología , Trasplante de Corazón/patología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Transgénicos , Modelos Animales
7.
Adv Neurobiol ; 23: 83-123, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31667806

RESUMEN

Brain glycogen synthesis is a regulated, multi-step process that begins with glucose transport across the blood brain barrier and culminates with the actions of glycogen synthase and the glycogen branching enzyme to elongate glucose chains and introduce branch points in a growing glycogen molecule. This review focuses on the synthesis of glycogen in the brain, with an emphasis on glycogen synthase, but draws on salient studies in mammalian muscle and liver as well as baker's yeast, with the goal of providing a more comprehensive view of glycogen synthesis and highlighting potential areas for further study in the brain. In addition, deficiencies in the glycogen biosynthetic enzymes which lead to glycogen storage diseases in humans are discussed, highlighting effects on the brain and discussing findings in genetically modified animal models that recapitulate these diseases. Finally, implications of glycogen synthesis in neurodegenerative and other diseases that impact the brain are presented.


Asunto(s)
Encéfalo/enzimología , Glucógeno Sintasa/química , Glucógeno Sintasa/metabolismo , Animales , Glucosa/metabolismo , Glucógeno/metabolismo , Humanos
8.
Cell Rep ; 29(6): 1410-1418.e6, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693883

RESUMEN

Browning induction or transplantation of brown adipose tissue (BAT) or brown/beige adipocytes derived from progenitor or induced pluripotent stem cells (iPSCs) can represent a powerful strategy to treat metabolic diseases. However, our poor understanding of the mechanisms that govern the differentiation and activation of brown adipocytes limits the development of such therapy. Various genetic factors controlling the differentiation of brown adipocytes have been identified, although most studies have been performed using in vitro cultured pre-adipocytes. We investigate here the differentiation of brown adipocytes from adipose progenitors in the mouse embryo. We demonstrate that the formation of multiple lipid droplets (LDs) is initiated within clusters of glycogen, which is degraded through glycophagy to provide the metabolic substrates essential for de novo lipogenesis and LD formation. Therefore, this study uncovers the role of glycogen in the generation of LDs.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis/genética , Tejido Adiposo Pardo/metabolismo , Embrión de Mamíferos/metabolismo , Glucógeno/metabolismo , Gotas Lipídicas/metabolismo , Adipocitos Marrones/ultraestructura , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/ultraestructura , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Glucógeno/ultraestructura , Humanos , Gotas Lipídicas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , PPAR gamma/genética , PPAR gamma/metabolismo , ARN Interferente Pequeño , Transcriptoma
9.
PLoS Med ; 5(1): e27, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18232732

RESUMEN

BACKGROUND: Stored glycogen is an important source of energy for skeletal muscle. Human genetic disorders primarily affecting skeletal muscle glycogen turnover are well-recognised, but rare. We previously reported that a frameshift/premature stop mutation in PPP1R3A, the gene encoding RGL, a key regulator of muscle glycogen metabolism, was present in 1.36% of participants from a population of white individuals in the UK. However, the functional implications of the mutation were not known. The objective of this study was to characterise the molecular and physiological consequences of this genetic variant. METHODS AND FINDINGS: In this study we found a similar prevalence of the variant in an independent UK white population of 744 participants (1.46%) and, using in vivo (13)C magnetic resonance spectroscopy studies, demonstrate that human carriers (n = 6) of the variant have low basal (65% lower, p = 0.002) and postprandial muscle glycogen levels. Mice engineered to express the equivalent mutation had similarly decreased muscle glycogen levels (40% lower in heterozygous knock-in mice, p < 0.05). In muscle tissue from these mice, failure of the truncated mutant to bind glycogen and colocalize with glycogen synthase (GS) decreased GS and increased glycogen phosphorylase activity states, which account for the decreased glycogen content. CONCLUSIONS: Thus, PPP1R3A C1984DeltaAG (stop codon 668) is, to our knowledge, the first prevalent mutation described that directly impairs glycogen synthesis and decreases glycogen levels in human skeletal muscle. The fact that it is present in approximately 1 in 70 UK whites increases the potential biomedical relevance of these observations.


Asunto(s)
Codón sin Sentido , Mutación del Sistema de Lectura , Glucógeno/biosíntesis , Músculo Esquelético/enzimología , Fosfoproteínas Fosfatasas/fisiología , Adulto , Animales , Diabetes Mellitus Tipo 2/enzimología , Femenino , Frecuencia de los Genes , Glucógeno/análisis , Glucógeno Fosforilasa/metabolismo , Glucógeno Sintasa/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Datos de Secuencia Molecular , Músculo Esquelético/química , Fosfoproteínas Fosfatasas/genética , Periodo Posprandial , Relación Estructura-Actividad , Reino Unido , Población Blanca/genética
10.
Mol Cell Biol ; 24(16): 7179-87, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15282316

RESUMEN

Glycogen serves as a repository of glucose in many mammalian tissues. Mice lacking this glucose reserve in muscle, heart, and several other tissues were generated by disruption of the GYS1 gene, which encodes an isoform of glycogen synthase. Crossing mice heterozygous for the GYS1 disruption resulted in a significant underrepresentation of GYS1-null mice in the offspring. Timed matings established that Mendelian inheritance was followed for up to 18.5 days postcoitum (dpc) and that approximately 90% of GYS1-null animals died soon after birth due to impaired cardiac function. Defects in cardiac development began between 11.5 and 14.5 dpc. At 18.5 dpc, the hearts were significantly smaller, with reduced ventricular chamber size and enlarged atria. Consistent with impaired cardiac function, edema, pooling of blood, and hemorrhagic liver were seen. Glycogen synthase and glycogen were undetectable in cardiac muscle and skeletal muscle from the surviving null mice, and the hearts showed normal morphology and function. Congenital heart disease is one of the most common birth defects in humans, at up to 1 in 50 live births. The results provide the first direct evidence that the ability to synthesize glycogen in cardiac muscle is critical for normal heart development and hence that its impairment could be a significant contributor to congenital heart defects.


Asunto(s)
Glucógeno Sintasa/metabolismo , Glucógeno/metabolismo , Corazón/crecimiento & desarrollo , Miocardio/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Ecocardiografía , Femenino , Glucógeno Sintasa/genética , Corazón/anatomía & histología , Corazón/fisiología , Humanos , Hígado/citología , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/química , Miocardio/citología , Isoformas de Proteínas/genética , Tasa de Supervivencia , Transgenes
11.
Biochem J ; 395(1): 137-45, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16356168

RESUMEN

Glycogen, a branched polymer of glucose, forms an energy re-serve in numerous organisms. In mammals, the two largest glyco-gen stores are in skeletal muscle and liver, which express tissue-specific glycogen synthase isoforms. MGSKO mice, in which mGys1 (mouse glycogen synthase) is disrupted, are devoid of muscle glycogen [Pederson, Chen, Schroeder, Shou, DePaoli-Roach and Roach (2004) Mol. Cell. Biol. 24, 7179-7187]. The GSL30 mouse line hyper-accumulates glycogen in muscle [Manchester, Skurat, Roach, Hauschka and Lawrence (1996) Proc. Natl. Acad. Sci. U.S.A. 93, 10707-10711]. We performed a microarray analysis of mRNA from the anterior tibialis, medial gastrocnemius and liver of MGSKO mice, and from the gastroc-nemius of GSL30 mice. In MGSKO mice, transcripts of 79 genes varied in their expression in the same direction in both the anterior tibialis and gastrocnemius. These included several genes encoding proteins proximally involved in glycogen metabolism. The Ppp1r1a [protein phosphatase 1 regulatory (inhibitor) sub-unit 1A] gene underwent the greatest amount of downregulation. In muscle, the downregulation of Pfkfb1 and Pfkfb3, encoding isoforms of 6-phosphofructo-2-kinase/fructose-2,6-bisphospha-tase, is consistent with decreased glycolysis. Pathways for branched-chain amino acid, and ketone body utilization appear to be downregulated, as is the capacity to form the gluconeogenic precursors alanine, lactate and glutamine. Expression changes among several members of the Wnt signalling pathway were identified, suggesting an as yet unexplained role in glycogen meta-bolism. In liver, the upregulation of Pfkfb1 and Pfkfb3 expression is consistent with increased glycolysis, perhaps as an adaptation to altered muscle metabolism. By comparing changes in muscle expression between MGSKO and GSL30 mice, we found a subset of 44 genes, the expression of which varied as a function of muscle glycogen content. These genes are candidates for regulation by glycogen levels. Particularly interesting is the observation that 11 of these genes encode cardiac or slow-twitch isoforms of muscle contractile proteins, and are upregulated in muscle that has a greater oxidative capacity in MGSKO mice.


Asunto(s)
Perfilación de la Expresión Génica , Glucógeno/metabolismo , Músculo Esquelético/metabolismo , Animales , Regulación de la Expresión Génica , Glucógeno Sintasa/deficiencia , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Músculo Esquelético/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Proteínas Wnt/genética
12.
Diabetes ; 54(12): 3466-73, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16306363

RESUMEN

Glycogen is an important component of whole-body glucose metabolism. MGSKO mice lack skeletal muscle glycogen due to disruption of the GYS1 gene, which encodes muscle glycogen synthase. MGSKO mice were 5-10% smaller than wild-type littermates with less body fat. They have more oxidative muscle fibers and, based on the activation state of AMP-activated protein kinase, more capacity to oxidize fatty acids. Blood glucose in fed and fasted MGSKO mice was comparable to wild-type littermates. Serum insulin was lower in fed but not in fasted MGSKO animals. In a glucose tolerance test, MGSKO mice disposed of glucose more effectively than wild-type animals and had a more sustained elevation of serum insulin. This result was not explained by increased conversion to serum lactate or by enhanced storage of glucose in the liver. However, glucose infusion rate in a euglycemic-hyperinsulinemic clamp was normal in MGSKO mice despite diminished muscle glucose uptake. During the clamp, MGSKO animals accumulated significantly higher levels of liver glycogen as compared with wild-type littermates. Although disruption of the GYS1 gene negatively affects muscle glucose uptake, overall glucose tolerance is actually improved, possibly because of a role for GYS1 in tissues other than muscle.


Asunto(s)
Glucosa/metabolismo , Glucógeno Sintasa/deficiencia , Animales , Grasas de la Dieta/farmacología , Femenino , Prueba de Tolerancia a la Glucosa , Crecimiento , Insulina/sangre , Glucógeno Hepático/metabolismo , Masculino , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/enzimología , Músculo Esquelético/patología
13.
J Neonatal Perinatal Med ; 9(3): 313-6, 2016 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-27589546

RESUMEN

The incidence of perinatal and neonatal Listeriosis is underestimated due undiagnosed stillbirths, misdiagnosis of NL and underreporting of single case reports. Recent outbreaks reinforce the need for better surveillance and targeted health education in certain population groups especially during pregnancy.


Asunto(s)
Queso/envenenamiento , Enfermedades Transmitidas por los Alimentos/diagnóstico , Enfermedades Transmitidas por los Alimentos/epidemiología , Listeriosis/diagnóstico , Listeriosis/epidemiología , Adulto , Ampicilina/administración & dosificación , Antibacterianos/administración & dosificación , Femenino , Enfermedades Transmitidas por los Alimentos/microbiología , Conocimientos, Actitudes y Práctica en Salud , Humanos , Incidencia , Lactante , Recién Nacido , Listeriosis/microbiología , Vigilancia de la Población , Embarazo , Complicaciones Infecciosas del Embarazo/epidemiología , Factores de Riesgo , Estados Unidos
14.
Exp Hematol ; 29(2): 244-55, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11166464

RESUMEN

OBJECTIVE: The human bone marrow contains mesenchymal stem cells capable of differentiating along multiple mesenchymal cell lineages. Using a non-human primate model, we sought to determine whether the systemic infusion of baboon-derived mesenchymal stem cells was associated with toxicity and whether these cells were capable of homing to and persisting within the bone marrow. MATERIALS AND METHODS: Five baboons (Papio anubis) were administered lethal irradiation followed by intravenous autologous hematopoietic progenitor cells combined with either autologous (n = 3) or allogeneic (n = 2) mesenchymal stem cells that had been expanded in culture. In four of these baboons, the mesenchymal stem cells were genetically modified with a retroviral vector encoding either the enhanced green fluorescent protein gene (n = 3) or the human placental alkaline phosphatase gene (n = 1) for tracking purposes. A sixth animal received only intravenous gene marked autologous mesenchymal stem cells but no hematopoietic stem cells or conditioning irradiation. RESULTS: Following culture, baboon mesenchymal stem cells appeared morphologically as a homogeneous population of spindle-shaped cells that were identified by the monoclonal antibodies SH-3 and SH-4. These cells did not express the hematopoietic markers CD34 or CD45. Baboon mesenchymal stem cells isolated from primary culture were capable of differentiating along both adipogenic and osteogenic lineages. There was no acute or chronic toxicity associated with the intravenous infusion of mesenchymal stem cells. In all five recipients of gene marked mesenchymal stem cells, transgene was detected in post-transplant bone marrow biopsies. In two animals receiving autologous mesenchymal stem cells, including the one non-conditioned recipient, transgene could be detected over 1 year following infusion. In one recipient of allogeneic gene marked mesenchymal stem cells, transgene was detected in the bone marrow at 76 days following infusion. CONCLUSION: These data demonstrate that baboon mesenchymal stem cells: 1) are not associated with significant toxicity when administered intravenously, 2) are capable of homing to the bone marrow following intravenous infusion, and 3) have the capacity to establish residence within the bone marrow for an extended duration following systemic administration.


Asunto(s)
Médula Ósea , Mesodermo/citología , Papio , Trasplante de Células Madre , Células Madre/citología , Fosfatasa Alcalina/genética , Animales , Anticuerpos Monoclonales , Antígenos CD34/análisis , Médula Ósea/química , Separación Celular , Células Cultivadas , ADN Recombinante/análisis , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas Fluorescentes Verdes , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Antígenos Comunes de Leucocito/análisis , Proteínas Luminiscentes/genética , Masculino , Mesodermo/inmunología , Reacción en Cadena de la Polimerasa , Transfección , Transgenes
15.
Hum Gene Ther ; 12(12): 1527-41, 2001 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-11506695

RESUMEN

Human mesenchymal stem cells (MSCs) are capable of differentiating into multiple mesenchymal lineages including chondrocytes, osteocytes, adipocytes, and marrow stromal cells. Using a nonhuman primate model, we evaluated nonhuman primate MSCs as targets for gene therapy. Baboon MSCs (bMSCs) cultured from bone marrow aspirates appeared as a homogeneous population of spindle-shaped cells. bMSCs were capable of differentiating into adipocytes and osteocytes in vitro and chondrocytes in vivo. bMSCs were genetically modified with a bicistronic vector encoding the human erythropoietin (hEPO) gene and the green fluorescent protein (GFP) gene. Transduction efficiencies ranged from 72 to 99% after incubation of MSCs with retroviral supernatant. Transduced cells produced from 1.83 x 10(5) to 7.12 x 10(5) mIU of hEPO per 10(6) cells per 24 hr in vitro before implantation. To determine the capacity of bMSCs to express hEPO in vivo, transduced bMSCs were injected intramuscularly in NOD/SCID mice. In a separate experiment, transduced bMSCs were loaded into immunoisolatory devices (IIDs) and surgically implanted into either autologous or allogeneic baboon recipients. Human EPO was detected in the serum of NOD/SCID mice for up to 28 days and in the serum of five baboons for between 9 and 137 days. NOD/SCID mice experienced sharp rises in hematocrit after intramuscular injection of hEPO-transduced bMSCs. The baboon that expressed hEPO for 137 days experienced a statistically significant (p < 0.04) rise in its hematocrit. These data demonstrate that nonhuman primate MSCs can be engineered to deliver a secreted and biologically active gene product. Therefore, human MSCs may be an effective target for future human gene therapy trials.


Asunto(s)
Eritropoyetina/genética , Eritropoyetina/metabolismo , Terapia Genética/métodos , Mesodermo/citología , Mesodermo/metabolismo , Células Madre/citología , Células Madre/metabolismo , Adulto , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes , Hematócrito , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Modelos Genéticos , Papio , Fenotipo , Retroviridae/genética , Factores de Tiempo , Transducción Genética
16.
Transplantation ; 64(5): 709-16, 1997 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9311707

RESUMEN

BACKGROUND: We demonstrated previously that a nonmyeloablative preparative regimen can induce mixed chimerism and allograft tolerance in cynomolgus monkeys. METHODS: The current studies were designed to clarify the importance and toxicity of various elements of the allotolerance conditioning regimen by: fractionating or reducing the whole-body irradiation (WBI) dosage; adding deoxyspergualine; or deleting donor bone marrow, cyclosporine, irradiation, or splenectomy. RESULTS: Monkeys treated without donor bone marrow, cyclosporine, or irradiation did not develop chimerism or long-term allograft survival. One of three monkeys treated without splenectomy developed chimerism but died of a surgical complication. The other two did not develop chimerism and rejected by day 117. Six of six monkeys treated with 300 cGy of fractionated WBI developed chimerism. Five of these recipients had long-term graft survival. Only two of four monkeys treated with 250 cGy developed chimerism, so a 2-week course of deoxyspergualine was added. This led to chimerism in two monkeys, but one died of ureteral stenosis and the other died of allograft rejection. An unanticipated high incidence of ureteral complications felt to be secondary to rejection episodes and ischemic injury was observed in the long-term surviving animals. CONCLUSIONS: All parameters of the original preparative regimen seem to be essential for consistent success. The degree of lymphocyte depletion was proportional to the WBI dose. Long-term graft survival was observed only in recipients achieving lymphocyte chimerism of > 1.5%. In this model, lymphocyte depletion seems to be the best predictor of chimerism, and significant lymphocyte chimerism seems to be important in achieving tolerance.


Asunto(s)
Macaca fascicularis/inmunología , Quimera por Trasplante/fisiología , Acondicionamiento Pretrasplante/métodos , Animales , Ciclosporina/uso terapéutico , Citometría de Flujo , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Guanidinas/uso terapéutico , Tolerancia Inmunológica , Inmunosupresores/uso terapéutico , Trasplante de Riñón/inmunología , Recuento de Leucocitos/efectos de la radiación , Depleción Linfocítica , Masculino , Pancitopenia/etiología , Quimera por Trasplante/inmunología , Trasplante Homólogo , Irradiación Corporal Total/efectos adversos
17.
Transplantation ; 67(6): 915-8, 1999 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-10199743

RESUMEN

Simultaneous pancreas-kidney transplant from living donors has been recently proposed as an effective therapeutic option in selected uremic patients with type I diabetes. We report the first simultaneous pancreas-kidney transplant performed between identical twins. Posttransplant, the recipient has been maintained on low dose cyclosporine to avoid recurrent auto-immune insulitis. At the 1-year follow-up, both donor and recipient are well with normal renal function and excellent glucose control. Simultaneous pancreas-kidney transplant between identical twins can be performed successfully using cyclosporine to prevent recurrent auto-immune insulitis.


Asunto(s)
Enfermedades en Gemelos , Trasplante de Riñón , Trasplante de Páncreas , Gemelos Monocigóticos , Adulto , Nefropatías Diabéticas/cirugía , Femenino , Estudios de Seguimiento , Humanos , Fallo Renal Crónico/cirugía
18.
Transplantation ; 72(10): 1653-5, 2001 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-11726826

RESUMEN

Highly sensitized patients are forced to stay on transplant waiting lists for many years and ultimately may never find a donor. Peripheral blood stem cell (PBSC) transplantation may provide a strategy to decrease host alloreactivity through the production of a chimeric state. We investigated alloreactivity and chimerism in a highly sensitized 40-year-old patient with sickle cell disease who underwent a nonradiation based conditioning regimen consisting of fludarabine, ATG, and high dose melphalan, for allogeneic stem cell transplant. Host monocytes and lymphocytes became donor in origin by day 14. PRA, initially 100% pretransplant, fell to 0 by day 263. Anti-red blood cells antibody became undetectable by day 152. The use of a new nonradiation-based conditioning regimen enabled successful engraftment of allogeneic donor PBSCs and the elimination of alloantibody. As new less toxic conditioning regimens are developed, PBSC transplantation might provide a new solution to allosensitization.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Isoanticuerpos/análisis , Acondicionamiento Pretrasplante , Adulto , Humanos , Pulmón/fisiopatología
19.
Transplantation ; 69(5): 914-7, 2000 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10755550

RESUMEN

The effect of long-term pharmacologic immunosuppression (PI) on anti-Galalpha1-3Gal (alphaGal) antibody (Ab) levels has not been determined previously in humans. In this study, we measured alpha Gal Ab levels by ELISA in 14 healthy volunteers (controls) and in 70 patients with grafts (kidney, heart, liver) who had received different combinations of PI (including cyclosporine, tacrolimus, azathioprine, mycophenolate mofetil, and steroids) for >3 months. There was great variation in Gal IgM (<80-fold) and IgG (<160-fold). There was no difference in Gal IgM or Gal IgG between any one group and any other. In kidney patients with either high (mean 68%) or low (mean 6%) panel-reactive alloantibodies, there was no difference in alpha Gal Ab level or serum cytotoxicity to pig cells. In vitro immunoadsorption of alphaGal Ab from the serum did not change panel-reactive alloantibody positivity. Therapy with OKT3, a mouse product that might stimulate alphaGal Ab production, led to no significant change in patient Ab levels. We conclude that long-term (>3 months) PI does not reduce Gal Ab levels sufficiently to be of clinical value in xenotransplantation.


Asunto(s)
Anticuerpos/análisis , Disacáridos/inmunología , Trasplante de Corazón/inmunología , Terapia de Inmunosupresión , Trasplante de Riñón/inmunología , Trasplante de Hígado/inmunología , Adulto , Anciano , Animales , Sangre/inmunología , Línea Celular/inmunología , Citotoxicidad Inmunológica , Femenino , Humanos , Inmunoglobulina G/análisis , Inmunoglobulina M/análisis , Inmunosupresores/uso terapéutico , Masculino , Ratones , Persona de Mediana Edad , Muromonab-CD3/uso terapéutico , Porcinos/inmunología
20.
Transplantation ; 68(11): 1708-16, 1999 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-10609947

RESUMEN

BACKGROUND: We have previously demonstrated that induction of mixed lymphohematopoietic chimerism resulted in donor specific renal allograft tolerance without the need for chronic immunosuppression in nonhuman primates. Here we have tested whether tolerance can be similarly induced for baboon to cynomolgus renal xenografts. METHODS: After preconditioning with anti-thymocyte globulin (ATG), nonlethal total body irradiation, and thymic irradiation, cynomolgus monkeys underwent splenectomy, native nephrectomies, and baboon marrow and renal transplants. Postoperative cyclosporine was given for 28 days. RESULTS: In Group 1 (n=2, survival= 13, 14 days), both animals developed anti-donor immunoglobulin G, had biopsy findings consistent with humoral rejection, and showed rapidly progressive xenograft failure. In Group 2 (n=5, survival=1, 16, 33, 112, 190 days), 15-deoxyspergualine was added to the regimen (Day 0-13). In one long-term survivor, donor specific hyporesponsiveness was first observed (mixed lymphocyte culture [(MLR]) on Day 48. MLR reactivity returned on Day 64 together with the development of anti-donor antibody and subsequent xenograft failure on Day 112. Donor specific T-cell hyporesponsiveness was detected in the other long-term survivor for the first 133 days, after which a donor-specific skin xenograft was placed, (survival 24 days). Following the skin graft rejection, a rise in the MLR, development of anti-donor antibody and progressive rejection of the renal xenograft were observed. CONCLUSIONS: Antibody-mediated rejection seems to constitute the major difference between concordant xenografts and allografts. Addition of 15-deoxyspergualine for 2 weeks posttransplant extended concordant primate xenograft survival to 6 months without chronic immunosuppression. In contrast to the allogeneic model, renal transplant acceptance in this xenogeneic system was interrupted by placement of a donor-specific skin graft.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Tolerancia Inmunológica/fisiología , Trasplante de Riñón/inmunología , Trasplante Heterólogo/inmunología , Animales , Anticuerpos Heterófilos/análisis , Anticuerpos Monoclonales/farmacología , Femenino , Rechazo de Injerto/inmunología , Supervivencia de Injerto/efectos de los fármacos , Guanidinas/farmacología , Inmunosupresores/farmacología , Macaca fascicularis , Masculino , Ratones , Papio , Trasplante de Piel/inmunología , Linfocitos T/fisiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA