Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Ann Neurol ; 94(5): 969-986, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37526361

RESUMEN

OBJECTIVE: GM2 gangliosidosis is usually fatal by 5 years of age in its 2 major subtypes, Tay-Sachs and Sandhoff disease. First reported in 1881, GM2 gangliosidosis has no effective treatment today, and children succumb to the disease after a protracted neurodegenerative course and semi-vegetative state. This study seeks to further develop adeno-associated virus (AAV) gene therapy for human translation. METHODS: Cats with Sandhoff disease were treated by intracranial injection of vectors expressing feline ß-N-acetylhexosaminidase, the enzyme deficient in GM2 gangliosidosis. RESULTS: Hexosaminidase activity throughout the brain and spinal cord was above normal after treatment, with highest activities at the injection sites (thalamus and deep cerebellar nuclei). Ganglioside storage was reduced throughout the brain and spinal cord, with near complete clearance in many regions. While untreated cats with Sandhoff disease lived for 4.4 ± 0.6 months, AAV-treated cats lived to 19.1 ± 8.6 months, and 3 of 9 cats lived >21 months. Correction of the central nervous system was so effective that significant increases in lifespan led to the emergence of otherwise subclinical peripheral disease, including megacolon, enlarged stomach and urinary bladder, soft tissue spinal cord compression, and patellar luxation. Throughout the gastrointestinal tract, neurons of the myenteric and submucosal plexuses developed profound pathology, demonstrating that the enteric nervous system was inadequately treated. INTERPRETATION: The vector formulation in the current study effectively treats neuropathology in feline Sandhoff disease, but whole-body targeting will be an important consideration in next-generation approaches. ANN NEUROL 2023;94:969-986.


Asunto(s)
Gangliosidosis GM2 , Enfermedad de Sandhoff , Niño , Animales , Gatos , Humanos , Enfermedad de Sandhoff/genética , Enfermedad de Sandhoff/terapia , Enfermedad de Sandhoff/veterinaria , Insuficiencia Multiorgánica/terapia , Vectores Genéticos , Sistema Nervioso Central/patología , Terapia Genética
2.
Brain ; 145(2): 655-669, 2022 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-34410345

RESUMEN

GM1 gangliosidosis is a fatal neurodegenerative disease caused by a deficiency of lysosomal ß-galactosidase. In its most severe form, GM1 gangliosidosis causes death by 4 years of age, and no effective treatments exist. Previous work has shown that injection of the brain parenchyma with an adeno-associated viral (AAV) vector provides pronounced therapeutic benefit in a feline GM1 model. To develop a less invasive treatment for the brain and increase systemic biodistribution, intravenous injection of AAV9 was evaluated. AAV9 expressing feline ß-galactosidase was intravenously administered at 1.5×1013 vector genomes/kg body weight to six GM1 cats at ∼1 month of age. The animals were divided into two cohorts: (i) a long-term group, which was followed to humane end point; and (ii) a short-term group, which was analysed 16 weeks post-treatment. Clinical assessments included neurological exams, CSF and urine biomarkers, and 7 T MRI and magentic resonance spectroscopy (MRS). Post-mortem analysis included ß-galactosidase and virus distribution, histological analysis and ganglioside content. Untreated GM1 animals survived 8.0 ± 0.6 months while intravenous treatment increased survival to an average of 3.5 years (n = 2) with substantial improvements in quality of life and neurological function. Neurological abnormalities, which in untreated animals progress to the inability to stand and debilitating neurological disease by 8 months of age, were mild in all treated animals. CSF biomarkers were normalized, indicating decreased CNS cell damage in the treated animals. Urinary glycosaminoglycans decreased to normal levels in the long-term cohort. MRI and MRS showed partial preservation of the brain in treated animals, which was supported by post-mortem histological evaluation. ß-Galactosidase activity was increased throughout the CNS, reaching carrier levels in much of the cerebrum and normal levels in the cerebellum, spinal cord and CSF. Ganglioside accumulation was significantly reduced by treatment. Peripheral tissues such as heart, skeletal muscle, and sciatic nerve also had normal ß-galactosidase activity in treated GM1 cats. GM1 histopathology was largely corrected with treatment. There was no evidence of tumorigenesis or toxicity. Restoration of ß-galactosidase activity in the CNS and peripheral organs by intravenous gene therapy led to profound increases in lifespan and quality of life in GM1 cats. These data support the promise of intravenous gene therapy as a safe, effective treatment for GM1 gangliosidosis.


Asunto(s)
Gangliosidosis GM1 , Enfermedades Neurodegenerativas , Animales , Biomarcadores , Gatos , Dependovirus/genética , Gangliósido G(M1)/uso terapéutico , Gangliósidos , Gangliosidosis GM1/genética , Gangliosidosis GM1/patología , Gangliosidosis GM1/terapia , Terapia Genética/métodos , Humanos , Calidad de Vida , Distribución Tisular , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
3.
Mol Ther ; 25(4): 892-903, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28236574

RESUMEN

GM1 gangliosidosis is a fatal neurodegenerative disease that affects individuals of all ages. Favorable outcomes using adeno-associated viral (AAV) gene therapy in GM1 mice and cats have prompted consideration of human clinical trials, yet there remains a paucity of objective biomarkers to track disease status. We developed a panel of biomarkers using blood, urine, cerebrospinal fluid (CSF), electrodiagnostics, 7 T MRI, and magnetic resonance spectroscopy in GM1 cats-either untreated or AAV treated for more than 5 years-and compared them to markers in human GM1 patients where possible. Significant alterations were noted in CSF and blood of GM1 humans and cats, with partial or full normalization after gene therapy in cats. Gene therapy improved the rhythmic slowing of electroencephalograms (EEGs) in GM1 cats, a phenomenon present also in GM1 patients, but nonetheless the epileptiform activity persisted. After gene therapy, MR-based analyses revealed remarkable preservation of brain architecture and correction of brain metabolites associated with microgliosis, neuroaxonal loss, and demyelination. Therapeutic benefit of AAV gene therapy in GM1 cats, many of which maintain near-normal function >5 years post-treatment, supports the strong consideration of human clinical trials, for which the biomarkers described herein will be essential for outcome assessment.


Asunto(s)
Biomarcadores , Gangliosidosis GM1/genética , Gangliosidosis GM1/metabolismo , Terapia Genética , Animales , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/orina , Gatos , Dependovirus/clasificación , Dependovirus/genética , Modelos Animales de Enfermedad , Electroencefalografía , Gangliosidosis GM1/mortalidad , Gangliosidosis GM1/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Hipocalcemia/metabolismo , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Resultado del Tratamiento
4.
Mol Genet Metab ; 116(1-2): 80-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25971245

RESUMEN

Sandhoff disease (SD) is a fatal neurodegenerative disease caused by a mutation in the enzyme ß-N-acetylhexosaminidase. Children with infantile onset SD develop seizures, loss of motor tone and swallowing problems, eventually reaching a vegetative state with death typically by 4years of age. Other symptoms include vertebral gibbus and cardiac abnormalities strikingly similar to those of the mucopolysaccharidoses. Isolated fibroblasts from SD patients have impaired catabolism of glycosaminoglycans (GAGs). To evaluate mucopolysaccharidosis-like features of the feline SD model, we utilized radiography, MRI, echocardiography, histopathology and GAG quantification of both central nervous system and peripheral tissues/fluids. The feline SD model exhibits cardiac valvular and structural abnormalities, skeletal changes and spinal cord compression that are consistent with accumulation of GAGs, but are much less prominent than the severe neurologic disease that defines the humane endpoint (4.5±0.5months). Sixteen weeks after intracranial AAV gene therapy, GAG storage was cleared in the SD cat cerebral cortex and liver, but not in the heart, lung, skeletal muscle, kidney, spleen, pancreas, small intestine, skin, or urine. GAG storage worsens with time and therefore may become a significant source of pathology in humans whose lives are substantially lengthened by gene therapy or other novel treatments for the primary, neurologic disease.


Asunto(s)
Terapia Genética , Enfermedad de Sandhoff/genética , Enfermedad de Sandhoff/terapia , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/uso terapéutico , Adenoviridae/genética , Estructuras Animales/patología , Animales , Gatos , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Mucopolisacaridosis/genética , Mucopolisacaridosis/patología , Mucopolisacaridosis/terapia , Fenotipo , Enfermedad de Sandhoff/fisiopatología , Enfermedad de Sandhoff/orina
5.
Mol Ther ; 21(7): 1306-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23689599

RESUMEN

Salutary responses to adeno-associated viral (AAV) gene therapy have been reported in the mouse model of Sandhoff disease (SD), a neurodegenerative lysosomal storage disease caused by deficiency of ß-N-acetylhexosaminidase (Hex). While untreated mice reach the humane endpoint by 4.1 months of age, mice treated by a single intracranial injection of vectors expressing human hexosaminidase may live a normal life span of 2 years. When treated with the same therapeutic vectors used in mice, two cats with SD lived to 7.0 and 8.2 months of age, compared with an untreated life span of 4.5 ± 0.5 months (n = 11). Because a pronounced humoral immune response to both the AAV1 vectors and human hexosaminidase was documented, feline cDNAs for the hexosaminidase α- and ß-subunits were cloned into AAVrh8 vectors. Cats treated with vectors expressing feline hexosaminidase produced enzymatic activity >75-fold normal at the brain injection site with little evidence of an immune infiltrate. Affected cats treated with feline-specific vectors by bilateral injection of the thalamus lived to 10.4 ± 3.7 months of age (n = 3), or 2.3 times as long as untreated cats. These studies support the therapeutic potential of AAV vectors for SD and underscore the importance of species-specific cDNAs for translational research.


Asunto(s)
Enfermedades de los Gatos/enzimología , Enfermedades de los Gatos/terapia , Enfermedad de Sandhoff/enzimología , Enfermedad de Sandhoff/terapia , beta-N-Acetilhexosaminidasas/metabolismo , Animales , Enfermedades de los Gatos/genética , Gatos , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos/genética , Enfermedad de Sandhoff/genética , beta-N-Acetilhexosaminidasas/genética
6.
Mol Ther Methods Clin Dev ; 17: 258-270, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31970203

RESUMEN

GM1 gangliosidosis (GM1) is a fatal neurodegenerative lysosomal storage disease that occurs most commonly in young children, with no effective treatment available. Long-term follow-up of GM1 cats treated by bilateral thalamic and deep cerebellar nuclei (DCN) injection of adeno-associated virus (AAV)-mediated gene therapy has increased lifespan to 8 years of age, compared with an untreated lifespan of ~8 months. Due to risks associated with cerebellar injection in humans, the lateral ventricle was tested as a replacement route to deliver an AAVrh8 vector expressing feline ß-galactosidase (ß-gal), the defective enzyme in GM1. Treatment via the thalamus and lateral ventricle corrected storage, myelination, astrogliosis, and neuronal morphology in areas where ß-gal was effectively delivered. Oligodendrocyte number increased, but only in areas where myelination was corrected. Reduced AAV and ß-gal distribution were noted in the cerebellum with subsequent increases in storage, demyelination, astrogliosis, and neuronal degeneration. These postmortem findings were correlated with endpoint MRI and magnetic resonance spectroscopy (MRS). Compared with the moderate dose with which most cats were treated, a higher AAV dose produced superior survival, currently 6.5 years. Thus, MRI and MRS can predict therapeutic efficacy of AAV gene therapy and non-invasively monitor cellular events within the GM1 brain.

7.
Metabolism ; 57(3): 404-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18249215

RESUMEN

Niacin has recently been shown to increase serum total concentrations of the adipocyte-derived protein adiponectin. Adiponectin possesses important vascular anti-inflammatory and metabolic properties that have been attributed to the active high-molecular weight (HMW) complex of the protein. Our purpose was to examine the influence of extended-release niacin on the distribution of HMW and low-molecular weight (LMW) adiponectin complexes. Fifteen men with the metabolic syndrome were treated for 6 weeks with extended-release niacin. Serum total adiponectin concentrations increased by 46% after the niacin intervention (P < .05). High-molecular weight adiponectin accounted for 63% of the increase in total adiponectin, which was reflected by a shift in the HMW/LMW adiponectin ratio from 0.69 to 0.86 (+25%) (P < .05). Serum insulin concentrations increased by 20% after the niacin intervention despite an increase in HMW adiponectin concentrations (P < .05). These results suggest that the increase in total adiponectin concentrations observed with extended-release niacin is primarily due to an increase in the active HMW complex. Therefore, at least part of the cardioprotective benefits of niacin may be attributed to a shift in the HMW/LMW adiponectin ratio in obese men with the metabolic syndrome.


Asunto(s)
Adiponectina/sangre , Hipolipemiantes/farmacología , Niacina/farmacología , Adiponectina/química , Adulto , Composición Corporal/fisiología , Peso Corporal/fisiología , Enfermedades Cardiovasculares/sangre , Preparaciones de Acción Retardada , Diabetes Mellitus Tipo 2/sangre , Humanos , Hipolipemiantes/administración & dosificación , Resistencia a la Insulina/fisiología , Masculino , Síndrome Metabólico/sangre , Persona de Mediana Edad , Peso Molecular , Niacina/administración & dosificación
8.
Hum Gene Ther ; 29(3): 312-326, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28922945

RESUMEN

Tay-Sachs disease (TSD) is a fatal neurodegenerative disorder caused by a deficiency of the enzyme hexosaminidase A (HexA). TSD also occurs in sheep, the only experimental model of TSD that has clinical signs of disease. The natural history of sheep TSD was characterized using serial neurological evaluations, 7 Tesla magnetic resonance imaging, echocardiograms, electrodiagnostics, and cerebrospinal fluid biomarkers. Intracranial gene therapy was also tested using AAVrh8 monocistronic vectors encoding the α-subunit of Hex (TSD α) or a mixture of two vectors encoding both the α and ß subunits separately (TSD α + ß) injected at high (1.3 × 1013 vector genomes) or low (4.2 × 1012 vector genomes) dose. Delay of symptom onset and/or reduction of acquired symptoms were noted in all adeno-associated virus-treated sheep. Postmortem evaluation showed superior HexA and vector genome distribution in the brain of TSD α + ß sheep compared to TSD α sheep, but spinal cord distribution was low in all groups. Isozyme analysis showed superior HexA formation after treatment with both vectors (TSD α + ß), and ganglioside clearance was most widespread in the TSD α + ß high-dose sheep. Microglial activation and proliferation in TSD sheep-most prominent in the cerebrum-were attenuated after gene therapy. This report demonstrates therapeutic efficacy for TSD in the sheep brain, which is on the same order of magnitude as a child's brain.


Asunto(s)
Dependovirus , Terapia Genética , Enfermedad de Tay-Sachs/terapia , Cadena alfa de beta-Hexosaminidasa/biosíntesis , Cadena beta de beta-Hexosaminidasa/biosíntesis , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/enzimología , Modelos Animales de Enfermedad , Ecocardiografía , Humanos , Imagen por Resonancia Magnética , Microglía/enzimología , Ovinos , Enfermedad de Tay-Sachs/diagnóstico por imagen , Enfermedad de Tay-Sachs/enzimología , Enfermedad de Tay-Sachs/genética , Cadena alfa de beta-Hexosaminidasa/genética , Cadena beta de beta-Hexosaminidasa/genética
9.
Am J Vet Res ; 68(1): 57-62, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17199419

RESUMEN

OBJECTIVE: To assess serum concentrations of adiponectin and characterize adiponectin protein complexes in healthy dogs. ANIMALS: 11 healthy dogs. PROCEDURES: Sera collected from 10 dogs were evaluated via velocity sedimentation and ultracentrifugation, SDS-PAGE, western immunoblotting, and radioimmunoassay. Visceral adipose tissue (approx 90 g) was collected from the falciform ligament of a healthy dog undergoing elective ovariohysterectomy, and adiponectin gene expression was assessed via a real-time PCR procedure. RESULTS: Adiponectin gene expression was detected in visceral adipose tissue. Serum adiponectin concentrations ranged from 0.85 to 1.5 microg/mL (mean concentration, 1.22 microg/mL). In canine serum, adiponectin was present as a multimer, consisting of a low-molecular-weight complex (180 kd); as 3 (180-, 90-, and 60-kd) complexes under denaturing conditions; as 2 (90- and 60-kd) complexes under reducing conditions; and as a dimer, a monomer, and globular head region (60, 30, and 28 kd, respectively) under reducing-denaturing conditions. It is likely that adiponectin also circulates as a high-molecular-weight (360- to 540-kd) complex in canine serum, but resolution of this complex was not possible via SDS-PAGE. CONCLUSIONS AND CLINICAL RELEVANCE: After exposure to identical experimental conditions, adiponectin protein complexes in canine serum were similar to those detected in human and rodent sera. Circulating adiponectin concentrations in canine serum were slightly lower than concentrations in human serum. Adiponectin gene expression was identified in canine visceral adipose tissue. Results suggest that adiponectin could be used as an early clinical marker for metabolic derangements, including obesity, insulin resistance, and diabetes mellitus in dogs.


Asunto(s)
Adiponectina/sangre , Perros/sangre , Adiponectina/biosíntesis , Adiponectina/genética , Tejido Adiposo Blanco/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting/veterinaria , Femenino , Masculino , Datos de Secuencia Molecular , Radioinmunoensayo/veterinaria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Alineación de Secuencia
10.
Neuroscience ; 340: 117-125, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27793778

RESUMEN

Sandhoff disease (SD) is a lysosomal storage disorder characterized by the absence of hydrolytic enzyme ß-N-acetylhexosaminidase (Hex), which results in storage of GM2 ganglioside in neurons and unremitting neurodegeneration. Neuron loss initially affects fine motor skills, but rapidly progresses to loss of all body faculties, a vegetative state, and death by five years of age in humans. A well-established feline model of SD allows characterization of the disease in a large animal model and provides a means to test the safety and efficacy of therapeutic interventions before initiating clinical trials. In this study, we demonstrate a robust central nervous system (CNS) inflammatory response in feline SD, primarily marked by expansion and activation of the microglial cell population. Quantification of major histocompatibility complex II (MHC-II) labeling revealed significant up-regulation throughout the CNS with areas rich in white matter most severely affected. Expression of the leukocyte chemokine macrophage inflammatory protein-1 alpha (MIP-1α) was also up-regulated in the brain. SD cats were treated with intracranial delivery of adeno-associated viral (AAV) vectors expressing feline Hex, with a study endpoint 16weeks post treatment. AAV-mediated gene delivery repressed the expansion and activation of microglia and normalized MHC-II and MIP-1α levels. These data reiterate the profound inflammatory response in SD and show that neuroinflammation is abrogated after AAV-mediated restoration of enzymatic activity.


Asunto(s)
Encéfalo/inmunología , Terapia Genética , Enfermedad de Sandhoff/inmunología , Enfermedad de Sandhoff/terapia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Astrocitos/inmunología , Astrocitos/patología , Encéfalo/patología , Gatos , Dependovirus/genética , Modelos Animales de Enfermedad , Genes MHC Clase II/fisiología , Vectores Genéticos , Gliosis/inmunología , Gliosis/patología , Gliosis/terapia , Inmunohistoquímica , Microglía/inmunología , Microglía/patología , Neuronas/inmunología , Neuronas/patología , Reacción en Cadena de la Polimerasa , Enfermedad de Sandhoff/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Exp Neurol ; 263: 102-12, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25284324

RESUMEN

The GM2 gangliosidoses, Tay-Sachs disease (TSD) and Sandhoff disease (SD), are progressive neurodegenerative disorders that are caused by a mutation in the enzyme ß-N-acetylhexosaminidase (Hex). Due to the recent emergence of novel experimental treatments, biomarker development has become particularly relevant in GM2 gangliosidosis as an objective means to measure therapeutic efficacy. Here we describe blood, cerebrospinal fluid (CSF), magnetic resonance imaging (MRI), and electrodiagnostic methods for evaluating disease progression in the feline SD model and application of these approaches to assess AAV-mediated gene therapy. SD cats were treated by intracranial injections of the thalami combined with either the deep cerebellar nuclei or a single lateral ventricle using AAVrh8 vectors encoding feline Hex. Significantly altered in untreated SD cats, blood and CSF based biomarkers were largely normalized after AAV gene therapy. Also reduced after treatment were expansion of the lysosomal compartment in peripheral blood mononuclear cells and elevated activity of secondary lysosomal enzymes. MRI changes characteristic of the gangliosidoses were documented in SD cats and normalized after AAV gene therapy. The minimally invasive biomarkers reported herein should be useful to assess disease progression of untreated SD patients and those in future clinical trials.


Asunto(s)
Biomarcadores/análisis , Modelos Animales de Enfermedad , Terapia Genética/métodos , Enfermedad de Sandhoff/sangre , Enfermedad de Sandhoff/líquido cefalorraquídeo , Animales , Encéfalo/patología , Gatos , Dependovirus , Progresión de la Enfermedad , Vectores Genéticos , Leucocitos Mononucleares/patología , Lisosomas/patología , Imagen por Resonancia Magnética , Enfermedad de Sandhoff/patología , beta-N-Acetilhexosaminidasas/administración & dosificación , beta-N-Acetilhexosaminidasas/genética
12.
Sci Transl Med ; 6(231): 231ra48, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24718858

RESUMEN

Progressive debilitating neurological defects characterize feline G(M1) gangliosidosis, a lysosomal storage disease caused by deficiency of lysosomal ß-galactosidase. No effective therapy exists for affected children, who often die before age 5 years. An adeno-associated viral vector carrying the therapeutic gene was injected bilaterally into two brain targets (thalamus and deep cerebellar nuclei) of a feline model of G(M1) gangliosidosis. Gene therapy normalized ß-galactosidase activity and storage throughout the brain and spinal cord. The mean survival of 12 treated G(M1) animals was >38 months, compared to 8 months for untreated animals. Seven of the eight treated animals remaining alive demonstrated normalization of disease, with abrogation of many symptoms including gait deficits and postural imbalance. Sustained correction of the G(M1) gangliosidosis disease phenotype after limited intracranial targeting by gene therapy in a large animal model suggests that this approach may be useful for treating the human version of this lysosomal storage disorder.


Asunto(s)
Encéfalo/patología , Terapia Genética , Enfermedades del Sistema Nervioso/terapia , Animales , Cruzamiento , Gatos , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Lisosomas/enzimología , Imagen por Resonancia Magnética , Masculino , Especificidad de Órganos , Análisis de Supervivencia , beta-Galactosidasa/genética , beta-Galactosidasa/uso terapéutico
13.
Biochem Biophys Res Commun ; 345(1): 332-9, 2006 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-16682005

RESUMEN

Adiponectin is an adipokine with profound insulin-sensitizing, anti-inflammatory, and anti-atherogenic properties. Plasma levels of adiponectin are reduced in insulin resistant states such as obesity, type 2 diabetes and cardiovascular disease. However, the mechanism(s) by which adiponectin concentrations are decreased during disease development is unclear. Studies have shown that endothelin-1 (ET-1), a vasoconstrictor peptide, affects adipocyte glucose metabolism and secretion of adipokines such as leptin, resistin, and adiponectin. The goal of our study was to determine the mechanism by which ET-1 decreases adiponectin secretion. 3T3-L1 adipocytes were treated for 24h with ET-1 (10nM) and then stimulated with vehicle or insulin (100 nM) for a period of 1-2h. Chronic ET-1 (24h) treatment significantly decreased basal and insulin-stimulated adiponectin secretion by 66% and 47%, respectively. Inhibition of phosphatidylinositol 4,5-bisphosphate (PIP(2)) hydrolysis by the PLCbeta inhibitor, U73122, or exogenous addition of PIP(2):histone carrier complex (1.25:0.625 microM) ameliorated the decrease in basal and insulin-stimulated adiponectin secretion observed with ET-1. However, treatment with exogenous PIP(2):histone carrier complex and the actin depolymerizing agent latrunculin B (20 microM) did not reverse the ET-1-mediated decrease in adiponectin secretion. In conclusion, we demonstrate that ET-1 inhibits basal and insulin-stimulated adiponectin secretion through PIP(2) modulation of the actin cytoskeleton.


Asunto(s)
Actinas/metabolismo , Adipocitos/metabolismo , Adiponectina/metabolismo , Endotelina-1/farmacología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Transducción de Señal/fisiología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Ratones , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA