Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 12(547)2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32522805

RESUMEN

Heterozygous mutations of the gene encoding the postsynaptic protein SHANK3 are associated with syndromic forms of autism spectrum disorders (ASDs). One of the earliest clinical symptoms in SHANK3-associated ASD is neonatal skeletal muscle hypotonia. This symptom can be critical for the early diagnosis of affected children; however, the mechanism mediating hypotonia in ASD is not completely understood. Here, we used a combination of patient-derived human induced pluripotent stem cells (hiPSCs), Shank3Δ11(-/-) mice, and Phelan-McDermid syndrome (PMDS) muscle biopsies from patients of different ages to analyze the role of SHANK3 on motor unit development. Our results suggest that the hypotonia in SHANK3 deficiency might be caused by dysfunctions in all elements of the voluntary motor system: motoneurons, neuromuscular junctions (NMJs), and striated muscles. We found that SHANK3 localizes in Z-discs in the skeletal muscle sarcomere and co-immunoprecipitates with α-ACTININ. SHANK3 deficiency lead to shortened Z-discs and severe impairment of acetylcholine receptor clustering in hiPSC-derived myotubes and in muscle from Shank3Δ11(-/-) mice and patients with PMDS, indicating a crucial role for SHANK3 in the maturation of NMJs and striated muscle. Functional motor defects in Shank3Δ11(-/-) mice could be rescued with the troponin activator Tirasemtiv that sensitizes muscle fibers to calcium. Our observations give insight into the function of SHANK3 besides the central nervous system and imply potential treatment strategies for SHANK3-associated ASD.


Asunto(s)
Trastorno Autístico , Células Madre Pluripotentes Inducidas , Animales , Humanos , Ratones , Proteínas de Microfilamentos , Músculo Esquelético , Mutación/genética , Proteínas del Tejido Nervioso/genética , Unión Neuromuscular
2.
Biometals ; 30(5): 643-661, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28717982

RESUMEN

Zn is essential for growth and development. The bioavailability of Zn is affected by several factors such as other food components. It is therefore of interest, to understand uptake mechanisms of Zn delivering compounds to identify ways to bypass the inhibitory effects of these factors. Here, we studied the effect of Zn amino acid conjugates (ZnAAs) on the bioavailabilty of Zn. We used Caco-2 cells and enterocytes differentiated from human induced pluripotent stem cells from a control and Acrodermatitis enteropathica (AE) patient, and performed fluorescence based assays, protein biochemistry and atomic absorption spectrometry to characterize cellular uptake and absorption of ZnAAs. The results show that ZnAAs are taken up by AA transporters, leading to an intracellular enrichment of Zn mostly uninhibited by Zn uptake antagonists. Enterocytes from AE patients were unable to gain significant Zn through exposure to ZnCl2 but did not show differences with respect to ZnAAs. We conclude that ZnAAs may possess an advantage over classical Zn supplements such as Zn salts, as they may be able to increase bioavailability of Zn, and may be more efficient in patients with AE.


Asunto(s)
Acrodermatitis/tratamiento farmacológico , Aminoácidos/farmacocinética , Complejos de Coordinación/farmacocinética , Enterocitos/efectos de los fármacos , Zinc/deficiencia , Zinc/farmacocinética , Acrodermatitis/metabolismo , Acrodermatitis/patología , Aminoácidos/química , Aminoácidos/metabolismo , Animales , Disponibilidad Biológica , Transporte Biológico , Células CACO-2 , Proteínas Portadoras/metabolismo , Diferenciación Celular , Complejos de Coordinación/química , Complejos de Coordinación/metabolismo , Enterocitos/citología , Enterocitos/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Zinc/química , Zinc/metabolismo
3.
Sci Rep ; 7: 45190, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28345660

RESUMEN

Phelan McDermid Syndrome (PMDS) is a genetic disorder characterized by features of Autism spectrum disorders. Similar to reports of Zn deficiency in autistic children, we have previously reported high incidence of Zn deficiency in PMDS. However, the underlying mechanisms are currently not well understood. Here, using inductively coupled plasma mass-spectrometry to measure the concentration of Zinc (Zn) and Copper (Cu) in hair samples from individuals with PMDS with 22q13.3 deletion including SHANK3 (SH3 and multiple ankyrin repeat domains 3), we report a high rate of abnormally low Zn/Cu ratios. To investigate possible underlying mechanisms, we generated enterocytes from PMDS patient-derived induced pluripotent stem cells and used Caco-2 cells with knockdown of SHANK3. We detected decreased expression of Zn uptake transporters ZIP2 and ZIP4 on mRNA and protein level correlating with SHANK3 expression levels, and found reduced levels of ZIP4 protein co-localizing with SHANK3 at the plasma membrane. We demonstrated that especially ZIP4 exists in a complex with SHANK3. Furthermore, we performed immunohistochemistry on gut sections from Shank3αß knockout mice and confirmed a link between enterocytic SHANK3, ZIP2 and ZIP4. We conclude that apart from its well-known role in the CNS, SHANK3 might play a specific role in the GI tract.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Trastornos de los Cromosomas/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Zinc/deficiencia , Adolescente , Células CACO-2 , Proteínas de Transporte de Catión/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Niño , Preescolar , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 22/genética , Cromosomas Humanos Par 22/metabolismo , Cobre , Enterocitos/citología , Enterocitos/metabolismo , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Cabello/química , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Lactante , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Adulto Joven
4.
Neural Plast ; 2016: 3760702, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27247802

RESUMEN

Disturbances in neuronal differentiation and function are an underlying factor of many brain disorders. Zinc homeostasis and signaling are important mediators for a normal brain development and function, given that zinc deficiency was shown to result in cognitive and emotional deficits in animal models that might be associated with neurodevelopmental disorders. One underlying mechanism of the observed detrimental effects of zinc deficiency on the brain might be impaired proliferation and differentiation of stem cells participating in neurogenesis. Thus, to examine the molecular mechanisms regulating zinc metabolism and signaling in differentiating neurons, using a protocol for motor neuron differentiation, we characterized the expression of zinc homeostasis genes during neurogenesis using human induced pluripotent stem cells (hiPSCs) and evaluated the influence of altered zinc levels on the expression of zinc homeostasis genes, cell survival, cell fate, and neuronal function. Our results show that zinc transporters are highly regulated genes during neuronal differentiation and that low zinc levels are associated with decreased cell survival, altered neuronal differentiation, and, in particular, synaptic function. We conclude that zinc deficiency in a critical time window during brain development might influence brain function by modulating neuronal differentiation.


Asunto(s)
Homeostasis/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Zinc/metabolismo , Apoptosis/fisiología , Supervivencia Celular/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Neuronas/citología , Transducción de Señal/fisiología
5.
CNS Neurol Disord Drug Targets ; 14(8): 1041-53, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26295815

RESUMEN

A dyshomeostasis of zinc ions has been reported for many psychiatric and neurodegenerative disorders including schizophrenia, attention deficit hyperactivity disorder, depression, autism, Parkinson's and Alzheimer's disease. Furthermore, alterations in zinc-levels have been associated with seizures and traumatic brain injury. Thus, altering zinclevels within the brain is emerging as a new target for the prevention and treatment of psychiatric and neurological diseases. However, given the restriction of zinc uptake into the brain by the blood-brain barrier, methods for controlled regulation and manipulation of zinc concentrations within the brain are rare. Here, we performed in vivo studies investigating the possibility of brain targeted zinc delivery using zinc-loaded nanoparticles which are able to cross the blood-brain barrier. After injecting these nanoparticles, we analyzed the regional and time-dependent distribution of zinc and nanoparticles within the brain. Moreover, we evaluated whether the presence of zinc-loaded nanoparticles alters the expression of zinc sensitive genes and proteins such as metallothioneins and zinc transporters and quantified possible toxic effects. Our results show that zinc loaded g7 nanoparticles offer a promising approach as a novel non - invasive method to selectively enrich zinc in the brain within a small amount of time.


Asunto(s)
Encéfalo/efectos de los fármacos , Fármacos del Sistema Nervioso Central/administración & dosificación , Portadores de Fármacos , Nanopartículas , Zinc/administración & dosificación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Cationes Bivalentes/administración & dosificación , Cationes Bivalentes/farmacocinética , Cationes Bivalentes/toxicidad , Fármacos del Sistema Nervioso Central/farmacocinética , Fármacos del Sistema Nervioso Central/toxicidad , Portadores de Fármacos/química , Portadores de Fármacos/toxicidad , Evaluación Preclínica de Medicamentos , Glicopéptidos/química , Glicopéptidos/toxicidad , Inmunohistoquímica , Ácido Láctico/química , Ácido Láctico/toxicidad , Ratones Endogámicos BALB C , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanopartículas/química , Nanopartículas/toxicidad , Ácido Poliglicólico/química , Ácido Poliglicólico/toxicidad , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Zinc/farmacocinética , Zinc/toxicidad
6.
Biometals ; 27(4): 715-30, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25007851

RESUMEN

Recent studies suggest that synaptic pathology in autism spectrum disorder (ASD) might be caused by the disruption of a signaling pathway at excitatory glutamatergic synapses, which can be influenced by environmental factors. Some factors, such as prenatal zinc deficiency, dysfunction of metallothioneins as well as deletion of COMMD1, all affect brain metal-ion homeostasis and have been associated with ASD. Given that COMMD1 regulates copper levels and that copper and zinc have antagonistic properties, here, we followed the idea that copper overload might induce a local zinc deficiency affecting key players of a putative ASD pathway such as ProSAP/Shank proteins as reported before. Our results show that increased copper levels indeed interfere with intracellular zinc concentrations and affect synaptic ProSAP/Shank levels, which similarly are altered by manipulation of copper and zinc levels through overexpression and knockdown of COMMD1. In line with this, acute and prenatal copper overload lead to local zinc deficiencies in mice. Pups exposed to prenatal copper overload furthermore show a reduction in ProSAP/Shank protein levels in the brain as well as a decreased NMDAR subunit 1 concentration. Thus, it might be likely that brain metal ion status influences a distinct pathway in excitatory synapses associated with genetic forms of ASD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cobre/fisiología , Sinapsis/metabolismo , Zinc/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Trastorno Autístico/metabolismo , Encéfalo/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Hipocampo/citología , Homeostasis , Humanos , Intercambio Materno-Fetal , Ratones Endogámicos C3H , Proteínas del Tejido Nervioso/metabolismo , Embarazo , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo
7.
Metallomics ; 6(5): 960-77, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24643462

RESUMEN

Many neurodegenerative and neuropsychiatric disorders have been reported to coincide with the dysregulation of metal ions in the body and central nervous system. However, in most cases, it is not the imbalance of a single divalent metal ion but a plethora of metal ions reported to be altered. Given that different divalent metal ions are often able to bind to a protein in a competitive manner, although with different affinities, and that they might use similar transporters for uptake and regulation, it is likely that the imbalance of one metal ion will downstream affect the homeostasis of other metal ions. Thus, based on this assumption, we hypothesize that the dysregulation of a specific metal ion will lead to a characteristic biometal profile. Similar profiles might therefore be detected in various neurological disorders. Moreover, if such shared biometal profiles exist across different neurological disorders, it is possible that shared behavioural impairments in these disorders result from the imbalance in metal ion homeostasis. Thus, here, we evaluate the reported excess or deficiency of metal ions in various neurological disorders and aim to integrate reported alterations in metal ions to generate a characteristic biometal profile for the disorder. Based on this, we try to predict which alterations in biometals will be caused by the overload or deficiency of one particular metal ion. Moreover, investigating the behavioural phenotypes of rodent models suffering from alterations in biometals, we assess whether a shared behavioural phenotype exists for disorders with similar biometal profiles. Our results show that observed behavioural aspects of some neurological disorders are reflected in their specific biometal profile and mirrored by mouse models suffering from similar biometal deregulations.


Asunto(s)
Metales/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Humanos
8.
Brain ; 137(Pt 1): 137-52, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24277719

RESUMEN

Proteins of the ProSAP/Shank family act as major organizing scaffolding elements within the postsynaptic density of excitatory synapses. Deletions, mutations or the downregulation of these molecules has been linked to autism spectrum disorders, the related Phelan McDermid Syndrome or Alzheimer's disease. ProSAP/Shank proteins are targeted to synapses depending on binding to zinc, which is a prerequisite for the assembly of the ProSAP/Shank scaffold. To gain insight into whether the previously reported assembly of ProSAP/Shank through zinc ions provides a crossing point between genetic forms of autism spectrum disorder and zinc deficiency as an environmental risk factor for autism spectrum disorder, we examined the interplay between zinc and ProSAP/Shank in vitro and in vivo using neurobiological approaches. Our data show that low postsynaptic zinc availability affects the activity dependent increase in ProSAP1/Shank2 and ProSAP2/Shank3 levels at the synapse in vitro and that a loss of synaptic ProSAP1/Shank2 and ProSAP2/Shank3 occurs in a mouse model for acute and prenatal zinc deficiency. Zinc-deficient animals displayed abnormalities in behaviour such as over-responsivity and hyperactivity-like behaviour (acute zinc deficiency) and autism spectrum disorder-related behaviour such as impairments in vocalization and social behaviour (prenatal zinc deficiency). Most importantly, a low zinc status seems to be associated with an increased incidence rate of seizures, hypotonia, and attention and hyperactivity issues in patients with Phelan-McDermid syndrome, which is caused by haploinsufficiency of ProSAP2/Shank3. We suggest that the molecular underpinning of prenatal zinc deficiency as a risk factor for autism spectrum disorder may unfold through the deregulation of zinc-binding ProSAP/Shank family members.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/metabolismo , Saposinas/metabolismo , Sinapsis/fisiología , Zinc/deficiencia , Animales , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Conducta Animal/fisiología , Western Blotting , Células Cultivadas , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Deleción Cromosómica , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/fisiopatología , Cromosomas Humanos Par 22/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Ratones , Técnicas de Cultivo de Órganos , Embarazo , ARN Interferente Pequeño/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Fluorescencia , Transfección , Vocalización Animal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA