Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 533(1): 64-69, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-32921413

RESUMEN

Obesity is the main risk factor behind insulin resistance and type 2 diabetes. Still, the mechanism behind adipocyte dysfunction is not yet resolved. Recently, we reported that rapid actin remodeling correlates with adipose cell size changes after short-term overfeeding. Therefore, we hypothesized that the actin-driven myocardin-related transcription factor (MRTF-A) contributes to impaired mature adipocyte function. Primary human adipocytes were subjected to adenoviral overexpression of MRTF-A or MRTF-B, followed by Western blot analysis and tracer glucose uptake assay. Further, we assessed cell size distribution, insulin response, MRTF-A localization, actin organization and degree of polymerization in adipocytes isolated from Ob/Ob mice. Overexpression of MRTF-A, but not MRTF-B, markedly suppressed PPARγ expression. Further, MRTF-A expression resulted in decreased IRS-1 level, shifted phosphorylation of Akt (pS473/pT308), IRS-1 (pS302) and AS160 (pT642), and lowered insulin-stimulated glucose uptake. Hypertrophic adipocytes from Ob/Ob mice displayed an increased proportion of polymerized actin, and increased nuclear translocation of MRTF-A compared with control (Ob/+). Similar with human adipocytes overexpressing MRTF-A, adipocytes isolated from Ob/Ob mice had reduced expression of IRS-1 and PPARγ, as well as impaired insulin response. Together, these data demonstrate that MRTF-A negatively influences insulin sensitivity and the expression of key targets in fully mature human adipocytes. This suggests that MRTF-A is poised to exert a transcriptional response in hypertrophic adipocytes, contributing to adipocyte dysfunction and insulin resistance.


Asunto(s)
Adipocitos/metabolismo , Resistencia a la Insulina , PPAR gamma/metabolismo , Transactivadores/metabolismo , Animales , Células Cultivadas , Regulación hacia Abajo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Ratones Obesos , PPAR gamma/genética , Transactivadores/genética , Regulación hacia Arriba
2.
Neurobiol Dis ; 132: 104560, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31419548

RESUMEN

Body weight has been shown to be a predictor of clinical progression in Huntington's disease (HD). Alongside widespread neuronal pathology, both HD patients and the R6/2 mouse model of HD exhibit weight loss and increased energy expenditure, providing a rationale for targeting whole-body energy metabolism in HD. Leptin-deficient mice display low energy expenditure and increased body weight. We therefore hypothesized that normalizing energy metabolism in R6/2 mice, utilizing leptin- deficiency, would lead to a slower disease progression in the R6/2 mouse. In this study, we show that R6/2 mice on a leptin-deficient genetic background display increased body weight and increased fat mass compared to R6/2 mice, as well as wild type littermates. The increased body weight was accompanied by low energy expenditure, illustrated by a reduction in respiratory exchange rate. Leptin-deficient R6/2 mice had large white adipocytes with white adipocyte gene expression characteristics, in contrast to white adipose tissue in R6/2 mice, where white adipose tissue showed signs of browning. Leptin-deficient R6/2 mice did not exhibit improved neuropathological measures. Our results indicate that lowering energy metabolism in HD, by increasing fat mass and reducing respiratory exchange rate, is not sufficient to affect neuropathology. Further studies targeting energy metabolism in HD are warranted.


Asunto(s)
Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Enfermedad de Huntington/metabolismo , Leptina/deficiencia , Pérdida de Peso/fisiología , Animales , Femenino , Enfermedad de Huntington/genética , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Obesos , Ratones Transgénicos
3.
Mol Cell Neurosci ; 88: 118-129, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29289683

RESUMEN

The three factors, p53, the microRNA-34 family and Sirtuin 1 (SIRT1), interact in a positive feedback loop involved in cell cycle progression, cellular senescence and apoptosis. Each factor in this triad has roles in metabolic regulation, maintenance of mitochondrial function, and regulation of brain-derived neurotrophic factor (BDNF). Thus, this regulatory network holds potential importance for the pathophysiology of Huntington's disease (HD), an inherited neurodegenerative disorder in which both mitochondrial dysfunction and impaired neurotrophic signalling are observed. We investigated expression of the three members of this regulatory triad in the R6/2 HD mouse model. Compared to wild-type littermates, we found decreased levels of miR-34a-5p, increased SIRT1 mRNA and protein levels, and increased levels of p53 protein in brain tissue from R6/2 mice. The upregulation of SIRT1 did not appear to lead to an increased activity of the enzyme, as based on measures of p53 acetylation. In other words, the observed changes did not reflect the known interactions between these factors, indicating a general perturbation of the p53, miR-34a and SIRT1 pathway in HD. This is the first study investigating the entire triad during disease progression in an HD model. Given the importance of these three factors alone and within the triad, our results indicate that outside factors are regulating - or dysregulating - this pathway in HD.


Asunto(s)
Enfermedad de Huntington/genética , MicroARNs/genética , Sirtuina 1/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/fisiología , Línea Celular , Modelos Animales de Enfermedad , Enfermedad de Huntington/metabolismo , Ratones Transgénicos , Transducción de Señal , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
4.
J Neurochem ; 137(5): 670-2, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27059524

RESUMEN

This Editorial highlights a study published in the current issue of Journal of Neurochemistry by Dobson et al. (), investigating whether the immunomodulatory agent, laquinimod exerts an immunomodulatory effect on isolated Huntington's disease monocytes. In Huntington's disease (HD) a central immune activation is mirrored in the periphery by a low-grade immune response and monocytes isolated from HD gene carriers have been shown pathologically hyperreactive in response to stimulation. This hyperreactive immune system has become recognized as an important feature of HD pathogenesis and the employment of a strategy to affect this hyperreactivity could be a potential disease-modifying avenue in HD. Read the highlighted article 'Laquinimod dampens hyperactive cytokine production in Huntington's disease patient myeloid cells' on page 782.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/inmunología , Factores Inmunológicos/uso terapéutico , Inmunomodulación/inmunología , Animales , Citocinas/antagonistas & inhibidores , Citocinas/inmunología , Humanos , Factores Inmunológicos/farmacología , Inmunomodulación/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/inmunología , Quinolonas/farmacología , Quinolonas/uso terapéutico , Resultado del Tratamiento
5.
J Neurochem ; 137(5): 820-37, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27016395

RESUMEN

Huntington's disease (HD) is an inherited and fatal polyglutamine neurodegenerative disorder caused by an expansion of the CAG triplet repeat coding region within the HD gene. Progressive dysfunction and loss of striatal GABAergic medium spiny neurons (MSNs) may account for some of the characteristic symptoms in HD patients. Interestingly, in HD, MSNs expressing neuropeptide Y (NPY) are spared and their numbers is even up-regulated in HD patients. Consistent with this, we report here on increased immuno-linked NPY (IL-NPY) levels in human cerebrospinal fluid (hCSF) from HD patients (Control n = 10; early HD n = 9; mid HD n = 11). As this antibody-based detection of NPY may provide false positive differences as a result of the antibody-based detections of only fragments of NPY, the initial finding was validated by investigating the proteolytic stability of NPY in hCSF using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) and selective inhibitors. A comparison between resulting NPY-fragments and detailed epitope analysis verified significant differences in IL-NPY1-36/3-36 and NPY1-30 levels between HD patients and control subjects with no significant differences between early vs mid HD cases. Ex vivo degradomics analysis demonstrated that NPY is initially degraded to NPY1-30 by cathepsin D in both HD patients and control subjects. Yet, NPY1-30 is then further differentially hydrolyzed by thimet oligopeptidase (TOP) in HD patients and by neprilysin (NEP) in control subjects. Furthermore, altered hCSF TOP-inhibitor Dynorphin A1-13 (Dyn-A1-13 ) and TOP-substrate Dyn-A1-8 levels indicate an impaired Dyn-A-TOP network in HD patients. Thus, we conclude that elevated IL-NPY-levels in conjunction with TOP-/NEP-activity/protein as well as Dyn-A1-13 -peptide levels may serve as a potential biomarker in human CSF of HD. Huntington's disease (HD) patients' cerebrospinal fluid (CSF) exhibits higher neuropeptide Y (NPY) levels. Further degradomics studies show that CSF-NPY is initially degraded to NPY1-30 by Cathepsin D. The NPY1-30 fragment is then differentially degraded in HD vs control involving Neprilysin (NEP), Thimet Oligopeptidase (TOP), and TOP-Dynorphin-A network. Together, these findings may help in search for HD biomarkers.


Asunto(s)
Enfermedad de Huntington/líquido cefalorraquídeo , Enfermedad de Huntington/diagnóstico , Neuropéptido Y/líquido cefalorraquídeo , Fragmentos de Péptidos/líquido cefalorraquídeo , Proteolisis , Adulto , Anciano , Animales , Biomarcadores/líquido cefalorraquídeo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Persona de Mediana Edad , Ratas
6.
Neurobiol Dis ; 73: 388-98, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25447230

RESUMEN

Inflammation is a growing area of research in neurodegeneration. In Huntington's disease (HD), a fatal inherited neurodegenerative disease caused by a CAG-repeat expansion in the gene encoding huntingtin, patients have increased plasma levels of inflammatory cytokines and circulating monocytes that are hyper-responsive to immune stimuli. Several mouse models of HD also show elevated plasma levels of inflammatory cytokines. To further determine the degree to which these models recapitulate observations in HD patients, we evaluated various myeloid cell populations from different HD mouse models to determine whether they are similarly hyper-responsive, as well as measuring other aspects of myeloid cell function. Myeloid cells from each of the three mouse models studied, R6/2, HdhQ150 knock-in and YAC128, showed increased cytokine production when stimulated. However, bone marrow CD11b(+) cells did not show the same hyper-responsive phenotype as spleen and blood cells. Furthermore, macrophages isolated from R6/2 mice show increased levels of phagocytosis, similar to findings in HD patients. Taken together, these results show significant promise for these mouse models to be used to study targeting innate immune pathways identified in human cells, thereby helping to understand the role the peripheral immune system plays in HD progression.


Asunto(s)
Citocinas/inmunología , Modelos Animales de Enfermedad , Enfermedad de Huntington/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Células Mieloides/inmunología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
7.
BMC Neurol ; 15: 8, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25651913

RESUMEN

BACKGROUND: Phosphorylcholine is one of the major epitopes of oxidised low density lipoprotein. Low levels of IgM antibodies against phosphorylcholine (anti-PC) are associated with development of myocardial infarction and stroke. It has been shown that patients with Alzheimer's disease and other dementias have significantly lower serum anti-PC levels compared to controls, suggesting that low levels of atheroprotective anti-PC may play a role in AD and dementia. METHODS: We quantified levels of anti-PC levels using an ELISA in plasma from 176 controls, 125 patients with Alzheimer's disease, 19 patients with vascular dementia and 63 patients with other dementias. RESULTS: We observed similar plasma anti-PC levels in controls, patients with Alzheimer's disease, and other dementias. CONCLUSIONS: Our data suggests that anti-PC is not useful as a biomarker for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/sangre , Anticuerpos/sangre , Fosforilcolina/inmunología , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Estudios de Casos y Controles , Demencia/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad
8.
Brain ; 137(Pt 3): 819-33, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24459107

RESUMEN

Huntington's disease is an inherited neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. The peripheral innate immune system contributes to Huntington's disease pathogenesis and has been targeted successfully to modulate disease progression, but mechanistic understanding relating this to mutant huntingtin expression in immune cells has been lacking. Here we demonstrate that human Huntington's disease myeloid cells produce excessive inflammatory cytokines as a result of the cell-intrinsic effects of mutant huntingtin expression. A direct effect of mutant huntingtin on the NFκB pathway, whereby it interacts with IKKγ, leads to increased degradation of IκB and subsequent nuclear translocation of RelA. Transcriptional alterations in intracellular immune signalling pathways are also observed. Using a novel method of small interfering RNA delivery to lower huntingtin expression, we show reversal of disease-associated alterations in cellular function-the first time this has been demonstrated in primary human cells. Glucan-encapsulated small interfering RNA particles were used to lower huntingtin levels in human Huntington's disease monocytes/macrophages, resulting in a reversal of huntingtin-induced elevated cytokine production and transcriptional changes. These findings improve our understanding of the role of innate immunity in neurodegeneration, introduce glucan-encapsulated small interfering RNA particles as tool for studying cellular pathogenesis ex vivo in human cells and raise the prospect of immune cell-directed HTT-lowering as a therapeutic in Huntington's disease.


Asunto(s)
Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Células Mieloides/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Transducción de Señal/genética , Regulación de la Expresión Génica/inmunología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Inmunidad Innata/genética , Células Mieloides/inmunología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , ARN Interferente Pequeño/uso terapéutico , Transducción de Señal/inmunología , Células U937
9.
J Perinat Med ; 43(2): 159-64, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25014513

RESUMEN

OBJECTIVE: To survey the placental gene expression of inflammatory markers and growth factors in non-smoking obese women with an uncomplicated pregnancy without associated morbidity and delivery at term compared with normal weight women. METHODS: Placental tissue samples from 32 obese women (body mass index, BMI≥35.0 kg/m2) were compared with samples from 94 normal weight women (BMI 18.5-25.0 kg/m2) matched for age (±1 year), gestational age (±3 days), parity and mode of delivery. Semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) was used to analyse toll receptor-2 and -4, interleukin-6 and -8, tumour necrosis factor-α, leptin, adiponectin, insulin-like growth factor-1 and -2, hepatocyte growth factor, hepatocyte growth factor receptor and insulin receptor. RESULTS: There was no significant difference in gene expression in placental tissue samples from obese and normal weight women. CONCLUSION: We found no difference in the occurrence of inflammatory marker and growth factor mRNA levels in placental tissue samples from a large group of obese women without associated morbidity and with healthy infants compared to a closely matched control group of healthy normal weight women. Compared with the previous studies, this anomalous finding may be explained by the absence of associated morbidity in the obese women in our study.


Asunto(s)
Biomarcadores/metabolismo , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Obesidad/metabolismo , Placenta/metabolismo , Complicaciones del Embarazo/metabolismo , Adulto , Estudios de Casos y Controles , Femenino , Expresión Génica , Humanos , Inflamación/etiología , Obesidad/complicaciones , Embarazo , Complicaciones del Embarazo/etiología , Adulto Joven
10.
Mov Disord ; 29(10): 1319-22, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25044107

RESUMEN

BACKGROUND: Differential diagnosis of parkinsonian disorders is challenging because of overlapping symptoms, especially during early stages of disease. No validated biomarkers are available for early and accurate diagnosis of multiple system atrophy and other parkinsonian disorders. It has been reported that flt3 ligand levels in cerebrospinal fluid could clearly differentiate patients with Parkinson's disease from patients with multiple system atrophy, with 99% sensitivity and 95% specificity. METHODS: We measured flt3 ligand levels in cerebrospinal fluid of subjects with Parkinson's disease (n = 37), multiple system atrophy (n = 30), and progressive supranuclear palsy (n = 19). RESULTS: In our cohort, no significant difference was found in flt3 ligand levels between Parkinson's disease, multiple system atrophy, and progressive supranuclear palsy. CONCLUSIONS: Our results suggest that cerebrospinal fluid flt3 ligand levels do not differentiate between parkinsonian disorders.


Asunto(s)
Proteínas de la Membrana/líquido cefalorraquídeo , Trastornos Parkinsonianos/líquido cefalorraquídeo , Anciano , Análisis de Varianza , Humanos , Persona de Mediana Edad , Atrofia de Múltiples Sistemas/líquido cefalorraquídeo , Índice de Severidad de la Enfermedad , Parálisis Supranuclear Progresiva/líquido cefalorraquídeo
11.
Mov Disord ; 29(12): 1511-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25164424

RESUMEN

This study was undertaken to determine whether the production of melatonin, a hormone regulating sleep in relation to the light/dark cycle, is altered in Huntington's disease. We analyzed the circadian rhythm of melatonin in a 24-hour study of cohorts of control, premanifest, and stage II/III Huntington's disease subjects. The mean and acrophase melatonin concentrations were significantly reduced in stage II/III Huntington's disease subjects compared with controls. We also observed a nonsignificant trend toward reduced mean and acrophase melatonin in premanifest Huntington's disease subjects. Onset of melatonin rise was significantly more temporally spread in both premanifest and stage II/III Huntington's disease subjects compared with controls. A nonsignificant trend also was seen for reduced pulsatile secretion of melatonin. Melatonin concentrations are reduced in Huntington's disease. Altered melatonin patterns may provide an explanation for disrupted sleep and circadian behavior in Huntington's disease, and represent a biomarker for disease state. Melatonin therapy may help the sleep disorders seen in Huntington's disease.


Asunto(s)
Enfermedad de Huntington/sangre , Melatonina/sangre , Adulto , Anciano , Femenino , Análisis de Fourier , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
12.
J Neurosci ; 32(1): 133-42, 2012 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-22219276

RESUMEN

Huntington's disease (HD) is caused by an expanded polyglutamine tract in the protein huntingtin (htt). Although HD has historically been viewed as a brain-specific disease, htt is expressed ubiquitously, and recent studies indicate that mutant htt might cause changes to the immune system that could contribute to pathogenesis. Monocytes from HD patients and mouse models are hyperactive in response to stimulation, and increased levels of inflammatory cytokines and chemokines are found in pre-manifest patients that correlate with pathogenesis. In this study, wild-type (WT) bone marrow cells were transplanted into two lethally irradiated transgenic mouse models of HD that ubiquitously express full-length htt (YAC128 and BACHD mice). Bone marrow transplantation partially attenuated hypokinetic and motor deficits in HD mice. Increased levels of synapses in the cortex were found in HD mice that received bone marrow transplants. Importantly, serum levels of interleukin-6, interleukin-10, CXC chemokine ligand 1, and interferon-γ were significantly higher in HD than WT mice but were normalized in mice that received a bone marrow transplant. These results suggest that immune cell dysfunction might be an important modifier of pathogenesis in HD.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/terapia , Células de la Médula Ósea/inmunología , Trasplante de Médula Ósea/métodos , Enfermedad de Huntington/inmunología , Enfermedad de Huntington/terapia , Terapia de Inmunosupresión/métodos , Animales , Enfermedades Autoinmunes del Sistema Nervioso/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedad de Huntington/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Hum Mol Genet ; 20(11): 2225-37, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21421997

RESUMEN

Huntington's disease (HD) is a devastating, neurodegenerative condition, which lacks effective treatment. Normal Huntingtin (HTT) and mutant Huntingtin (mHTT) are expressed in multiple tissues and can alter transcription of microRNAs (miRs). Importantly, miRs are present in a bio-stable form in human peripheral blood plasma and have recently been shown to be useful biomarkers in other diseases. We therefore sought to identify potential miR biomarkers of HD that are present in, and have functional consequences for, neuronal and non-neuronal tissues. In a cell line over-expressing mHTT-Exon-1, miR microarray analysis was used to identify candidate miRs. We then examined their presence and bio-stability in control and HD plasma. We found that miR-34b is significantly elevated in response to mHTT-Exon-1, and its blockade alters the toxicity of mHTT-Exon-1 in vitro. We also show that miR-34b is detectable in plasma from small input volumes and is insensitive to freeze-thaw-induced RNA degradation. Interestingly, miR-34b is significantly elevated in plasma from HD gene carriers prior to symptom onset. This is the first study suggesting that plasma miRs might be used as biomarkers for HD.


Asunto(s)
Enfermedad de Huntington/genética , MicroARNs/sangre , Adulto , Anciano , Biomarcadores/sangre , Muerte Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Exones , Femenino , Humanos , Enfermedad de Huntington/sangre , Inmunohistoquímica , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Mutación , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Transfección , Regulación hacia Arriba , Adulto Joven
14.
Acta Paediatr ; 102(10): 1010-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23845107

RESUMEN

AIM: Staphylococci are a major contribution for neonatal sepsis, which is the main risk factor for bronchopulmonary dysplasia. This study investigated the expression of pro-inflammatory mediators in endothelial and respiratory cells from newborns exposed to staphylococci. METHODS: Human vascular endothelial cells and small airway epithelial cells were incubated with neonatal blood isolates of Staphylococcus epidermidis (n = 14) and Staphylococcus aureus (n = 14). The extracellular release of IL-8, IL-10, sICAM-1, ICAM-1 mRNA and the expression of membrane bound ICAM-1 were assessed by ELISA, RT-PCR and immunofluorescence microscopy. RESULTS: Staphylococcus epidermidis induced higher levels of IL-8 (mean 38.5 ng/mL) and ICAM-1 mRNA (mean ratio 1.037) in the small airway epithelial cells than S. aureus (IL-8 mean 22.2 ng/mL, p < 0.01 and ICAM-1 mRNA mean ratio 0.715, p < 0.01). In the endothelial cells, ICAM-1 remained more integrated in the cell membranes after exposure to S. epidermidis compared with S. aureus, which induced disintegration and release of soluble ICAM-1 into the supernatants. CONCLUSION: Staphylococcus epidermidis induced a higher chemoattractive response than S. aureus. A persistent transmigration of granulocytes into the lung tissue in neonatal S. epidermidis sepsis might contribute to the development of bronchopulmonary dysplasia.


Asunto(s)
Células Endoteliales/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-8/metabolismo , Mucosa Respiratoria/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/patogenicidad , Staphylococcus epidermidis/patogenicidad , Biomarcadores/metabolismo , Células Cultivadas , Células Endoteliales/microbiología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Recién Nacido , Interleucina-10/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/microbiología , Sepsis/metabolismo , Staphylococcus aureus/aislamiento & purificación , Staphylococcus epidermidis/aislamiento & purificación
15.
J Huntingtons Dis ; 12(3): 253-266, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37718850

RESUMEN

BACKGROUND: Metabolic alterations contribute to disease onset and prognosis of Huntington's disease (HD). Weight loss in the R6/2 mouse model of HD is a consistent feature, with onset in mid-to-late stage of disease. OBJECTIVE: In the present study, we aimed to investigate molecular and functional changes in white adipose tissue (WAT) that occur at weight loss in R6/2 mice. We further elaborated on the effect of leptin-deficiency and early obesity in R6/2 mice. METHODS: We performed analyses at 12 weeks of age; a time point that coincides with the start of weight loss in our R6/2 mouse colony. Gonadal (visceral) and inguinal (subcutaneous) WAT depot weights were monitored, as well as adipocyte size distribution. Response to isoprenaline-stimulated glycerol release and insulin-stimulated glucose uptake in adipocytes from gonadal WAT was assessed. RESULTS: In R6/2 mice, WAT depot weights were comparable to wildtype (WT) mice, and the response to insulin and isoprenaline in gonadal adipocytes was unaltered. Leptin-deficient R6/2 mice exhibited distinct changes compared to leptin-deficient WT mice. At 12 weeks, female leptin-deficient R6/2 mice had reduced body weight accompanied by an increased proportion of smaller adipocytes, while in contrast; male mice displayed a shift towards larger adipocyte sizes without a significant body weight reduction at this timepoint. CONCLUSIONS: We here show that there are early sex-specific changes in adipocyte cell size distribution in WAT of R6/2 mice and leptin-deficient R6/2 mice.

16.
Front Aging Neurosci ; 15: 1089005, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37261266

RESUMEN

Introduction: Dysfunction of the cerebral vasculature is considered one of the key components of Alzheimer's disease (AD), but the mechanisms affecting individual brain vessels are poorly understood. Methods: Here, using in vivo two-photon microscopy in superficial cortical layers and ex vivo imaging across brain regions, we characterized blood-brain barrier (BBB) function and neurovascular coupling (NVC) at the level of individual brain vessels in adult female 5xFAD mice, an aggressive amyloid-ß (Aß) model of AD. Results: We report a lack of abnormal increase in adsorptive-mediated transcytosis of albumin and preserved paracellular barrier for fibrinogen and small molecules despite an extensive load of Aß. Likewise, the NVC responses to somatosensory stimulation were preserved at all regulatory segments of the microvasculature: penetrating arterioles, precapillary sphincters, and capillaries. Lastly, the Aß plaques did not affect the density of capillary pericytes. Conclusion: Our findings provide direct evidence of preserved microvascular function in the 5xFAD mice and highlight the critical dependence of the experimental outcomes on the choice of preclinical models of AD. We propose that the presence of parenchymal Aß does not warrant BBB and NVC dysfunction and that the generalized view that microvascular impairment is inherent to Aß aggregation may need to be revised.

17.
Mol Metab ; 57: 101439, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35007790

RESUMEN

OBJECTIVE: In Huntington's disease (HD), the disease-causing huntingtin (HTT) protein is ubiquitously expressed and causes both central and peripheral pathology. In clinical HD, a higher body mass index has been associated with slower disease progression, indicating the role of metabolic changes in disease pathogenesis. Underlying mechanisms of metabolic changes in HD remain poorly understood, but recent studies suggest the involvement of hypothalamic dysfunction. The present study aimed to investigate whether modulation of hypothalamic HTT levels would affect metabolic phenotype and disease features in HD using mouse models. METHODS: We used the R6/2 and BACHD mouse models that express different lengths of mutant HTT to develop lean- and obese phenotypes, respectively. We utilized adeno-associated viral vectors to overexpress either mutant or wild-type HTT in the hypothalamus of R6/2, BACHD, and their wild-type littermates. The metabolic phenotype was assessed by body weight measurements over time and body composition analysis using dual-energy x-ray absorptiometry at the endpoint. R6/2 mice were further characterized using behavioral analyses, including rotarod, nesting-, and hindlimb clasping tests during early- and late-time points of disease progression. Finally, gene expression analysis was performed in R6/2 mice and wild-type littermates in order to assess transcriptional changes in the hypothalamus and adipose tissue. RESULTS: Hypothalamic overexpression of mutant HTT induced significant gender-affected body weight gain in all models, including wild-type mice. In R6/2 females, early weight gain shifted to weight loss during the corresponding late stage of disease despite increased fat accumulation. Body weight changes were accompanied by behavioral alterations. During the period of early weight gain, R6/2 mice displayed a comparable locomotor capacity to wild-type mice. When assessing behavior just prior to weight loss onset in R6/2 mice, decreased locomotor performance was observed in R6/2 females with hypothalamic overexpression of mutant HTT. Transcriptional downregulation of beta-3 adrenergic receptor (B3AR), adipose triglyceride lipase (ATGL), and peroxisome proliferator-activated receptor-gamma (PPARγ) in gonadal white adipose tissue was accompanied by distinct alterations in hypothalamic gene expression profiles in R6/2 females after mutant HTT overexpression. No significant effect on metabolic phenotype in R6/2 was seen in response to wild-type HTT overexpression. CONCLUSIONS: Taken together, our findings provide further support for the role of HTT in metabolic control via hypothalamic neurocircuits. Understanding the specific central neurocircuits and their peripheral link underlying metabolic imbalance in HD may open up avenues for novel therapeutic interventions.


Asunto(s)
Enfermedad de Huntington , Animales , Modelos Animales de Enfermedad , Femenino , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Hipotálamo/metabolismo , Ratones , Ratones Transgénicos , Fenotipo
18.
Front Neurosci ; 16: 1027269, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36408416

RESUMEN

Structural changes and neuropathology in the hypothalamus have been suggested to contribute to the non-motor manifestations of Huntington's disease (HD), a neurodegenerative disorder caused by an expanded cytosine-adenine-guanine (CAG) repeat in the huntingtin (HTT) gene. In this study, we investigated whether hypothalamic HTT expression causes transcriptional changes. Hypothalamic RNA was isolated from two different HD mouse models and their littermate controls; BACHD mice with ubiquitous expression of full-length mutant HTT (mHTT) and wild-type mice with targeted hypothalamic overexpression of either wild-type HTT (wtHTT) or mHTT fragments. The mHTT and wtHTT groups showed the highest number of differentially expressed genes compared to the BACHD mouse model. Gene Set Enrichment Analysis (GSEA) with leading-edge analysis showed that suppressed sterol- and cholesterol metabolism were shared between hypothalamic wtHTT and mHTT overexpression. Most distinctive for mHTT overexpression was the suppression of neuroendocrine networks, in which qRT-PCR validation confirmed significant downregulation of neuropeptides with roles in feeding behavior; hypocretin neuropeptide precursor (Hcrt), tachykinin receptor 3 (Tacr3), cocaine and amphetamine-regulated transcript (Cart) and catecholamine-related biological processes; dopa decarboxylase (Ddc), histidine decarboxylase (Hdc), tyrosine hydroxylase (Th), and vasoactive intestinal peptide (Vip). In BACHD mice, few hypothalamic genes were differentially expressed compared to age-matched WT controls. However, GSEA indicated an enrichment of inflammatory- and gonadotropin-related processes at 10 months. In conclusion, we show that both wtHTT and mHTT overexpression change hypothalamic transcriptome profile, specifically mHTT, altering neuroendocrine circuits. In contrast, the ubiquitous expression of full-length mHTT in the BACHD hypothalamus moderately affects the transcriptomic profile.

19.
Neurobiol Dis ; 44(1): 1-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21624468

RESUMEN

Weight loss is the most important non-neurological complication of Huntington's disease (HD). It correlates with disease progression and affects the quality of life of HD patients, suggesting that it could be a valuable target for therapeutic intervention. The mechanism underlying weight loss in HD is unknown. Mutant huntingtin, the protein that causes the disease, is not only expressed in the brain, but also along the gastrointestinal (GI) tract. Here we demonstrate that the GI tract of HD mice is affected. At the anatomical level we observed loss of enteric neuropeptides, as well as decreased mucosal thickness and villus length. Exploring the functions of the GI system we found impaired gut motility, diarrhea, and malabsorption of food. The degree of malabsorption was inversely associated with body weight, suggesting that GI dysfunction plays an important role in weight loss in HD mice. In summary, these observations suggest that the GI tract is affected in HD mice and that GI dysfunction contributes to nutritional deficiencies and weight loss.


Asunto(s)
Enfermedades Gastrointestinales/fisiopatología , Enfermedad de Huntington/fisiopatología , Pérdida de Peso/fisiología , Envejecimiento/fisiología , Animales , Peso Corporal/fisiología , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/fisiología , Heces/química , Femenino , Mucosa Gástrica/fisiología , Mucosa Gástrica/ultraestructura , Enfermedades Gastrointestinales/etiología , Motilidad Gastrointestinal/fisiología , Enfermedad de Huntington/complicaciones , Inmunohistoquímica , Absorción Intestinal/fisiología , Mucosa Intestinal/fisiología , Mucosa Intestinal/ultraestructura , Síndromes de Malabsorción/genética , Síndromes de Malabsorción/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Agua/metabolismo
20.
Hum Mol Genet ; 18(20): 3942-54, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19628478

RESUMEN

Huntington's disease is a severe progressive neurodegenerative disorder caused by a CAG expansion in the IT15 gene, which encodes huntingtin. The disease primarily affects the neostriatum and cerebral cortex and also associates with increased incidence of diabetes. Here, we show that mutant huntingtin disrupts intracellular transport and insulin secretion by direct interference with microtubular beta-tubulin. We demonstrate that mutant huntingtin impairs glucose-stimulated insulin secretion in insulin-producing beta-cells, without altering stored levels of insulin. Using VSVG-YFP, we show that mutant huntingtin retards post-Golgi transport. Moreover, we demonstrate that the speed of insulin vesicle trafficking is reduced. Using immunoprecipitation of mutant and wild-type huntingtin in combination with mass spectrometry, we reveal an enhanced and aberrant interaction between mutant huntingtin and beta-tubulin, implying the underlying mechanism of impaired intracellular transport. Thus, our findings have revealed a novel pathogenetic process by which mutant huntingtin may disrupt hormone exocytosis from beta-cells and possibly impair vesicular transport in any cell that expresses the pathogenic protein.


Asunto(s)
Enfermedad de Huntington/metabolismo , Insulina/metabolismo , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Vesículas Transportadoras/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Unión Proteica , Transporte de Proteínas , Ratas , Vesículas Transportadoras/genética , Tubulina (Proteína)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA