Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(14): E1705-14, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25775548

RESUMO

Dominant mutations in p97/VCP (valosin-containing protein) cause a rare multisystem degenerative disease with varied phenotypes that include inclusion body myopathy, Paget's disease of bone, frontotemporal dementia, and amyotrophic lateral sclerosis. p97 disease mutants have altered N-domain conformations, elevated ATPase activity, and altered cofactor association. We have now discovered a previously unidentified disease-relevant functional property of p97 by identifying how the cofactors p37 and p47 regulate p97 ATPase activity. We define p37 as, to our knowledge, the first known p97-activating cofactor, which enhances the catalytic efficiency (kcat/Km) of p97 by 11-fold. Whereas both p37 and p47 decrease the Km of ATP in p97, p37 increases the kcat of p97. In contrast, regulation by p47 is biphasic, with decreased kcat at low levels but increased kcat at higher levels. By deleting a region of p47 that lacks homology to p37 (amino acids 69-92), we changed p47 from an inhibitory cofactor to an activating cofactor, similar to p37. Our data suggest that cofactors regulate p97 ATPase activity by binding to the N domain. Induced conformation changes affect ADP/ATP binding at the D1 domain, which in turn controls ATPase cycling. Most importantly, we found that the D2 domain of disease mutants failed to be activated by p37 or p47. Our results show that cofactors play a critical role in controlling p97 ATPase activity, and suggest that lack of cofactor-regulated communication may contribute to p97-associated disease pathogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Mutação , Trifosfato de Adenosina/metabolismo , Autofagia , Doenças Ósseas/metabolismo , Linhagem Celular Tumoral , Cromatografia em Gel , Complexo de Golgi , Homeostase , Humanos , Doenças Musculares/metabolismo , Doenças Neurodegenerativas/metabolismo , Fenótipo , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície , Proteína com Valosina
2.
Stress ; 19(2): 214-24, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26946982

RESUMO

Chronic stress has been associated with obesity, glucose intolerance, and insulin resistance. We developed a model of chronic psychosocial stress (CPS) in which subordinate mice are vulnerable to obesity and the metabolic-like syndrome while dominant mice exhibit a healthy metabolic phenotype. Here we tested the hypothesis that the metabolic difference between subordinate and dominant mice is associated with changes in functional pathways relevant for insulin sensitivity, glucose and lipid homeostasis. Male mice were exposed to CPS for four weeks and fed either a standard diet or a high-fat diet (HFD). We first measured, by real-time PCR candidate genes, in the liver, skeletal muscle, and the perigonadal white adipose tissue (pWAT). Subsequently, we used a probabilistic analysis approach to analyze different ways in which signals can be transmitted across the pathways in each tissue. Results showed that subordinate mice displayed a drastic downregulation of the insulin pathway in liver and muscle, indicative of insulin resistance, already on standard diet. Conversely, pWAT showed molecular changes suggestive of facilitated fat deposition in an otherwise insulin-sensitive tissue. The molecular changes in subordinate mice fed a standard diet were greater compared to HFD-fed controls. Finally, dominant mice maintained a substantially normal metabolic and molecular phenotype even when fed a HFD. Overall, our data demonstrate that subordination stress is a potent stimulus for the downregulation of the insulin signaling pathway in liver and muscle and a major risk factor for the development of obesity, insulin resistance, and type 2 diabetes mellitus.


Assuntos
Tecido Adiposo Branco/metabolismo , Dominação-Subordinação , Regulação para Baixo , Insulina/metabolismo , Fígado/metabolismo , Músculo Esquelético/metabolismo , Transdução de Sinais/fisiologia , Estresse Psicológico/metabolismo , Animais , Dieta Hiperlipídica , Glucose/metabolismo , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
3.
J Physiol ; 591(16): 3949-62, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23836683

RESUMO

Heterotrimeric G-proteins are critical players in the transduction mechanisms underlying odorant and pheromonal signalling. In the vomeronasal organ (VNO) of the adult mouse, two different G-protein complexes have been identified. Gαoß2γ8 is preferentially expressed in the basal neurons and coexpresses with type-2 vomeronasal pheromone receptors (V2Rs) whereas Gαi2ß2γ2 is found in the apical neurons and coexpresses with type-1 vomeronasal pheromone receptors (V1Rs). V2R-expressing neurons project to the posterior accessory olfactory bulb (AOB) whereas neurons expressing V1Rs send their axon to the anterior AOB. Gγ8 is also expressed in developing olfactory neurons where this protein is probably associated with Go. Here, we generated mice with a targeted deletion of the Gγ8 gene and investigated the behavioural effects and the physiological consequences of this mutation. Gγ8(-/-) mice show a normal development of the main olfactory epithelium; moreover, they do not display major deficits in odour perception. In contrast, the VNO undergoes a slow but remarkable loss of basal neurons starting from the fourth postnatal week, with a 40% reduction of cells at 2 months and 70% at 1 year. This loss is associated with a reduced early-gene expression in the posterior AOB of mice stimulated with pheromones. More interestingly, the Gγ8 deletion specifically leads to a reduced pheromone-mediated aggressiveness in both males and females, all other socio-sexual behaviours remaining unaltered. This study defines a specific role for Gγ8 in maintenance of the neuronal population of the VNO and in the mechanisms of pheromonal signalling that involve the aggressive behaviour towards conspecifics.


Assuntos
Agressão/fisiologia , Comportamento Animal/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Órgão Vomeronasal/fisiologia , Animais , Animais Recém-Nascidos , Feminino , Masculino , Camundongos , Camundongos Knockout , Neurônios/fisiologia , Bulbo Olfatório/fisiologia , Feromônios , Receptores de Feromônios/fisiologia , Reconhecimento Psicológico
4.
Biochem J ; 441(1): 511-22, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21880012

RESUMO

The peptides encoded by the VGF gene are gaining biomedical interest and are increasingly being scrutinized as biomarkers for human disease. An endocrine/neuromodulatory role for VGF peptides has been suggested but never demonstrated. Furthermore, no study has demonstrated so far the existence of a receptor-mediated mechanism for any VGF peptide. In the present study, we provide a comprehensive in vitro, ex vivo and in vivo identification of a novel pro-lipolytic pathway mediated by the TLQP-21 peptide. We show for the first time that VGF-immunoreactivity is present within sympathetic fibres in the WAT (white adipose tissue) but not in the adipocytes. Furthermore, we identified a saturable receptor-binding activity for the TLQP-21 peptide. The maximum binding capacity for TLQP-21 was higher in the WAT as compared with other tissues, and selectively up-regulated in the adipose tissue of obese mice. TLQP-21 increases lipolysis in murine adipocytes via a mechanism encompassing the activation of noradrenaline/ß-adrenergic receptors pathways and dose-dependently decreases adipocytes diameters in two models of obesity. In conclusion, we demonstrated a novel and previously uncharacterized peripheral lipolytic pathway encompassing the VGF peptide TLQP-21. Targeting the sympathetic nerve-adipocytes interaction might prove to be a novel approach for the treatment of obesity-associated metabolic complications.


Assuntos
Neuropeptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Animais , Composição Corporal , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/metabolismo , Masculino , Camundongos , Células NIH 3T3 , Fatores de Crescimento Neural , Obesidade/induzido quimicamente , Obesidade/metabolismo , Ligação Proteica , Transporte Proteico , Receptores de Superfície Celular
5.
Mol Ther Methods Clin Dev ; 27: 452-463, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36419468

RESUMO

Sanfilippo syndrome type B (mucopolysaccharidosis type IIIB) is a recessive genetic disorder that severely affects the brain due to a deficiency in the enzyme α-N-acetylglucosaminidase (NAGLU), leading to intra-lysosomal accumulation of partially degraded heparan sulfate. There are no effective treatments for this disorder. In this project, we carried out an ex vivo correction of neural stem cells derived from Naglu -/- mice (iNSCs) induced pluripotent stem cells (iPSC) using a modified enzyme in which human NAGLU is fused to an insulin-like growth factor II receptor binding peptide in order to improve enzyme uptake. After brain transplantation of corrected iNSCs into Naglu -/- mice and long-term evaluation of their impact, we successfully detected NAGLU-IGFII activity in all transplanted animals. We found decreased lysosomal accumulation and reduced astrocytosis and microglial activation throughout transplanted brains. We also identified a novel neuropathological phenotype in untreated Naglu -/- brains with decreased levels of the neuronal marker Map2 and accumulation of synaptophysin-positive aggregates. Upon transplantation, we restored levels of Map2 expression and significantly reduced formation of synaptophysin-positive aggregates. Our findings suggest that genetically engineered iNSCs can be used to effectively deliver the missing enzyme to the brain and treat Sanfilippo type B-associated neuropathology.

6.
Elife ; 102021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34723800

RESUMO

Ataxia Telangiectasia (A-T) and Ataxia with Ocular Apraxia Type 1 (AOA1) are devastating neurological disorders caused by null mutations in the genome stability genes, A-T mutated (ATM) and Aprataxin (APTX), respectively. Our mechanistic understanding and therapeutic repertoire for treating these disorders are severely lacking, in large part due to the failure of prior animal models with similar null mutations to recapitulate the characteristic loss of motor coordination (i.e., ataxia) and associated cerebellar defects. By increasing genotoxic stress through the insertion of null mutations in both the Atm (nonsense) and Aptx (knockout) genes in the same animal, we have generated a novel mouse model that for the first time develops a progressively severe ataxic phenotype associated with atrophy of the cerebellar molecular layer. We find biophysical properties of cerebellar Purkinje neurons (PNs) are significantly perturbed (e.g., reduced membrane capacitance, lower action potential [AP] thresholds, etc.), while properties of synaptic inputs remain largely unchanged. These perturbations significantly alter PN neural activity, including a progressive reduction in spontaneous AP firing frequency that correlates with both cerebellar atrophy and ataxia over the animal's first year of life. Double mutant mice also exhibit a high predisposition to developing cancer (thymomas) and immune abnormalities (impaired early thymocyte development and T-cell maturation), symptoms characteristic of A-T. Finally, by inserting a clinically relevant nonsense-type null mutation in Atm, we demonstrate that Small Molecule Read-Through (SMRT) compounds can restore ATM production, indicating their potential as a future A-T therapeutic.


Assuntos
Ataxia Telangiectasia/genética , Atrofia/fisiopatologia , Cerebelo/patologia , Códon sem Sentido/genética , Células de Purkinje/metabolismo , Animais , Ataxia Telangiectasia/fisiopatologia , Atrofia/genética , Modelos Animais de Doenças , Feminino , Masculino , Camundongos
7.
Stem Cell Res ; 37: 101434, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30999275

RESUMO

Patient-derived induced pluripotent stem cells (iPSCs) have become a promising resource for exploring genetics of complex diseases, discovering new drugs, and advancing regenerative medicine. Increasingly, laboratories are creating their own banks of iPSCs derived from diverse donors. However, there are not yet standardized guidelines for qualifying these cell lines, i.e., distinguishing between bona fide human iPSCs, somatic cells, and imperfectly reprogrammed cells. Here, we report the establishment of a panel of 30 iPSCs from CD34+ peripheral blood mononuclear cells, of which 10 were further differentiated in vitro into all three germ layers. We characterized these different cell types with commonly used pluripotent and lineage specific markers, and showed that NES, TUBB3, and OTX2 cannot be reliably used as ectoderm differentiation markers. Our work highlights the importance of marker selection in iPSC authentication, and the need for the field to establish definitive standard assays.


Assuntos
Antígenos de Diferenciação/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Ectoderma/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Leucócitos Mononucleares/metabolismo , Células Cultivadas , Ectoderma/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/citologia
8.
Mol Ther Methods Clin Dev ; 10: 113-127, 2018 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-30101150

RESUMO

Sanfilippo syndrome type B (mucopolysaccharidosis type IIIB [MPS IIIB]) is a lysosomal storage disorder primarily affecting the brain that is caused by a deficiency in the enzyme α-N-acetylglucosaminidase (NAGLU), leading to intralysosomal accumulation of heparan sulfate. There are currently no treatments for this disorder. Here we report that, ex vivo, lentiviral correction of Naglu-/- neural stem cells derived from Naglu-/- mice (iNSCs) corrected their lysosomal pathology and allowed them to secrete a functional NAGLU enzyme that could be taken up by deficient cells. Following long-term transplantation of these corrected iNSCs into Naglu-/- mice, we detected NAGLU activity in the majority of engrafted animals. Successfully transplanted Naglu-/- mice showed a significant decrease in storage material, a reduction in astrocyte activation, and complete prevention of microglial activation within the area of engrafted cells and neighboring regions, with beneficial effects extending partway along the rostrocaudal axis of the brain. Our results demonstrate long-term engraftment of iNSCs in the brain that are capable of cross-correcting pathology in Naglu-/- mice. Our findings suggest that genetically engineered iNSCs could potentially be used to deliver enzymes and treat MPS IIIB.

9.
Anticancer Res ; 38(5): 2627-2634, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29715082

RESUMO

BACKGROUND: Anti-metabolites are less-myelosuppressive than DNA-damaging anticancer drugs and may be useful against brain tumors. MATERIALS AND METHODS: We evaluated the asparagine/glutamine-deaminating agent Erwinaze with/without temozolomide against brain tumor cells and mouse medulloblastomas. RESULTS: Erwinaze treatment of cell lines and neurospheres led to dose-dependent reductions of cells (reversible by L-glutamine), with half maximal inhibitory concentrations (IC50s) of 0.12->10 IU/ml. Erwinaze at <1 IU/ml reduced temozolomide IC50s by 3.6- to 13-fold (300-1,200 µM to 40-330 µM). Seven-week-old SMO/SMO mice treated with Erwinaze (regardless of temozolomide treatment) had better survival 11 weeks post-therapy, compared to those not treated with Erwinaze (81.25% vs. 46.15, p=0.08). Temozolomide-treated mice developed 10% weight loss, impairing survival. All 16 mice treated with temozolomide (regardless of Erwinaze treatment) succumbed by 40-weeks of age, whereas 5/8 animals treated with Erwinaze alone and 2/6 controls survived (p=0.035). CONCLUSION: Erwinaze enhances cytotoxicity of temozolomide in vitro, and improves survival in SMO/SMO mice, likely by reducing cerebrospinal fluid glutamine. Temozolomide-associated toxicity prevented demonstration of any potential combinatorial advantage with Erwinaze in vivo.


Assuntos
Antineoplásicos/uso terapêutico , Asparaginase/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Cerebelares/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Meduloblastoma/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Asparaginase/administração & dosagem , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Meios de Cultura Livres de Soro , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Dickeya chrysanthemi/enzimologia , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Glioblastoma/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Glutamina/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Tolerância a Radiação , Esferoides Celulares/efeitos dos fármacos , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Ther Methods Clin Dev ; 8: 42-51, 2018 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-29159202

RESUMO

Antibodies against recombinant proteins can significantly reduce their effectiveness in unanticipated ways. We evaluated the humoral response of mice with the lysosomal storage disease mucopolysaccharidosis type I treated with weekly intravenous recombinant human alpha-l-iduronidase (rhIDU). Unlike patients, the majority of whom develop antibodies to recombinant human alpha-l-iduronidase, only approximately half of the treated mice developed antibodies against recombinant human alpha-l-iduronidase and levels were low. Serum from antibody-positive mice inhibited uptake of recombinant human alpha-l-iduronidase into human fibroblasts by partial inhibition compared to control serum. Tissue and cellular distributions of rhIDU were altered in antibody-positive mice compared to either antibody-negative or naive mice, with significantly less recombinant human alpha-l-iduronidase activity in the heart and kidney in antibody-positive mice. In the liver, recombinant human alpha-l-iduronidase was preferentially found in sinusoidal cells rather than in hepatocytes in antibody-positive mice. Antibodies against recombinant human alpha-l-iduronidase enhanced uptake of recombinant human alpha-l-iduronidase into macrophages obtained from MPS I mice. Collectively, these results imply that a humoral immune response against a therapeutic protein can shift its distribution preferentially into macrophage-lineage cells, causing decreased availability of the protein to the cells that are its therapeutic targets.

11.
PLoS One ; 12(10): e0186818, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29073173

RESUMO

Hemogenic endothelium (HE) undergoes endothelial-to-hematopoietic transition (EHT) to generate blood, a process that requires progressive down-regulation of endothelial genes and induction of hematopoietic ones. Previously, we have shown that the transcription factor HoxA3 prevents blood formation by inhibiting Runx1 expression, maintaining endothelial gene expression and thus blocking EHT. In the present study, we show that HoxA3 also prevents blood formation by inhibiting Notch pathway. HoxA3 induced upregulation of Jag1 ligand in endothelial cells, which led to cis-inhibition of the Notch pathway, rendering the HE nonresponsive to Notch signals. While Notch activation alone was insufficient to promote blood formation in the presence of HoxA3, activation of Notch or downregulation of Jag1 resulted in a loss of the endothelial phenotype which is a prerequisite for EHT. Taken together, these results demonstrate that Notch pathway activation is necessary to downregulate endothelial markers during EHT.


Assuntos
Células Endoteliais/metabolismo , Hematopoese/fisiologia , Proteínas de Homeodomínio/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Regulação para Baixo/fisiologia , Células Endoteliais/citologia , Proteínas de Homeodomínio/genética , Proteína Jagged-1/biossíntese , Proteína Jagged-1/genética , Camundongos , Receptores Notch/genética
12.
Front Nutr ; 1(30)2015.
Artigo em Inglês | MEDLINE | ID: mdl-25621284

RESUMO

BACKGROUND: Eating disorders are associated with physical morbidity and appear to have causal factors like stressful life events and negative affect. Binge eating disorder (BED) is characterized by eating in a discrete period of time a larger than normal amount of food, a sense of lack of control over eating, and marked distress. There are still unmet needs for the identification of mechanisms regulating excessive eating, which is in part due to the lack of appropriate animal models. We developed a naturalistic murine model of subordination stress induced hyperphagia associated with the development of obesity. Here we tested the hypotheses that the eating responses of subordinate mice recapitulate the BED and that limiting hyperphagia could prevent stress-associated metabolic changes. METHODS: Adult male mice were exposed to a model of chronic subordination stress associated with the automated acquisition of food intake and we performed a detailed meal pattern analysis. Additionally, using a pair-feeding protocol was test the hypothesis that the manifestation of obesity and the metabolic syndrome could be prevented by limiting hyperphagia. RESULTS: The architecture of feeding of subordinate mice was disrupted during the stress protocol due to disproportionate amount of food ingested at higher rate and with shorter satiety ratio than control mice. Subordinate mice hyperphagia was further exacerbated in response to either hunger or to the acute application of a social defeat. Notably, the obese phenotype but not the fasting hyperglycemia of subordinate mice was abrogated by preventing hyperphagia in a pair feeding paradigm. CONCLUSION: Overall these results support the validity of our chronic subordination stress to model binge eating disorder allowing for the determination of the underlying molecular mechanisms and the generation of testable predictions for innovative therapies, based on the understanding of the regulation and the control of food intake.

13.
Aging Cell ; 12(4): 695-705, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23648059

RESUMO

The contribution that oxidative damage to DNA and/or RNA makes to the aging process remains undefined. In this study, we used the hMTH1-Tg mouse model to investigate how oxidative damage to nucleic acids affects aging. hMTH1-Tg mice express high levels of the hMTH1 hydrolase that degrades 8-oxodGTP and 8-oxoGTP and excludes 8-oxoguanine from both DNA and RNA. Compared to wild-type animals, hMTH1-overexpressing mice have significantly lower steady-state levels of 8-oxoguanine in both nuclear and mitochondrial DNA of several organs, including the brain. hMTH1 overexpression prevents the age-dependent accumulation of DNA 8-oxoguanine that occurs in wild-type mice. These lower levels of oxidized guanines are associated with increased longevity and hMTH1-Tg animals live significantly longer than their wild-type littermates. Neither lipid oxidation nor overall antioxidant status is significantly affected by hMTH1 overexpression. At the cellular level, neurospheres derived from adult hMTH1-Tg neural progenitor cells display increased proliferative capacity and primary fibroblasts from hMTH1-Tg embryos do not undergo overt senescence in vitro. The significantly lower levels of oxidized DNA/RNA in transgenic animals are associated with behavioral changes. These mice show reduced anxiety and enhanced investigation of environmental and social cues. Longevity conferred by overexpression of a single nucleotide hydrolase in hMTH1-Tg animals is an example of lifespan extension associated with healthy aging. It provides a link between aging and oxidative damage to nucleic acids.


Assuntos
Comportamento Animal , Enzimas Reparadoras do DNA/metabolismo , Comportamento Exploratório , Regulação da Expressão Gênica no Desenvolvimento , Longevidade , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Senescência Celular , Enzimas Reparadoras do DNA/genética , Feminino , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oxirredução , Estresse Oxidativo , Monoéster Fosfórico Hidrolases/genética , Fatores de Tempo
14.
Psychoneuroendocrinology ; 38(12): 2933-42, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24060458

RESUMO

Stress and hypercaloric food are recognized risk factors for obesity, Metabolic Syndrome (MetS) and Type 2 Diabetes (T2D). Given the complexity of these metabolic processes and the unavailability of animal models, there is poor understanding of their underlying mechanisms. We established a model of chronic psychosocial stress in which subordinate mice are vulnerable to weight gain while dominant mice are resilient. Subordinate mice fed a standard diet showed marked hyperphagia, high leptin, low adiponectin, and dyslipidemia. Despite these molecular signatures of MetS and T2D, subordinate mice fed a standard diet were still euglycemic. We hypothesized that stress predisposes subordinate mice to develop T2D when synergizing with other risk factors. High fat diet aggravated dyslipidemia and the MetS thus causing a pre-diabetes-like state in subordinate mice. Contrary to subordinates, dominant mice were fully protected from stress-induced metabolic disorders when fed both a standard- and a high fat-diet. Dominant mice showed a hyperphagic response that was similar to subordinate but, unlike subordinates, showed a significant increase in VO2, VCO2, and respiratory exchange ratio when compared to control mice. Overall, we demonstrated a robust stress- and social status-dependent effect on the development of MetS and T2D and provided insights on the physiological mechanisms. Our results are reminiscent of the effect of the individual socioeconomic status on human health and provide an animal model to study the underlying molecular mechanisms.


Assuntos
Hiperfagia/psicologia , Resistência à Insulina , Síndrome Metabólica/metabolismo , Síndrome Metabólica/psicologia , Meio Social , Estresse Psicológico/psicologia , Adiponectina/metabolismo , Animais , Calorimetria Indireta , Diabetes Mellitus Tipo 2/sangue , Dieta , Dieta Hiperlipídica , Ingestão de Energia , Metabolismo Energético , Teste de Tolerância a Glucose , Hiperfagia/etiologia , Imuno-Histoquímica , Leptina/sangue , Masculino , Camundongos , Fatores de Risco , Predomínio Social , Estresse Psicológico/complicações
15.
FEMS Microbiol Lett ; 333(2): 146-52, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22640171

RESUMO

This study describes an efficient transformation system for the introduction of plasmid DNA into Bifidobacterium bifidum PRL2010 and Bifidobacterium asteroides PRL2011, for which to the best of our knowledge no transformation data have been reported previously. The method is based on electroporation of bifidobacterial cells, which were made competent by an optimized methodology based on varying media and growth conditions. Furthermore, the transformation protocol was applied in order to design a PRL2010-derivative, which carries antibiotic resistance against chloramphenicol and which was used to monitor PRL2010 colonization in a murine model.


Assuntos
Bifidobacterium/genética , DNA Bacteriano/genética , Eletroporação/métodos , Engenharia Genética/métodos , Transformação Bacteriana , Animais , Carga Bacteriana , Bifidobacterium/crescimento & desenvolvimento , Bifidobacterium/metabolismo , Cloranfenicol/metabolismo , Meios de Cultura/metabolismo , Farmacorresistência Bacteriana , Escherichia coli/genética , Escherichia coli/metabolismo , Fezes/microbiologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Plasmídeos/genética , Reprodutibilidade dos Testes , Especificidade da Espécie
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 35(6): 1461-71, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21093519

RESUMO

Exposure to stressful life events is intimately linked with vulnerability to neuropsychiatric disorders such as major depression. Pre-clinical animal models offer an effective tool to disentangle the underlying molecular mechanisms. In particular, the 129SvEv strain is often used to develop transgenic mouse models but poorly characterized as far as behavior and neuroendocrine functions are concerned. Here we present a comprehensive characterization of 129SvEv male mice's vulnerability to social stress-induced depression-like disorders and physiological comorbidities. We employed a well characterized mouse model of chronic social stress based on social defeat and subordination. Subordinate 129SvEv mice showed body weight gain, hyperphagia, increased adipose fat pads weight and basal plasma corticosterone. Home cage phenotyping revealed a suppression of spontaneous locomotor activity and transient hyperthermia. Subordinate 129SvEv mice also showed marked fearfulness, anhedonic-like response toward a novel but palatable food, increased anxiety in the elevated plus maze and social avoidance of an unfamiliar male mouse. A direct measured effect of the stressfulness of the living environment, i.e. the amount of daily aggression received, predicted the degree of corticosterone level and locomotor activity but not of the other parameters. This is the first study validating a chronic subordination stress paradigm in 129SvEv male mice. Results demonstrated remarkable stress vulnerability and establish the validity to use this mouse strain as a model for depression-like disorders.


Assuntos
Depressão/psicologia , Dominação-Subordinação , Camundongos da Linhagem 129/fisiologia , Camundongos da Linhagem 129/psicologia , Estresse Psicológico/psicologia , Agressão/fisiologia , Agressão/psicologia , Animais , Ansiedade/complicações , Ansiedade/psicologia , Doença Crônica , Corticosterona/sangue , Depressão/sangue , Depressão/complicações , Depressão/fisiopatologia , Modelos Animais de Doenças , Febre/complicações , Febre/fisiopatologia , Hiperfagia/complicações , Hiperfagia/psicologia , Masculino , Camundongos , Atividade Motora/fisiologia , Comportamento Social , Estresse Psicológico/sangue , Estresse Psicológico/complicações , Estresse Psicológico/fisiopatologia , Aumento de Peso
17.
Psychopharmacology (Berl) ; 208(1): 99-107, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19921154

RESUMO

BACKGROUND: The organophosphate chlorpyrifos (CPF) is a pesticide largely used worldwide. Studies from animal models indicate that CPF exposure during development at low doses can target different neurotransmitter systems in the absence of overt cholinergic effects. METHODS: Late gestational exposure (gestational days 14-17) to CPF at the dose of 6 mg/kg was evaluated in CD-1 mice at adulthood. Neurobehavioural effects likely involving serotonin (5-hydroxytryptamine, 5HT) transmission were assessed both in males and females, through the light-dark exploration test to assess CPF effects on anxiety profiles and the forced swimming test to evaluate the response to the 5HT transporter (5HTT) inhibitor fluvoxamine (30 mg/kg). In females only, we evaluated the effects of gestational exposure to CPF on maternal aggression, under basal condition or after injection of fluvoxamine. RESULTS: Gestational CPF exposure increased anxiety levels only in female mice, as shown by the augmented thigmotaxis behaviour and the lower latency to enter in the dark compartment. In the forced swimming test, no differences between CPF and control mice were found when assessed under basal condition (saline administration), but both male and female CPF mice missed to show the typical behavioural effects of the 5HTT inhibitor fluvoxamine. During maternal aggression, CPF females showed lower propensity to and intensity of aggressive behaviour, together with mild decreased responsiveness to fluvoxamine administration. CONCLUSIONS: Overall, the present results confirm a specific and sex-dependent vulnerability of affective/emotional domains to developmental CPF exposure. Furthermore, data provide clear indication on the disrupting effects of prenatal CPF on serotoninergic transmission.


Assuntos
Comportamento Animal/efeitos dos fármacos , Clorpirifos/toxicidade , Fluvoxamina/farmacologia , Inseticidas/toxicidade , Animais , Ansiedade/induzido quimicamente , Modelos Animais de Doenças , Feminino , Masculino , Exposição Materna , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Fatores Sexuais , Natação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA