• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 106
  • 72
  • 14
  • 5
  • 4
  • 3
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 238
  • 238
  • 238
  • 108
  • 94
  • 71
  • 38
  • 33
  • 33
  • 28
  • 27
  • 25
  • 23
  • 22
  • 22
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
71

Dieta hiperlipídica, inflamação e programação metabólica : efeitos na sinalização de insulina em camundongos recém-desmamados e adultos / High-fat diet, inflammation and metabolic programming : effects on insulin signaling in newly weaned and adult offspring of mice

Fante, Thaís de, 1990- 27 August 2018 (has links)
Orientadores: Adriana Souza Torsoni, Marciane Milanski / Dissertação (mestrado) - Universidade Estadual de Campinas, Faculdade de Ciências Aplicadas / Made available in DSpace on 2018-08-27T16:45:13Z (GMT). No. of bitstreams: 1 Fante_Thaisde_M.pdf: 3029616 bytes, checksum: 5e68ab17d3076784e4f20c127d76838f (MD5) Previous issue date: 2015 / Resumo: O estilo de vida moderno tem levado ao aumento na prevalência de obesidade e suas co-morbidades em gestantes e na população cada vez mais jovem. Muitos dos efeitos do consumo direto de dieta hiperlipídica (DH) no metabolismo de glicose e lipídios já são bem estabelecidos. No entanto, considera-se importante avaliar se o consumo de DH durante períodos críticos do desenvolvimento seria capaz de ativar mecanismos epigenéticos, perpetuando mudanças no metabolismo da prole e criando um ciclo vicioso que não poderia ser interrompido. O objetivo desse estudo foi avaliar o efeito potencial da programação metabólica em prejudicar a sinalização de insulina na prole recém desmamada de mães alimentadas com dieta hiperlipídica durante a gestação e lactação. Além disso, investigamos se a exposição precoce a um ambiente obesogênico seria capaz de exacerbar o prejuízo no metabolismo de glicose na vida adulta de animais reexpostos à dieta hiperlipídica. Para isso, camundongos fêmeas da linhagem Swiss foram alimentados com dieta controle ou DH durante os períodos de adaptação, gestação e lactação, e os tecidos da prole macho foram analisados nos dias 28 e 82. Os resultados mostram que a prole de mães obesas (HC-O) apresentou maior ganho de peso, adiposidade e ingestão alimentar que a prole de mães controle (CC-O). Além do mais, apresentou prejuízos na sinalização de insulina em tecidos periféricos como fígado, adiposo e músculo, e centrais, como o hipotálamo, provavelmente devido à maior ativação de vias inflamatórias. A reexposição à DH parece agir como um fator agravante para o desenvolvimento do fenótipo obeso, levando a resistência sistêmica à insulina e hiperleptinemia. É válido ressaltar que o tecido adiposo parece ser o tecido mais afetado na prole adulta após a reexposição da dieta (HH-O), o que pode contribuir para a desregulação metabólica observada. Em conjunto, nossos resultados sugerem que o consumo materno de dieta hiperlipídica durante a gestação e lactação pode ocasionar alterações no metabolismo glicídico da prole tanto em animais recém desmamados quanto adultos. Por fim, a obesidade materna leva à maior susceptibilidade ao desenvolvimento de obesidade e prejuízos na sinalização de insulina na prole que não podem ser revertidos pelo consumo de uma dieta controle, no entanto, podem ser agravados especialmente quando os animais são reexpostos à DH / Abstract: Modern lifestyle has resulted in an increase in the prevalence of obesity and its comorbidities in pregnancy and young population. Many effects from direct consumption of a high-fat diet (HFD) on glucose and lipid metabolism are well established. However, it is important to assess whether maternal consumption of HFD during critical periods of development can trigger epigenetic mechanisms, perpetuating changes in offspring metabolism and creating a vicious circle that cannot be broken. This study evaluated the potential effect of metabolic programming in impairing the insulin signaling in recently weaned offspring of obese dams. In addition, we investigated if early exposure to obesogenic environment is able to exacerbate the impairment of glucose metabolism in adult life in response to a high-fat diet. For this, Swiss female mice were fed with Stardard chow (SC) or HFD before and during mating, gestation and lactation. Tissues from male offspring were obtained at d28 and d82 to analyze activation of key proteins of inflammatory and insulin signaling pathways by Western Blot. Offspring of obese dams (HC-O) showed greater weight gain, adiposity and food intake than offspring of control dams (CC-O). Furthermore, they showed impairment in insulin signaling in central and peripheral tissues, associated to increased activation of inflammatory pathways. The HFD re-exposure seems to be an aggravating factor in development of obese phenotype leading to systemic insulin resistance and hyperleptinaemia. Moreover, adipose tissue was ultimately the most affected tissue in adult offspring after HFD rechallenged (HH-O) which may have contributed to the metabolic deregulation observed. Together our results suggest that maternal consumption of high-fat diet during pregnancy and lactation can cause changes in glucose metabolism of offspring in both weaned and adult animals. Additionally, maternal obesity leads to increase susceptibility to the development of obesity and impairment in insulin signaling in offspring that cannot be reversed by SC consumption, but can be aggravated especially when re-exposed to HFD / Mestrado / Metabolismo e Biologia Molecular / Mestra em Ciências da Nutrição e do Esporte e Metabolismo
72

Evaluation of an Organic Mineral Complex on the Development of Cardiovascular Disease Risk Following a 10-week High-Fat Diet

January 2020 (has links)
abstract: According to the World Health Organization, obesity has nearly tripled since 1975 and forty-one million children under the age of 5 are overweight or obese (World Health Organization, 2018). Exercise is a potential intervention to prevent obesity-induced cardiovascular complications as exercise training has been shown to aid nitric oxide (NO) production as well as preserving endothelial function in obese mice (Silva et al., 2016). A soil-derived organic mineral compound (OMC) has been shown to lower blood sugar in diabetic mice (Deneau et al., 2011). Prior research has shown that, while OMC did not prevent high fat diet (HFD)-induced increases in body fat in male Sprague-Dawley rats, it was effective at preventing HFD-induced impaired vasodilation (M. S. Crawford et al., 2019). Six-weeks of HFD has been shown to impair vasodilation through oxidative-stress mediated scavenging of NO as well as upregulation of inflammatory pathways including inducible nitric oxide synthase (iNOS) and cyclooxygenase (Karen L. Sweazea et al., 2010). Therefore, the aim of the present study was to determine whether OMC alters protein expression of iNOS and endothelial NOS (eNOS) in the vasculature of rats fed a control or HFD with and without OMC supplementation. Six-week old male Sprague-Dawley rats were fed either a standard chow diet (CHOW) or a HFD composed of 60% kcal from fat for 10 weeks. The rats were administered OMC at doses of 0 mg/mL (control), 0.6 mg/mL, or 3.0 mg/mL added to their drinking water. Following euthanasia with sodium pentobarbital (200 mg/kg, i.p.), mesenteric arteries and the surrounding perivascular adipose tissue were isolated and prepared for Western Blot analyses. Mesenteric arteries from HFD rats had more uncoupled eNOS (p = 0.006) and iNOS protein expression (p = 0.027) than rats fed the control diet. OMC was not effective at preventing the uncoupling of eNOS or increase in iNOS induced by HFD. Perivascular adipose tissue (PVAT) showed no significant difference in iNOS protein expression between diet or OMC treatment groups. These findings suggest that OMC is not likely working through the iNOS or eNOS pathways to improve vasodilation in these rats, but rather, appears to be working through another mechanism. / Dissertation/Thesis / Masters Thesis Biology 2020
73

The Effects of Adolescent High Fat Diet on Adult Prefrontal Cortex-Dependent Behavior, Stress Responsivity, and Microglial Reactivity,

Lloyd, Kelsey 29 September 2021 (has links)
No description available.
74

Metabolické účinky chronického podávání metforminu u obézních myší v závislosti na složení vysokotukové diety / Metabolic effects of chronic metformin administration in obese mice depending on the composition of high-fat diet

Roubalová, Jana January 2011 (has links)
Obesity leads to many severe metabolic disorders, e.g. dyslipidemia, insulin resistance, ectopic fat accumulation in the liver and skeletal muscles, non-alcoholic fatty liver disease and finally diabetes mellitus type 2. Metformin (1,1-dimethylbiguanide) is the most favored medicament for the treatment and prevention of these disorders. It stimulates cellular glucose uptake and normalizes blood levels of lipid metabolites without triggering insulin secretion. Research on insulin resistance and diabetes is often realized through developing diet- induced obesity in laboratory animals. The aim of this project is to compare metabolic effects of two different high-fat diets named HFD and HSD. The HFD diet consists chiefly of n-6 polyunsaturated fatty acids (corn oil) and starch (100% glucose). The HSD diet contains mainly saturated fatty acids (lard) and sucrose (50% glucose and 50% fructose). I also studied metabolic effects of metformin by adding it continuously to the drinking water given to obese mice fed with the HFD or the HSD diet. Methods: Intraperitoneal glucose tolerance test (IPGTT), blood and tissue levels of lipid metabolites assessment, radio-immunological assessment of blood levels of insulin, assessment of AMPK activity in liver by western blotting. Results: Increased consumption of the...
75

Hepatic NAD salvage pathway is enhanced in mice on a high-fat diet

Penke, Melanie, Larsen, Per S., Schuster, Susanne, Dall, Morten, Jensen, Benjamin A.H., Gorski, Theresa, Meusel, Andrej, Richter, Sandy, Vienberg, Sara G., Treebak, Jonas T., Kiess, Wieland, Garten, Antje 02 March 2020 (has links)
Nicotinamide phosphoribosyltransferase (Nampt) is the rate-limiting enzyme for NAD salvage and the abundance of Nampt has been shown to be altered in non-alcoholic fatty liver disease. It is, however, unknown how hepatic Nampt is regulated in response to accumulation of lipids in the liver of mice fed a high-fat diet (HFD). HFD mice gained more weight, stored more hepatic lipids and had an impaired glucose tolerance compared with control mice. NAD levels as well as Nampt mRNA expression, protein abundance and activity were significantly increased in HFD mice. Enhanced NAD levels were associated with deacetylation of p53 and Nfκb indicating increased activation of Sirt1. Despite impaired glucose tolerance and increased hepatic lipid levels in HFD mice, NAD metabolism was significantly enhanced. Thus, improved NAD metabolism may be a compensatory mechanism to protect against negative impact of hepatic lipid accumulation.
76

Multi-Faceted Mechanisms of Exercise to Improve Metabolic and Cardiac Health

Pinckard, Kelsey Marie January 2021 (has links)
No description available.
77

High-fat diet attenuates Alzheimer's disease-related pathology and cognitive impairment in a transgenic mouse model

Merkel, Jonathan Leonard 05 May 2022 (has links)
This study investigated the effect of high-fat diet consumption on Alzheimer's disease-related pathology in the Tg6799 mouse model. We found that high-fat diet attenuates Alzheimer's disease-related pathology and cognitive dysfunction in Tg6799 mice and propose decreased beta-amyloid deposition due to improved autophagic-lysosomal flux as a potential, underlying mechanism.
78

The Effects of Obesity and Exercise on Healthspan, Cancer Incidence, and Lifespan in a Mouse Model of Radiation-Induced Cancer

Farber, Eadan 18 September 2020 (has links)
Background: The number of cancer survivors across North America is increasing, with estimates indicating that this population will grow to nearly 19 million individuals by the end of 2020. This increase can be attributed, in part, due to improvements in cancer treatments, of which radiation therapy is most commonly used. Unfortunately, exposure to radiation also increases the risk of secondary cancer development long-term. Moreover, obesity and physical inactivity are prevalent, modifiable, risk factors among cancer survivors, with both factors being linked to decrements in quality of life, increased cancer risk, and greater mortality risk. To date, there has been promising epidemiological and clinical data highlighting the role of exercise as a way to mitigate cancer risk and improve survival; however, longitudinal studies are lacking and the effects of radiation in these studies have been largely ignored. Therefore, there is a major clinical need to directly evaluate the combinatory long-term effects of radiation, exercise, and/or obesity to reveal their implications on healthspan, cancer incidence, and survival. Recent pre-clinical work from our group has shown that after being exposed to radiation, endurance exercise prevented several negative alterations to hematopoietic stem cells and their niche caused by high-fat diet (HFD)-induced obesity. We also showed that leukemic blast viability in vitro was greater when cultured in bone marrow supernatant from mice with HFD-induced obesity compared to bone marrow supernatant from mice without HFD-induced obesity. It is unknown, however, whether these findings extend to alterations in cancer risk across the lifespan. As such, the purpose of this study was to evaluate the effects of lifelong exercise and diet-induced obesity on healthspan, cancer incidence, and survival in an established mouse model of radiation-induced cancer. Methods: Male CBA mice (n=80) were randomly divided into either a control diet (CTRL; n=40) of 45% high-fat diet (HFD; n=40) and then further divided into either a sedentary group (SED; n=20) or exercise-trained group (EX; n=20). At age 13 weeks, all mice were exposed to a cancer inducing dose of whole-body ionizing radiation (3 Gy). A healthspan index score and endpoint monitoring were conducted throughout the study by blinded investigators. Results: When normalized to CTRL/SED, the highest healthspan score was in the CTRL/EX (score = +2.5), followed by HFD/EX (score = +1) and lastly HFD/SED (score = -0.5). Cancer incidence was significantly higher in the HFD/SED group when compared to the CTRL/EX group (p<0.05) and a trend for higher cancer incidence for HFD/SED was observed when compared to the CTRL/SED group (p=0.079). There was no significant difference between the HFD/SED and HFD/EX group in cancer incidence (p>0.05). Overall survival was significantly higher in the HFD/SED group compared to CTRL/SED group (p<0.05); however, risk of cancer-related mortality was 1.6-times higher in the HFD/SED group compared to the CTRL/SED group (RR=1.60; 95% CI, 1.00-2.56; p=0.0495) and 1.68-times that of the CTRL/EX group (RR=1.68; 95% CI, 1.02-2.78; p=0.0415). Conclusion: Our findings show that lifelong exercise training resulted in higher healthspan index, lower cancer incidence, and lower risk of cancer-related mortality following radiation exposure, with these effects being largely reversed by HFD-induced obesity. This study provides the rationale for future studies to uncover cellular and molecular mechanisms that could be underlying these results. Moreover, this study presents a proof of concept for the consideration of clinical studies in cancer survivors examining exercise as an intervention to reduce the long-term effects of radiation.
79

Dysregulation of of phospholipid-specific phagocytosis by B1 B cells in diet-induced obese mice

Vo, Hung 22 January 2016 (has links)
B1 B cells have received increasing attention recently due to their newly discovered phagocytic and microbicidal capabilities. Several studies have demonstrated that B1 cells can phagocytize polystyrene fluorescent particles, bacteria (Staphylococcus aureus, Escherichia coli), and even apoptotic cells. Nevertheless, little is known about the biological significance of this seemingly redundant function of B1 B cells as compared to that of conventional phagocytes. Here we investigate the unique phosphotidylcholine (PtC)-specific B1 B cell phagocytosis. PtC is a major phospholipid in the biological membrane and a classical antigen recognized by B1 B cell-derived natural antibodies. These antibodies play important roles in immune defense as well as tissue homeostasis. Here we report that B1 cells preferentially phagocytose PtC-coated beads, differing from that of conventional macrophages. We further attest that these beads were truly internalized and subsequently fused with hydrolytic lysosomes indicated by increasing fluorescent intensity of a pH-sensitive dye. Despite the differences in antigen specificity, phagocytosis of both B1 cells and macrophages can be inhibited by the microtubule-inhibitor, Colchicine, in a dose-dependent manner. Most intriguingly, upon chronic high-fat diet (HFD) consumption by the host, B1 cell phagocytosis starts to lose antigen-specificity for PtC. Morphologically, some of these B1 B cells in DIO mice show enlarged cytosol and engulfed more beads, indicating a transition to macrophage-like cells. Our study suggests for the first time that B1 B cells have unique phospholipid-specific phagocytosis capacity, which is affected by diet-induced obesity.
80

Role of DHS in translation control of islet β-cell replication during high fat induced obesity and glucose intolerance

Levasseur, Esther Marie 12 July 2017 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Insulin resistance in liver, muscle, and adipose tissue almost invariably occurs during obesity. To compensate, the insulin-producing β-cell increases insulin production by expanding cellular mass. The inability of the β-cell to fully compensate leads to hyperglycemia and ultimately type 2 diabetes. The enzyme deoxyhypusine synthase (DHS) catalyzes the spermidine-dependent posttranslational modification of Lys50 of eukaryotic translation initiation factor 5A (eIF5A) to form hypusine (Hyp). Studies have demonstrated this modification of eIF5A to contribute to cellular proliferation in cancerous cells, but its role in the physiologic proliferation of islet β-cells is unknown. I hypothesized eIF5A-Hyp to be required for the proliferation of islet β cells during the early phase of insulin resistance, allowing the β-cell to respond to the increased demand for insulin to maintain glucose homeostasis. To test this hypothesis, deletion of DHS was induced post-developmentally in β-cells by crossing Dhs-fl/fl mice with MIP1-CreERT mice, and animals were fed for 1 or 4 weeks with a 60% kcal from fat diet (HFD) or normal chow diet (NCD, 16% kcal from fat diet). NCD-fed and HFD-fed animals had normal glucose homeostasis after one week feeding, regardless of genotype. However, after 4 weeks of HFD, KO mice had significantly worse glucose intolerance compared to control mice. eIF5A-Hyp levels increased in β-cells of control animals and as expected remained low in the KO mice. β-cell proliferation was significantly increased after 1 week of HFD as measured by PCNA staining, however KO mice showed no increase. Cyclin D2 protein, but not mRNA, was increased in control animals fed a HFD; this protein increase was not observed in KO animals. Furthermore, polyribosomal profile of isolated islets of 1 week HFD-fed mice showed the Ccnd2 mRNA bound to the monoribosome fractions in the KO animals compared to the controls, resulting in changes of global translation. Interestingly, Ccnd1 polyribosome to monoribosome ratio showed no changes in translation compared to Ccnd2. Taken together, these results suggest that DHS (and, consequently, eIF5A-Hyp) is necessary for the adaptive proliferative and functional response of β-cells during high fat diet induced obesity and glucose intolerance.

Page generated in 0.066 seconds