Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Mucosal Immunol ; 11(1): 120-130, 2018 01.
Article in English | MEDLINE | ID: mdl-28401939

ABSTRACT

The multidrug resistance-1 (MDR1) gene encodes an ATP-dependent efflux transporter that is highly expressed in the colon. In mice, loss of MDR1 function results in colitis with similarities to human inflammatory bowel diseases (IBD). Here, we show that MDR1 has an unexpected protective role for the mitochondria where MDR1 deficiency results in mitochondrial dysfunction with increased mitochondrial reactive oxygen species (mROS) driving the development of colitis. Exogenous induction of mROS accelerates, while inhibition attenuates colitis in vivo; these effects are amplified in MDR1 deficiency. In human IBD, MDR1 is negatively correlated to SOD2 gene expression required for mROS detoxification. To provide direct evidential support, we deleted intestinal SOD2 gene in mice and showed an increased susceptibility to colitis. We exploited the genome-wide association data sets and found many (∼5%) of IBD susceptibility genes with direct roles in regulating mitochondria homeostasis. As MDR1 primarily protects against xenotoxins via its efflux function, our findings implicate a distinct mitochondrial toxin+genetic susceptibility interaction leading to mitochondrial dysfunction, a novel pathogenic mechanism that could offer many new therapeutic opportunities for IBD.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Colitis/genetics , Inflammation/genetics , Inflammatory Bowel Diseases/genetics , Intestines/immunology , Mitochondria/physiology , Superoxide Dismutase/genetics , Animals , Disease Models, Animal , Genetic Predisposition to Disease , Homeostasis , Humans , Metabolic Detoxication, Phase I/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism
2.
Diabetologia ; 54(9): 2392-403, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21667214

ABSTRACT

AIMS/HYPOTHESIS: Urocortins are the endogenous ligands for the corticotropin-releasing factor receptor type 2 (CRFR2), which is implicated in regulating energy balance and/or glucose metabolism. We determined the effects of chronic CRFR2 activation on metabolism in vivo, by generating and phenotyping transgenic mice overproducing the specific CRFR2 ligand urocortin 3. METHODS: Body composition, glucose metabolism, insulin sensitivity, energy efficiency and expression of key metabolic genes were assessed in adult male urocortin 3 transgenic mice (Ucn3(+)) under control conditions and following an obesogenic high-fat diet (HFD) challenge. RESULTS: Ucn3(+) mice had increased skeletal muscle mass with myocyte hypertrophy. Accelerated peripheral glucose disposal, increased respiratory exchange ratio and hypoglycaemia on fasting demonstrated increased carbohydrate metabolism. Insulin tolerance and indices of insulin-stimulated signalling were unchanged, indicating these effects were not mediated by increased insulin sensitivity. Expression of the transgene in Crfr2 (also known as Crhr2)-null mice negated key aspects of the Ucn3(+) phenotype. Ucn3(+) mice were protected from the HFD-induced hyperglycaemia and increased adiposity seen in control mice despite consuming more energy. Expression of uncoupling proteins 2 and 3 was higher in Ucn3(+) muscle, suggesting increased catabolic processes. IGF-1 abundance was upregulated in Ucn3(+) muscle, providing a potential paracrine mechanism in which urocortin 3 acts upon CRFR2 to link the altered metabolism and muscular hypertrophy observed. CONCLUSIONS/INTERPRETATION: Urocortin 3 acting on CRFR2 in skeletal muscle of Ucn3(+) mice results in a novel metabolically favourable phenotype, with lean body composition and protection against diet-induced obesity and hyperglycaemia. Urocortins and CRFR2 may be of interest as potential therapeutic targets for obesity.


Subject(s)
Dietary Fats/adverse effects , Hyperglycemia/metabolism , Hyperglycemia/prevention & control , Obesity/metabolism , Obesity/prevention & control , Urocortins/genetics , Urocortins/metabolism , Animals , Body Composition/drug effects , Body Composition/physiology , Dietary Fats/pharmacology , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glucose/metabolism , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Phenotype , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
3.
J Neuroendocrinol ; 21(11): 879-87, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19602102

ABSTRACT

Inter-individual differences in hypothalamic-pituitary-adrenal (HPA) axis activity underlie differential vulnerability to neuropsychiatric and metabolic disorders, although the basis of this variation is poorly understood. 11beta-Hydroxysteroid dehydrogenase type 1 (11beta-HSD1) has previously been shown to influence HPA axis activity. 129/MF1 mice null for 11beta-HSD1 (129/MF1 HSD1(-/-)) have greatly increased adrenal gland size and altered HPA activity, consistent with reduced glucocorticoid negative feedback. On this background, concentrations of plasma corticosterone and adrenocorticotrophic hormone (ACTH) were elevated in unstressed mice, and showed a delayed return to baseline after stress in HSD1-null mice with reduced sensitivity to exogenous glucocorticoid feedback compared to same-background genetic controls. In the present study, we report that the genetic background can dramatically alter this pattern. By contrast to HSD1(-/-) mice on a 129/MF1 background, HSD1(-/-) mice congenic on a C57Bl/6J background have normal basal plasma corticosterone and ACTH concentrations and exhibit normal return to baseline of plasma corticosterone and ACTH concentrations after stress. Furthermore, in contrast to 129/MF1 HSD1(-/-) mice, C57Bl/6J HSD1(-/-) mice have increased glucocorticoid receptor expression in areas of the brain involved in glucocorticoid negative feedback (hippocampus and paraventricular nucleus), suggesting this may be a compensatory response to normalise feedback control of the HPA axis. In support of this hypothesis, C57Bl/6J HSD1(-/-) mice show increased sensitivity to dexamethasone-mediated suppression of peak corticosterone. Thus, although 11beta-HSD1 appears to contribute to regulation of the HPA axis, the genetic background is crucial in governing the response to (and hence the consequences of) its loss. Similar variations in plasticity may underpin inter-individual differences in vulnerability to disorders associated with HPA axis dysregulation. They also indicate that 11beta-HSD1 inhibition does not inevitably activate the HPA axis.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adrenal Glands/pathology , Adrenocorticotropic Hormone/blood , Animals , Base Sequence , Circadian Rhythm , Corticosterone/blood , DNA Primers , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Polymerase Chain Reaction
4.
FASEB J ; 22(11): 3896-907, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18697839

ABSTRACT

Glucocorticoid hormones are critical to respond and adapt to stress. Genetic variations in the glucocorticoid receptor (GR) gene alter hypothalamic-pituitary-adrenal (HPA) axis activity and associate with hypertension and susceptibility to metabolic disease. Here we test the hypothesis that reduced GR density alters blood pressure and glucose and lipid homeostasis and limits adaption to obesogenic diet. Heterozygous GR(betageo/+) mice were generated from embryonic stem (ES) cells with a gene trap integration of a beta-galactosidase-neomycin phosphotransferase (betageo) cassette into the GR gene creating a transcriptionally inactive GR fusion protein. Although GR(betageo/+) mice have 50% less functional GR, they have normal lipid and glucose homeostasis due to compensatory HPA axis activation but are hypertensive due to activation of the renin-angiotensin-aldosterone system (RAAS). When challenged with a high-fat diet, weight gain, adiposity, and glucose intolerance were similarly increased in control and GR(betageo/+) mice, suggesting preserved control of intermediary metabolism and energy balance. However, whereas a high-fat diet caused HPA activation and increased blood pressure in control mice, these adaptions were attenuated or abolished in GR(betageo/+) mice. Thus, reduced GR density balanced by HPA activation leaves glucocorticoid functions unaffected but mineralocorticoid functions increased, causing hypertension. Importantly, reduced GR limits HPA and blood pressure adaptions to obesogenic diet.


Subject(s)
Blood Pressure/drug effects , Diet/adverse effects , Dietary Fats/adverse effects , Hypertension/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/metabolism , Adiposity/drug effects , Adiposity/genetics , Aldosterone/metabolism , Angiotensins/metabolism , Animals , Blood Glucose/metabolism , Cell Line , Dietary Fats/pharmacology , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Humans , Hypertension/chemically induced , Hypertension/genetics , Lipid Metabolism/genetics , Mice , Mice, Transgenic , Receptors, Glucocorticoid/genetics , Renin/metabolism , Weight Gain/drug effects , Weight Gain/genetics
5.
J Neuroendocrinol ; 17(6): 387-93, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15929744

ABSTRACT

The hypothalamus appears to be more responsive to ghrelin and growth hormone secretagogues (GHS) in fasting, as reflected by a two- to three-fold increase in the number of cells detected that express Fos protein in the arcuate nucleus, in 48-h fasted rats compared to fed controls. Moreover, this increased hypothalamic responsiveness to GHS in fasting is regulated by the central action of exogenous leptin and insulin, although it is unknown whether these hormones mediate the changes in hypothalamic responsiveness to GHS associated with the fasting/fed state. In the present study, we show that refeeding with normal rat chow for only 2 h at the end of a 48-h fast reversed the potentiation of the Fos response to GHRP-6 observed in fasted rats. Circulating leptin and insulin levels remained significantly lower in refed rats compared to ad lib-fed rats, suggesting that the change in the hypothalamic sensitivity brought about by refeeding was independent of these hormones. By contrast, 2 h of chow refeeding at the end of a fast restored plasma glucose levels to those of the fed state. Refeeding with sugar alone for 2 h at the end of a 48-h fast also reduced the potentiated Fos response in fasting, indicating that elevated blood glucose can influence the central responsiveness to ghrelin/GHS. By contrast, infusion of the ileal satiety factor, PYY(3-36) (known to increase postprandially) did not alter the central responsiveness to GHRP-6, although it suppressed feeding and body weight as expected. This study highlights the importance of nutritional status in regulating the action of exogenous GHS (and presumably endogenous ghrelin) on the hypothalamic circuits controlling food intake.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/physiology , Eating/physiology , Fasting/physiology , Oligopeptides/pharmacology , Animal Feed , Animals , Blood Glucose , Carbohydrates/pharmacology , Ghrelin , Insulin/blood , Leptin/blood , Male , Molecular Mimicry , Oligopeptides/chemistry , Peptide Fragments , Peptide Hormones/chemistry , Peptide YY/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar
6.
Neuroscience ; 126(1): 9-19, 2004.
Article in English | MEDLINE | ID: mdl-15145069

ABSTRACT

Exposure of the rat to restraint results in activation of the hypothalamic-pituitary-adrenal (HPA) axis, a characteristic pattern of c-fos expression in the brain and increased cardiovascular function. These responses adapt with repeated exposure of an individual to the same stress. Corticosterone secretion habituates, and c-fos mRNA expression in the paraventricular nucleus of the hypothalamus (PVN) decreases. The increased expression of corticotropin releasing hormone mRNA in the PVN also becomes less prominent, whereas vasopressin mRNA progressively increases. The neural mechanisms responsible for this adaptation remain obscure. Because of its role in conditioned learning, we have hypothesised that the amygdala might be involved in this adaptive process. Here we show that large neurotoxic lesions of the amygdala in male rats do not prevent acute stress activation of the HPA axis following 30 min restraint, whilst more discrete lesions of the central nucleus actually exacerbate the acute response. Rats with large amygdala lesions demonstrate delayed habituation of corticosterone and c-fos to repeated restraint, an affect not apparent with central nucleus lesions. Furthermore we show that neither type of lesion significantly reduced tachycardiac responses to single or repeated restraint as measured by telemetry. We conclude that the amygdala and the central nucleus are not necessary for HPA and cardiovascular activation in response to stress (though the central nucleus may modulate it), and that adaptation to repeated stress is only modestly dependent upon the amygdala.


Subject(s)
Adaptation, Physiological/physiology , Amygdala/physiology , Stress, Physiological/physiopathology , Acute Disease , Animals , Denervation , Heart Rate , Male , Paraventricular Hypothalamic Nucleus/physiology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/analysis , Rats , Rats, Inbred Strains , Recurrence , Restraint, Physical
7.
Biochem Biophys Res Commun ; 311(4): 915-9, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14623268

ABSTRACT

It has recently been suggested that gut-derived PYY(3-36) may be involved in the central mediation of post-prandial satiety signals. We have examined the acute effects of peripherally administered PYY(3-36) on food intake and hypothalamic gene expression of neuropeptides in mice. A single intraperitoneal injection of PYY(3-36) to mice that had been fasted for 24h resulted in a highly significant reduction in food intake at 6 and 24h post-injection but not at 48h. However, in freely fed mice, food intake was unaltered by PYY(3-36) administration. In the arcuate nucleus POMC mRNA expression was significantly elevated at 6h and remained elevated at 24h following PYY(3-36) injection. By contrast NPY mRNA expression in the arcuate nucleus was suppressed at 6h but not at 24h post-injection. In the lateral hypothalamus there were no differences in MCH mRNA expression at either time point. In conclusion, peripherally administered PYY(3-36) has a suppressive effect on food intake that is more prominent in recently fasted mice and lasts up to 24 h. This is associated with a short-lived suppression of NPY mRNA, a longer lasting increase in POMC mRNA but no change in MCH mRNA expression.


Subject(s)
Appetite Regulation/drug effects , Appetite Regulation/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Neuropeptides/metabolism , Peptide Fragments/administration & dosage , Peptide YY/administration & dosage , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Hypothalamic Hormones/metabolism , Hypothalamus/cytology , Injections, Intraperitoneal , Melanins/metabolism , Mice , Neuropeptide Y/metabolism , Pituitary Hormones/metabolism , Pro-Opiomelanocortin/metabolism
8.
Med Sci Sports Exerc ; 31(5): 670-4, 1999 May.
Article in English | MEDLINE | ID: mdl-10331886

ABSTRACT

PURPOSE: An athlete's ability to repeatedly perform at high intensities during intermittent exercise could be related to an accelerated plasma lactate removal ability during recovery periods. METHODS: We determined the decline in plasma lactate levels during passive recovery after an incremental exercise test to exhaustion on a bicycle ergometer in five trained and five untrained male subjects. Venous blood samples were taken during exercise and recovery for the analysis of plasma lactate concentration. The endurance fitness of the subjects was characterized using a variable known as the maximum turn point power output (MTP), measured in W x kg(-1). MTP describes the workload at which lactate levels rise significantly above resting concentrations. RESULTS: The decline in plasma lactate levels during recovery was determined at selected intervals from the exponential recovery curve plotted as a percentage of peak plasma lactate versus time. No significant relationships were found between the recovery parameters measured from the curve and the MTP values of these subjects (Spearman's rank order correlation; r(s) values from -0.042 to -0.31). CONCLUSIONS: Therefore, we can conclude that training confers no advantage to the decline in plasma lactate while recovering passively from exercise at equivalent relative maximal work intensities.


Subject(s)
Bicycling/physiology , Exercise/physiology , Lactic Acid/blood , Adult , Humans , Male , Oxygen Consumption/physiology , Physical Endurance/physiology , Respiratory Mechanics/physiology , Time Factors
9.
N Z Med J ; 112(1085): 127, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10326804
10.
J Appl Physiol (1985) ; 84(3): 877-83, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9480946

ABSTRACT

We measured brain and abdominal temperatures in eight male Sprague-Dawley rats (350-450 g) exercising voluntarily to a point of fatigue in two hot environments. Rats exercised, at the same time of the day, in three different trials, in random order: rest 23 degrees C, exercise 33 degrees C; rest 23 degrees C, exercise 38 degrees C; and rest 38 degrees C, exercise 38 degrees C. Running time to fatigue was 29.4 +/- 5.9 (SD), 22.1 +/- 3.7, and 14.3 +/- 2.9 min for the three trials, respectively. Abdominal temperatures, measured with intraperitoneal radiotelemeters, at fatigue in the three trials (39.9 +/- 0.3, 39.9 +/- 0.3, and 39.8 +/- 0.3 degrees C, respectively) were not significantly different from each other. Corresponding brain temperatures, measured with thermocouples in the hypothalamic region (40.2 +/- 0.4, 40.2 +/- 0.4, and 40.1 +/- 0.4 degrees C), also did not differ. Our results are consistent with the concept that there is a critical level of body temperature beyond which animals will not continue to exercise voluntarily in the heat. Also, in our study, brain temperature was higher than abdominal temperature throughout exercise; that is, selective brain cooling did not occur when body temperature was below the level limiting exercise.


Subject(s)
Abdomen/physiology , Body Temperature/physiology , Brain/physiology , Hot Temperature/adverse effects , Muscle Fatigue/physiology , Physical Exertion/physiology , Animals , Body Temperature Regulation/physiology , Hypothalamus/physiology , Male , Rats , Rats, Sprague-Dawley , Telemetry
14.
Mol Biochem Parasitol ; 65(1): 11-22, 1994 May.
Article in English | MEDLINE | ID: mdl-7935618

ABSTRACT

The temporal expression during gametogenesis and the cellular location of the sexual stage specific protein Pfs16, a putative integral membrane protein of Plasmodium falciparum, was investigated using two monoclonal antibodies, 2G7 and 93A3A2. Using sorbitol synchronised, in vitro gametocyte cultures along with immunofluorescence assays, the time at which Pfs16 is first expressed during gametogenesis has been estimated to 35 hours post merozoite invasion. By immunofluorescence assays on thin blood smears monoclonal antibodies specific for Pfs16 react strongly with the gametocyte and also with vesicles within the red blood cell cytoplasm, many of which connect with the gametocyte cell. Purification of parasitophorous vacuole membranes from mature and immature gametocytes and immunoelectron microscopy on gametocytes during gametogenesis have allowed us to locate Pfs16 to the parasitophorous vacuole membrane. During gametogenesis this membrane is shed along with the red blood cell membrane. Immunofluorescence assays and immunoelectron microscopy studies of emerged gametes indicate that in a minority of cases the parasitophorous vacuole membrane along with Pfs16 can be retained to some extent on the gamete surface.


Subject(s)
Antigens, Protozoan/metabolism , Membrane Proteins/metabolism , Plasmodium falciparum/immunology , Animals , Antibodies, Monoclonal , Female , Fluorescent Antibody Technique , Male , Microscopy, Immunoelectron , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Subcellular Fractions/immunology , Subcellular Fractions/metabolism
18.
J Clin Orthod ; 23(9): 636-7, 1989 Sep.
Article in English | MEDLINE | ID: mdl-2639155
19.
J Clin Orthod ; 21(8): 530-2, 1987 Aug.
Article in English | MEDLINE | ID: mdl-3482088
SELECTION OF CITATIONS
SEARCH DETAIL
...