Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters










Publication year range
1.
NPJ Aging Mech Dis ; 7(1): 14, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34210964

ABSTRACT

To identify potential factors influencing age-related cognitive decline and disease, we created MindCrowd. MindCrowd is a cross-sectional web-based assessment of simple visual (sv) reaction time (RT) and paired-associate learning (PAL). svRT and PAL results were combined with 22 survey questions. Analysis of svRT revealed education and stroke as potential modifiers of changes in processing speed and memory from younger to older ages (ntotal = 75,666, nwomen = 47,700, nmen = 27,966; ages 18-85 years old, mean (M)Age = 46.54, standard deviation (SD)Age = 18.40). To complement this work, we evaluated complex visual recognition reaction time (cvrRT) in the UK Biobank (ntotal = 158,249 nwomen = 89,333 nmen = 68,916; ages 40-70 years old, MAge = 55.81, SDAge = 7.72). Similarities between the UK Biobank and MindCrowd were assessed using a subset of MindCrowd (UKBb MindCrowd) selected to mirror the UK Biobank demographics (ntotal = 39,795, nwomen = 29,640, nmen = 10,155; ages 40-70 years old, MAge = 56.59, SDAge = 8.16). An identical linear model (LM) was used to assess both cohorts. Analyses revealed similarities between MindCrowd and the UK Biobank across most results. Divergent findings from the UK Biobank included (1) a first-degree family history of Alzheimer's disease (FHAD) was associated with longer cvrRT. (2) Men with the least education were associated with longer cvrRTs comparable to women across all educational attainment levels. Divergent findings from UKBb MindCrowd included more education being associated with shorter svRTs and a history of smoking with longer svRTs from younger to older ages.

2.
Sci Rep ; 11(1): 10248, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986309

ABSTRACT

Vascular contributions to cognitive impairment and dementia (VCID) include structural and functional blood vessel injuries linked to poor neurocognitive outcomes. Smoking might indirectly increase the likelihood of cognitive impairment by exacerbating vascular disease risks. Sex disparities in VCID have been reported, however, few studies have assessed the sex-specific relationships between smoking and memory performance and with contradictory results. We investigated the associations between sex, smoking, and cardiovascular disease with verbal learning and memory function. Using MindCrowd, an observational web-based cohort of ~ 70,000 people aged 18-85, we investigated whether sex modifies the relationship between smoking and cardiovascular disease with verbal memory performance. We found significant interactions in that smoking is associated with verbal learning performance more in women and cardiovascular disease more in men across a wide age range. These results suggest that smoking and cardiovascular disease may impact verbal learning and memory throughout adulthood differently for men and women.


Subject(s)
Cigarette Smoking/adverse effects , Memory/drug effects , Verbal Learning/drug effects , Adult , Aged , Aged, 80 and over , Cigarette Smoking/psychology , Cognition/drug effects , Cognition/physiology , Cognitive Dysfunction/physiopathology , Cohort Studies , Dementia, Vascular/etiology , Female , Humans , Male , Memory/physiology , Middle Aged , Sex Factors , Verbal Learning/physiology
3.
Diabetes Obes Metab ; 16 Suppl 1: 49-55, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25200296

ABSTRACT

Fatty acid (FA)-sensitive neurons are present in the brain, especially the hypothalamus, and play a key role in the neural control of energy homeostasis. Through neuronal output, FA may modulate feeding behaviour as well as insulin secretion and action. Subpopulations of neurons in the ventromedial and arcuate hypothalamic nuclei are selectively either inhibited or activated by FA. Molecular effectors of these FA effects probably include chloride or potassium ion channels. While intracellular metabolism and activation of the ATP-sensitive K⁺ channel appear to be necessary for some of the signalling effects of FA, at least half of the FA responses in ventromedial hypothalamic neurons are mediated by interaction with FAT/CD36, an FA transporter/receptor that does not require intracellular metabolism to activate downstream signalling. Thus, FA or their metabolites can modulate neuronal activity as a means of directly monitoring ongoing fuel availability by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. Recently, the role of lipoprotein lipase in FA sensing has also been shown in animal models not only in hypothalamus, but also in hippocampus and striatum. Finally, FA overload might impair neural control of energy homeostasis through enhanced ceramide synthesis and may contribute to obesity and/or type 2 diabetes pathogenesis in predisposed subjects.


Subject(s)
CD36 Antigens/metabolism , Fatty Acids, Nonesterified/metabolism , Feedback, Physiological , Lipid Metabolism , Models, Neurological , Neurons/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Appetite Regulation , Corpus Striatum/cytology , Corpus Striatum/metabolism , Fatty Acids, Nonesterified/blood , Hippocampus/cytology , Hippocampus/metabolism , Humans , Lipoprotein Lipase/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Organ Specificity , Ventromedial Hypothalamic Nucleus/cytology
4.
Diabetes Metab ; 40(1): 29-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24210646

ABSTRACT

Nutrient-sensitive neurons [to glucose and fatty acids (FAs)] are present at many sites throughout the brain, including the hypothalamus and brain stem, and play a key role in the neural control of energy and glucose homoeostasis. Through their neuronal output, FAs can modulate feeding behaviour as well as insulin secretion and activity. Central administration of oleate, for example, inhibits food intake and glucose production in rats. This suggests that daily variations in plasma FA concentrations could be detected by the central nervous system as a signal that contributes to regulation of energy balance. At the cellular level, subpopulations of neurons in the ventromedial and arcuate hypothalamic nuclei are selectively either inhibited or activated by FAs. Possible molecular effectors of these FA effects most likely include the chloride and potassium ion channels. While intracellular metabolism and activation of the ATP-sensitive K(+) channels appear to be necessary for some signalling effects of FAs, at least half the FA responses in ventromedial hypothalamic neurons are mediated by interaction with fatty acid translocase (FAT)/CD36, an FA transporter/receptor that does not require intracellular metabolism to activate downstream signalling. Thus, FAs and their metabolites can modulate neuronal activity by directly monitoring the ongoing fuel availability for brain nutrient-sensing neurons involved in the regulation of energy and glucose homoeostasis. Besides these physiological effects, FA overload or metabolic dysfunction may also impair neural control of energy homoeostasis and contribute to obesity and/or type 2 diabetes in predisposed subjects.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Fatty Acids/metabolism , Feeding Behavior , Hypothalamus/metabolism , Neurons/metabolism , Obesity/metabolism , Eating , Energy Metabolism , Female , Genetic Predisposition to Disease , Homeostasis , Humans , Hypothalamus/physiology , KATP Channels/metabolism , Lipid Metabolism , Male , Potassium Channels/metabolism , Signal Transduction
5.
Int J Obes (Lond) ; 37(12): 1515-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23478425

ABSTRACT

OBJECTIVE: Obesity is a costly, deadly public health problem for which new treatments are needed. Individual differences in meal pattern have been proposed to have a role in obesity risk. The present study tested the hypothesis that (i) the microstructure of chronic high-fat diet intake differs between genetically selected diet-induced obesity (DIO) and diet-resistant (DR) rats, and (ii) central administration of urocortin 2 (Ucn 2), a corticotropin-releasing factor type 2 agonist, decreases high-fat diet intake not only in lean DR rats, but also in obese DIO rats. DESIGN: Male, selectively bred DIO and DR rats (n=10/genotype) were chronically fed a high-fat diet. Food and water intake as well as ingestion microstructure were then compared under baseline conditions and following third intracerebroventricular injection of Ucn 2 (0, 0.1, 0.3, 1, 3 µg). RESULTS: Irrespective of genotype, Ucn 2 reduced nocturnal food intake with a minimum effective dose of 0.3 µg, suppressing high-fat diet intake by ∼40% at the 3 µg dose. Ucn 2 also made rats of both genotypes eat smaller and briefer meals, including at doses that did not reduce drinking. Obese DIO rats ate fewer but larger meals than DR rats, which they ate more quickly and consumed with two-third less water. CONCLUSIONS: Unlike leptin and insulin, Ucn 2 retains its full central anorectic efficacy to reduce high-fat diet intake even in obese, genetically prone DIO rats, which otherwise show a 'gorging' meal pattern. These results open new opportunities of investigation toward treating some forms of DIO.


Subject(s)
Appetite Depressants/pharmacology , Dietary Fats/administration & dosage , Eating/drug effects , Feeding Behavior/drug effects , Obesity/drug therapy , Urocortins/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Behavior, Animal , Diet, High-Fat , Disease Models, Animal , Drinking/drug effects , Injections, Intraventricular , Male , Obesity/etiology , Obesity/prevention & control , Rats , Rats, Mutant Strains , Time Factors
6.
Diabetes Metab ; 37(2): 83-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21185213

ABSTRACT

Nutrient sensitive neurons (glucose and fatty acids (FA)) are present in many sites throughout the brain, including the hypothalamus and brainstem, and play a key role in the neural control of energy and glucose homeostasis. Through neuronal output, FA may modulate feeding behaviour as well as both insulin secretion and action. For example, central administration of oleate inhibits food intake and glucose production in rats. This suggests that daily variations in plasma FA concentrations might be detected by the central nervous system as a signal which contributes to the regulation of energy balance. At the cellular level, subpopulations of neurons in the ventromedial and arcuate hypothalamic nuclei are selectively either inhibited or activated by FA. Possible molecular effectors of these FA effects likely include chloride or potassium ion channels. While intracellular metabolism and activation of the ATP-sensitive K(+) channel appear to be necessary for some of the signaling effects of FA, at least half of the FA responses in ventromedial hypothalamic neurons are mediated by interaction with FAT/CD36, a FA transporter/receptor that does not require intracellular metabolism to activate downstream signaling. Thus, FA or their metabolites can modulate neuronal activity as a means of directly monitoring ongoing fuel availability by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. Besides these physiological effects, FA overload or metabolic dysfunction might impair neural control of energy homeostasis and contribute to obesity and/or type 2 diabetes in predisposed subjects.


Subject(s)
Brain/physiology , Energy Metabolism/physiology , Fatty Acids/blood , Fatty Acids/metabolism , Homeostasis/physiology , Animals , Biological Transport , Humans , Hypothalamus/cytology , Neurons/physiology , Neuropeptides/physiology , Neurotransmitter Agents/physiology
7.
Int J Obes (Lond) ; 33 Suppl 1: S53-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19363509

ABSTRACT

Epidemiological studies suggest that maternal undernutrition, obesity and diabetes during gestation and lactation can all produce obesity in human offspring. Animal models provide a means of assessing the independent consequences of altering the pre- vs postnatal environments on a variety of metabolic, physiological and neuroendocrine functions, which lead to the development of offspring obesity, diabetes, hypertension and hyperlipidemia. During the gestational period, maternal malnutrition, obesity, type 1 and type 2 diabetes, and psychological and pharmacological stressors can all promote offspring obesity. Normal postnatal nutrition can sometimes reduce the adverse effect of some of these prenatal factors, but may also exacerbate the development of obesity and diabetes in offspring of dams that are malnourished during gestation. The genetic background of the individual is also an important determinant of outcome when the perinatal environment is perturbed. Individuals with an obesity-prone genotype are more likely to be adversely affected by factors such as maternal obesity and high-fat diets. Many perinatal manipulations are associated with reorganization of the central neural pathways which regulate food intake, energy expenditure and storage in ways that enhance the development of obesity and diabetes in offspring. Both leptin and insulin have strong neurotrophic properties so that an excess or an absence of either of them during the perinatal period may underlie some of these adverse developmental changes. As perinatal manipulations can permanently and adversely alter the systems that regulate energy homeostasis, it behooves us to gain a better understanding of the factors during this period that promote the development of offspring obesity as a means of stemming the tide of the emerging worldwide obesity epidemic.


Subject(s)
Disease Models, Animal , Energy Metabolism/physiology , Maternal Nutritional Physiological Phenomena/physiology , Obesity/etiology , Pregnancy Complications , Animals , Body Weight/physiology , Child , Exercise , Female , Humans , Infant, Newborn , Insulin/physiology , Lactation , Leptin/physiology , Mice , Mice, Inbred Strains , Obesity/genetics , Obesity/prevention & control , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Weaning
8.
Int J Obes (Lond) ; 30(4): 722-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16404411

ABSTRACT

OBJECTIVE: To determine the effect of exercise on weight gain and adiposity in obesity-prone and -resistant rats. DESIGN: Body weight gain, fat pad weights, food intake, plasma leptin and insulin levels were assessed in outbred male Sprague-Dawley rats, which remained sedentary or were given unrestricted access to running wheels either before or after they developed diet-induced obesity (DIO) or diet-resistance (DR) on a high energy (HE; 31% fat) diet. RESULTS: When fed a low fat (4.5%) chow diet, rats which would later develop DIO (n=6) after 3 weeks on HE diet ran the same amount as DR rats (n=6). Other rats were first made DIO (n=12) or DR (n=12) after 10 weeks on HE diet and then either kept sedentary or given running wheels for 4 weeks on HE diet. DIO and DR rats ran comparable amounts but only the DIO rats reduced their body weight gain, fat pad relative to body weights and plasma leptin levels significantly, compared to their sedentary controls. Exercise had no effect on food intake in either DIO or DR rats but reduced feed efficiency (weight gain/caloric intake) in both. CONCLUSION: Although DIO and DR rats ran similar amounts, the greater reduction in body weight gain and adiposity of exercising DIO rats suggests that they are more sensitive to some metabolic or physiologic system that prevents them from increasing their intake sufficiently to compensate for their net reduction in energy stores.


Subject(s)
Adiposity/physiology , Obesity/physiopathology , Physical Exertion/physiology , Weight Gain/physiology , Animals , Energy Intake/physiology , Insulin/blood , Leptin/blood , Male , Rats , Rats, Sprague-Dawley
9.
Diabetologia ; 47(11): 2032-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15599702

ABSTRACT

AIMS/HYPOTHESIS: NEFA play a key role in the setting of insulin resistance and hyperinsulinaemia, which are both features of the prediabetic state. In addition to the direct effects on pancreas and peripheral tissues, NEFA have been reported to act via changes in autonomic nervous system activity. The present study was aimed at studying the effects of a local increase in NEFA in the brain on glucose-induced insulin secretion (GIIS) and on insulin action. We hypothesised that cerebral NEFA beta oxidation is a prerequisite for these central effects. METHODS: Male Wistar rats were infused with Intralipid/heparin for 24 h through the carotid artery towards the brain (IL rats), after which we performed the GIIS test, a euglycaemic-hyperinsulinaemic clamp and c-fos immunochemistry. In another series of experiments, Intralipid/heparin infusion was coupled with lateral ventricular infusion of etomoxir, a CPT1 inhibitor, which was initiated 5 days previously. RESULTS: During the infusion period, there were no changes in plasma NEFA, insulin or glucose concentrations. IL rats displayed an increased GIIS compared with control rats (C rats) infused with saline/heparin, and their liver insulin sensitivity was decreased. Furthermore, lipid infusion induced a significant decrease in c-fos-like immunoreactive neurons in medial hypothalamic nuclei, and an increase in lateral hypothalamus. Neuronal activation profile was almost normalised in IL rats infused with etomoxir, and GIIS was strongly decreased, possibly because of the concomitant normalisation of hepatic glucose output. CONCLUSIONS/INTERPRETATION: These results strongly suggest that beta oxidation is required for the central effects of NEFA on GIIS.


Subject(s)
Brain/metabolism , Fatty Acids, Nonesterified/pharmacology , Glucose/pharmacology , Insulin/metabolism , Animals , Body Weight , Brain/drug effects , Energy Intake , Hyperinsulinism/physiopathology , Insulin Resistance , Insulin Secretion , Kinetics , Oxidation-Reduction , Rats
10.
J Clin Exp Neuropsychol ; 26(5): 698-705, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15370391

ABSTRACT

In this case study, we present two Holocaust survivors who appeared to have adapted well post-trauma, but developed severe PTSD symptomatology following the onset of neurologic illness in later life. These individuals were referred fro neuropsychological evaluations by their treating neurologists to assess their levels of cognitive functioning. We present the neuropsychological findings, and discuss possible mechanisms for emergence of PTSD symptoms. These case studies demonstrate the need for systematic research to further investigate the potential relationship between aging, degenerative disease, and PTSD symptoms in elderly trauma survivors.


Subject(s)
Holocaust , Mental Processes/physiology , Nervous System Diseases/etiology , Stress Disorders, Post-Traumatic/complications , Survivors , Age of Onset , Aged , Aged, 80 and over , Female , Humans , Life Change Events , Male , Neuropsychological Tests , Psychiatric Status Rating Scales
11.
Diabetes ; 53(8): 1959-65, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277373

ABSTRACT

Glucosensing neurons in the hypothalamic arcuate nucleus (ARC) were studied using electrophysiological and immunocytochemical techniques in neonatal male Sprague-Dawley rats. We identified glucose-excited and -inhibited neurons, which increase and decrease, respectively, their action potential frequency (APF) as extracellular glucose levels increase throughout the physiological range. Glucose-inhibited neurons were found predominantly in the medial ARC, whereas glucose-excited neurons were found in the lateral ARC. ARC glucose-excited neurons in brain slices dose-dependently increased their APF and decreased their ATP-sensitive K+ channel (KATP channel) currents as extracellular glucose levels increased from 0.1 to 10 mmol/l. However, glucose sensitivity was greatest as extracellular glucose decreased to <2.5 mmol/l. The glucokinase inhibitor alloxan increases KATP single-channel currents in glucose-excited neurons in a manner similar to low glucose. Leptin did not alter the activity of ARC glucose-excited neurons. Although insulin did not affect ARC glucose-excited neurons in the presence of 2.5 mmol/l (steady-state) glucose, they were stimulated by insulin in the presence of 0.1 mmol/l glucose. Neuropeptide Y (NPY) inhibited and alpha-melanocyte-stimulating hormone stimulated ARC glucose-excited neurons. ARC glucose-excited neurons did not show pro-opiomelanocortin immunoreactivity. These data suggest that ARC glucose-excited neurons may serve an integrative role in the regulation of energy balance.


Subject(s)
Alloxan/pharmacology , Arcuate Nucleus of Hypothalamus/physiology , Glucose/pharmacology , Neurons/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , In Vitro Techniques , Male , Membrane Potentials/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Tolbutamide/pharmacology
13.
Diabetes ; 50(12): 2673-81, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11723049

ABSTRACT

Glucosensing neurons in the ventromedial hypothalamic nucleus (VMN) were studied using visually guided slice-patch recording techniques in brain slices from 14- to 21-day-old male Sprague-Dawley rats. Whole-cell current-clamp recordings were made as extracellular glucose levels were increased (from 2.5 to 5 or 10 mmol/l) or decreased (from 2.5 to 0.1 mmol/l). Using these physiological conditions to define glucosensing neurons, two subtypes of VMN glucosensing neurons were directly responsive to alterations in extracellular glucose levels. Another three subtypes were not directly glucose-sensing themselves, but rather were presynaptically modulated by changes in extracellular glucose. Of the VMN neurons, 14% were directly inhibited by decreases in extracellular glucose (glucose-excited [GE]), and 3% were directly excited by decreases in extracellular glucose (glucose-inhibited [GI]). An additional 14% were presynaptically excited by decreased glucose (PED neurons). The other two subtypes of glucosensing neurons were either presynaptically inhibited (PIR; 11%) or excited (PER; 8%) when extracellular glucose was raised to > 2.5 mmol/l. GE neurons sensed decreased glucose via an ATP-sensitive K(+) (K(ATP)) channel. The inhibitory effect of increased glucose on PIR neurons appears to be mediated by a presynaptic gamma-aminobutyric acid-ergic glucosensing neuron that probably originates outside the VMN. Finally, all types of glucosensing neurons were both fewer in number and showed abnormal responses to glucose in a rodent model of diet-induced obesity and type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Neurons/physiology , Obesity/physiopathology , Synapses/physiology , Ventromedial Hypothalamic Nucleus/physiopathology , Adenosine Triphosphate/pharmacology , Animals , Cell Count , Diabetes Mellitus, Type 2/pathology , Diet , Glucose/pharmacology , Male , Neurons/drug effects , Neurons/pathology , Obesity/etiology , Obesity/pathology , Patch-Clamp Techniques , Potassium Channels/physiology , Rats , Rats, Sprague-Dawley , Ventromedial Hypothalamic Nucleus/pathology , gamma-Aminobutyric Acid/physiology
15.
Clin Neuropsychol ; 15(1): 13-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11778575

ABSTRACT

Despite the rapid increase of Hispanics in the U.S., there continues to be a lack of adequate psychological assessment tools to examine Spanish-speaking patients with cognitive or neuropsychological disturbances. We investigated the clinical utility of the Multilingual Aphasia Examination-Spanish (MAE-S) in the evaluation of language functions of Hispanic subjects post-traumatic brain injury (TBI). The performance of 40 TBI patients was compared to that of 40 age-, gender-, and education-matched normal controls. Subject groups differed on the Visual Naming (VN), Controlled Oral Word Association (COWA), and Token Test subtests. The VN and COWA subtests were the best discriminators of group membership. Distribution of scores for the patient group on the Rating of Articulation scale additionally indicate subtle articulatory difficulties post-TBI. For all subtests, trauma severity per Glasgow Coma Scale was the best predictor of language performance, over and above the contribution of other clinical and demographic variables. These results are consistent with prior reports of dysphasia post-TBI and suggest that the MAE-S is a sensitive and accurate measure to assess language disturbances in Hispanic populations.


Subject(s)
Aphasia/diagnosis , Brain Injury, Chronic/diagnosis , Hispanic or Latino/psychology , Multilingualism , Neuropsychological Tests/statistics & numerical data , Adult , Aphasia/psychology , Brain Injury, Chronic/psychology , Female , Humans , Male , Middle Aged , Psychometrics , Reproducibility of Results
16.
Int J Obes Relat Metab Disord ; 25 Suppl 5: S68-72, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11840219

ABSTRACT

The brain regulates energy homeostasis by balancing energy intake, expenditure and storage. To accomplish this, it has evolved specialized neurons that receive and integrate afferent neural and metabolic signals conveying information about the energy status of the body. These sensor-integrator-effector neurons are located in brain areas involved in homeostatic functions such as the hypothalamus, locus coeruleus, basal ganglia, limbic system and nucleus tractus solitarius. The ability to sense and regulate glucose metabolism is critical because of glucose's primacy as a metabolic substrate for neural function. Most neurons use glucose as an energy substrate, but glucosensing neurons also use glucose as a signaling molecule to regulate neuronal firing and transmitter release. There are two types of glucosensing neurons that either increase (glucose responsive, GR) or decrease (glucose sensitive, GS) their firing rate as brain glucose levels rise. Little is known about the mechanism by which GS neurons sense glucose. However, GR neurons appear to function much like the pancreatic beta-cell where glycolysis regulates the activity of an ATP-sensitive K(+) (K(ATP)) channel. The K(ATP) channel is composed of four pore-forming units (Kir6.2) and four sulfonylurea binding sites (SUR). Glucokinase (GK) appears to modulate K(ATP) channel activity via its gatekeeper role in the glycolytic production of ATP. Thus, GK may serve as a marker for GR neurons. Neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) neurons in the hypothalamic arcuate nucleus are critical components of the energy homeostasis pathways in the brain. Both express Kir6.2 and GK, as well as leptin receptors. They also receive visceral neural and intrinsic neuropeptide and transmitter inputs. Such metabolism-related signals can summate upon K(ATP) channel activity which then alters membrane potential, neuronal firing rate and peptide/transmitter release. The outputs of these neurons are integral components of effector systems which regulate energy homeostasis. Thus, arcuate NPY and POMC neurons are probably prototypes of this important class of sensor-integrator-effector neurons.


Subject(s)
Brain/physiology , Glucose/metabolism , Neurons/physiology , Animals , Energy Metabolism , Glucokinase/physiology , Homeostasis , Humans , Potassium Channels/physiology , Signal Transduction
17.
J Clin Exp Neuropsychol ; 22(5): 580-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11094393

ABSTRACT

We evaluated prospectively 210 patients with idiopathic Parkinson's disease (PD) to determine whether cognitive deterioration and disease disability affect subject drop out. Subjects who refused to return for follow-up testing had a greater degree of bradykinesia and overall disability, more advanced disease, fewer years of education and greater depressive symptomatology. However, discriminant analysis indicated that performance on the neuropsychological measures, rather than PD severity, significantly predicted whether patients return for follow-up testing. Our findings indicate that cognitive impairment uniquely contributes to subject attrition, which may distort dementia estimates in PD.


Subject(s)
Cognition , Dementia/etiology , Parkinson Disease/complications , Parkinson Disease/psychology , Patient Dropouts/psychology , Aged , Confounding Factors, Epidemiologic , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neuropsychological Tests , Parkinson Disease/physiopathology , Patient Selection , Prognosis , Prospective Studies , Severity of Illness Index
18.
Am J Physiol Regul Integr Comp Physiol ; 279(6): R2222-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080089

ABSTRACT

Chronic administration of sibutramine lowers body weight, presumably by altering brain monoamine metabolism. Here the effect of sibutramine on sympathoadrenal function (24-h urine norepinephrine and epinephrine levels) and arcuate nucleus (ARC) neuropeptide Y (NPY) and proopiomelanocortin (POMC) expression was assessed in diet-induced obese rats fed a low-fat diet. Chronic (10 wk) sibutramine [5 mg. kg(-1). day(-1) ip; rats fed ad libitum and injected with sibutramine (AS)] lowered body weight by 15% but only transiently (3-4 wk) reduced intake compared with vehicle-treated controls [rats fed chow ad libitum and injected with vehicle daily (AV)]. Other rats food restricted (RS) to 90% of the weight of AS rats and then given sibutramine restored their body weights to the level of AS rats when allowed libitum food intake. After reequilibration, RS rats were again energy restricted to reduce their weight to 90% of AS rats, and additional vehicle-treated rats (RV) were restricted to keep their body weights at the level of AS rats for 3 wk more. Terminally, total adipose depot weights and leptin levels paralleled body weights (AV > AS = RV > RS), although AS rats had heavier abdominal and lighter peripheral depots than RV rats of comparable body weights. Sibutramine treatment increased sympathetic activity, attenuated the increased ARC NPY, and decreased POMC mRNA levels induced by energy restriction in RV rats. Thus sibutramine lowered the defended body weight in association with compensatory changes in those central pathways involved in energy homeostasis.


Subject(s)
Appetite Depressants/pharmacology , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/physiology , Cyclobutanes/pharmacology , Feeding Behavior , Obesity/physiopathology , Adipose Tissue/anatomy & histology , Adipose Tissue/drug effects , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Body Weight/drug effects , Energy Intake , Epinephrine/urine , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Homeostasis/physiology , Leptin/blood , Neuropeptide Y/genetics , Norepinephrine/urine , Organ Size/drug effects , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , Rats
19.
Nutrition ; 16(10): 909-15, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11054596

ABSTRACT

There is an obesity epidemic in the industrialized world that is not simply explained by excess energy intake and decreased energy expenditure. Persistent obesity develops when genetically predisposed individuals are in a chronic state of positive energy balance. Once established, the obese body weight is avidly defended against both over- and underfeeding. Animal studies have shown that lean individuals who are genetically predisposed toward obesity have abnormalities of neural function that prime them to become obese when caloric density of the diet is raised. These neural abnormalities are gradually "corrected" as obesity becomes fully developed, suggesting that obesity is the normal state for such individuals. Thus, defense of the obese body weight may be perpetuated by the formation of new neural circuits involved in energy-homeostasis pathways that are not then easily abolished. Such neural plasticity can occur in both adult life and during nervous-system development. Early pre- and postnatal metabolic conditions (maternal diabetes, obesity, undernutrition) can lead genetically predisposed offspring to become even more obese as adults. This enhanced obesity is associated with altered brain neural circuitry, and these changes can then be passed on to subsequent generations in a feed-forward cycle of ever-increasing body weight. Thus, the metabolic perturbations associated with obesity during both brain development and adult life can produce "metabolic imprinting" on genetically predisposed neural circuits involved in energy homeostasis. Drugs that reduce body weight decrease the defended body weight and alter neural pathways involved in energy homeostasis but have no permanent effect on body weight or neural function in most individuals. Thus, early intervention in mothers, infants, children, and adults may be the only way to prevent the formation of permanent neural connections that promote and perpetuate obesity in genetically predisposed individuals.


Subject(s)
Diet , Energy Metabolism , Neural Pathways/physiology , Neuronal Plasticity/physiology , Obesity/physiopathology , Animals , Environment , Genetic Predisposition to Disease , Humans , Neuronal Plasticity/genetics , Obesity/genetics
20.
Am J Physiol Regul Integr Comp Physiol ; 279(4): R1357-64, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11004005

ABSTRACT

The relationship between stress and obesity was assessed in male rats selectively bred to develop either diet-induced obesity (DIO) or diet resistance (DR) when fed a high-energy, 31% fat diet for 3 wk followed by 2 wk on a hyperphagic liquid diet (Ensure). One-half of the rats of each phenotype were subjected to moderate daily, unpredictable stress (cage changing, exposure to conspecific, swim, and immobilization stress, intraperitoneal saline injection) during the 5 wk. Both stressed and unstressed DIO rats were 26% heavier and ate 27% more than comparable DR rats at onset and had 48% lower basal morning plasma corticosterone levels. Stressed DR rats gained less weight and had significant elevations of basal morning corticosterone but reduced basal sympathetic activity (24-h urine norepinephrine) over 5 wk compared with their unstressed DR controls. Terminally, there was a 35% increase in the paraventricular nucleus corticotropin-releasing hormone mRNA expression. On the other hand, stressed DIO rats showed only a transient early increase in open-field activity and a terminal increase in basal corticosterone levels as the only effects of stress. Thus DIO rats are hyporesponsive to chronic stress compared with DR rats. This is in keeping with several other known differences in hypothalamopituitary and autonomic function in this model.


Subject(s)
Diet , Feeding Behavior , Obesity/physiopathology , Obesity/psychology , Stress, Psychological/physiopathology , Weight Gain/physiology , Animals , Body Weight , Energy Intake , Energy Metabolism , Genetic Predisposition to Disease , Male , Obesity/genetics , Rats , Rats, Sprague-Dawley , Restraint, Physical , Sodium Chloride/administration & dosage , Sodium Chloride/pharmacology , Swimming , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...