Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alison M. Strack is active.

Publication


Featured researches published by Alison M. Strack.


Frontiers in Neuroendocrinology | 1993

Feast and Famine: Critical Role of Glucocorticoids with Insulin in Daily Energy Flow

Mary F. Dallman; Alison M. Strack; Susan F. Akana; Margaret J. Bradbury; Edward S. Hanson; Karen A. Scribner; Michael Smith

The hypothesis proposed in this review is that normal diurnal rhythms in the hypothalamic-pituitary-adrenal (HPA) axis are highly regulated by activity in medial hypothalamic nuclei to effect an interaction between corticosteroids and insulin such that optimal metabolism results in response to changes in the fed or fasted state of the animal. There are marked diurnal rhythms in function of the HPA axis under both basal and stress conditions. The HPA axis controls corticosteroid output from the adrenal and, in turn, forward elements of this axis are inhibited by feedback from circulating plasma corticosteroid levels. Basal activity in the HPA axis of mammals fed ad lib peaks about 2 h before the peak of the diurnal feeding rhythm, and is controlled by input from the suprachiasmatic nuclei. The rhythm in stress responsiveness is lowest at the time of the basal peak and highest at the time of the basal trough in the HPA axis activity. There are also diurnal rhythms in corticosteroid feedback sensitivity of basal and stress-induced ACTH secretion which peak at the time of the basal trough. These rhythms are all overridden when feeding, and thus insulin secretion, is disrupted. Corticosteroids interact with insulin on food intake and body composition, and corticosteroids also increase insulin secretion. Corticosteroids stimulate feeding at low doses but inhibit it at high doses; however, it is the high levels of insulin, induced by high levels of corticosteroids, that may inhibit feeding. The effects of corticosteroids on liver, fat, and muscle cell metabolism, with emphasis on their interactions with insulin, are briefly reviewed. Corticosteroids both synergize with and antagonize the effects of insulin. The effects of stress hormones, and their interactions with insulin on lipid and protein metabolism, followed by some of the metabolic effects of injury stress, with or without nutritional support, are evaluated. In the presence of elevated insulin stimulated by glucocorticoids and nutrition, stress causes less severe catabolic effects. In the central nervous system, regulation of function in the HPA axis is clearly affected by the activity of medial hypothalamic nuclei that also alter feeding, metabolism, and obesity in rats. Lesions of the arcuate (ARC) and ventromedial (VMN) paraventricular (PVN) nuclei result in obesity and hyperactivity in the HPA axis. Moreover, adrenalectomy inhibits or prevents development of the lesion-induced obesity. There are interactions among these nuclei; one mode of communication is via inputs of neuropeptide Y (NPY) cells in the ARC to the VMN, dorsomedial nuclei, and PVN.(ABSTRACT TRUNCATED AT 400 WORDS)


Annals of the New York Academy of Sciences | 1995

The Neural Network that Regulates Energy Balance Is Responsive to Glucocorticoids and Insulin and Also Regulates HPA Axis Responsivity at a Site Proximal to CRF Neurons

Mary F. Dallman; Susan F. Akana; Alison M. Strack; E. Simon Hanson; Raynard J. Sebastian

The structure of a large neural system that responds to and regulates energy balance and that encompasses that PVN and activity of the HPA axis has begun to emerge from these experiments (Fig. 6). Several large loops have been delineated within this context of the maintenance of energy balance. Corticosteroids stimulate both feeding and insulin secretion. The actions of corticosteroids in the periphery are catabolic, causing mobilization of energy stores; their actions in the central nervous system are stimulatory to energy acquisition (food intake). By contrast, the action of insulin in the periphery is anabolic, causing energy storage; its action in the central nervous system is inhibitory to energy acquisition (food intake). At the level of the CNS, insulin inhibits and corticosteroids stimulate expression of NPY mRNA in the arcuate nuclei, and these actions may explain, in part, the reciprocal actions of the hormones on energy acquisition. Thus over the long term, stimulation of insulin secretion by corticosteroids tends to supply an automatic brake on the effects of corticosteroids on feeding. The neural system that controls energy balance and responds to the reciprocal signals of corticosterone and insulin also regulates responsivity to restraint stress in the HPA axis. The low-amplitude ACTH responses to restraint, corticosteroid feedback, and prior stress-induced facilitation that are observed under conditions of relative fasting in the PM can be produced in the AM by a 14-h, overnight fast. By contrast, NPY injected ivt stimulates identical ACTH responses in the AM in fed rats and in rats fasted overnight, suggesting that NPY acts to stimulate CRF secretion at a site closer to the PVN than the stress of restraint, which is filtered through the neural energy balance system. In the periphery, corticosteroids and insulin also have reciprocal effects on energy storage; effects that are opposite those exerted in the CNS on energy acquisition. Thus, together, the two hormones may be construed as a bihormonal system that regulates overall energy balance. Although under normal conditions this system is well designed to accomplish energy balance, and provides a mechanism by which total energy stores may be increased appropriately (e.g., prior to hibernation or migration), it seems probable that under conditions of chronic stress, this regulatory system may be maladaptive. Chronic stress and glucocorticoid treatment cause increases in mean daily concentrations of both corticosteroids and insulin. Increases in the absolute levels of both hormones, with the normal ratio between them maintained, results in remodeling of body energy stores-away from muscle stores and toward fat stores, particularly abdominal fat stores. It seems quite likely that some conditions of abdominal obesity in man may be explained, at least in part, by increased activity in the HPA axis. Because abdominal obesity is associated with cardiovascular diseases, these responses, when they persist, are clearly maladaptive. Exploration of the role and control of the HPA axis in and by the larger neural network that regulates energy balance has to date been instructive. Clearly this work has just begun and is primarily still at the level of phenomenology. However, once the phenomenology is understood, mechanistic work can be performed that will flesh out our understanding of this very large and physiologically essential system.


Endocrinology | 1999

Starvation: Early Signals, Sensors, and Sequelae*

Mary F. Dallman; Susan F. Akana; Seema Bhatnagar; M. Elizabeth Bell; SuJean Choi; Alan Chu; Cydney Horsley; Nancy Levin; Onno C. Meijer; L. Soriano; Alison M. Strack; Victor Viau

To identify the sequences of changes in putative signals, reception of these and responses to starvation, we sampled fed and starved rats at 2- to 6-h intervals after removal of food 2 h before dark. Metabolites, hormones, hypothalamic neuropeptide expression, fat depots, and leptin expression were measured. At 2 h, insulin decreased, and FFA and corticosterone (B) increased; by 4 h, leptin and glucose levels decreased. Neuropeptide Y messenger RNA (mRNA) increased 6 h after food removal and thereafter. Adrenal and plasma B did not follow ACTH and were elevated throughout, with a nadir at the dark-light transition. Leptin correlated inversely with adrenal B. Fat stores decreased during the last 12 h. Leptin mRNA in perirenal and sc fat peaked during the dark period, resembling plasma leptin in fed rats. We conclude that 1) within the first 4 h, hormonal and metabolic signals relay starvation-induced information to the hypothalamus; 2) hypothalamic neuropeptide synthesis responds rapidly to the altered metabolic signals; 3) catabolic activity quickly predominates, reinforced by elevated B, not driven by ACTH, but possibly to a minor extent by leptin, and more by adrenal neural activity; and 4) leptin secretion decreases before leptin mRNA or fat depot weight, showing synthesis-independent regulation. (Endocrinology 140: 4015‐ 4023, 1999)


Annals of the New York Academy of Sciences | 2004

Chronic Stress-Induced Effects of Corticosterone on Brain: Direct and Indirect

Mary F. Dallman; Susan F. Akana; Alison M. Strack; K S Scribner; Norman C. Pecoraro; S. E. La Fleur; Hani Houshyar; Francisca Gomez

Abstract: Acutely, glucocorticoids act to inhibit stress‐induced corticotrophin‐releasing factor (CRF) and adrenocorticotrophic hormone (ACTH) secretion through their actions in brain and anterior pituitary (canonical feedback). With chronic stress, glucocorticoid feedback inhibition of ACTH secretion changes markedly. Chronically stressed rats characteristically exhibit facilitated ACTH responses to acute, novel stressors. Moreover, in adrenalectomized rats in which corticosterone was replaced, steroid concentrations in the higher range are required for facilitation of ACTH responses to occur after chronic stress or diabetes. Infusion of corticosterone intracerebroventricularly into adrenalectomized rats increases basal ACTH, tends to increase CRF, and allows facilitation of ACTH responses to repeated restraint. Therefore, with chronic stressors, corticosterone seems to act in brain in an excitatory rather than an inhibitory fashion. We believe, under conditions of chronic stress, that there is an indirect glucocorticoid feedback that is mediated through the effects of the steroid ± insulin on metabolism. Increased energy stores feedback on brain to inhibit hypothalamic CRF and decrease the expression of dopamine‐β‐hydroxylase in the locus coeruleus. These changes would be expected to decrease the level of discomfort and anxiety induced by chronic stress. Moreover, central neural actions of glucocorticoids abet the peripheral effects of the steroids by increasing the salience and ingestion of pleasurable foods.


Journal of Pharmacology and Experimental Therapeutics | 2007

Antiobesity Efficacy of a Novel Cannabinoid-1 Receptor Inverse Agonist, N-[(1S,2S)-3-(4-Chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl]-2-methyl-2-{[5-(trifluoromethyl)pyridin-2-yl]oxy}propanamide (MK-0364), in Rodents

Tung M. Fong; Xiao-Ming Guan; Donald J. Marsh; Chun-Pyn Shen; D. Sloan Stribling; Kim Rosko; Julie Lao; Hong Yu; Yue Feng; Jing C. Xiao; Lex H.T. Van der Ploeg; Mark T. Goulet; Williams K. Hagmann; Linus S. Lin; Thomas J. Lanza; James P. Jewell; Ping Liu; Shrenik K. Shah; Hongbo Qi; Xinchun Tong; Junying Wang; Suoyu S. Xu; Barbara Francis; Alison M. Strack; D. Euan MacIntyre; Lauren P. Shearman

The cannabinoid-1 receptor (CB1R) has been implicated in the control of energy balance. To explore the pharmacological utility of CB1R inhibition for the treatment of obesity, we evaluated the efficacy of N-[(1S,2S)-3-(4-chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl]-2-methyl-2-{[5-(trifluoromethyl)pyridin-2-yl]oxy}propanamide (MK-0364) and determined the relationship between efficacy and brain CB1R occupancy in rodents. MK-0364 was shown to be a highly potent CB1R inverse agonist that inhibited the binding and functional activity of various agonists with a binding Ki of 0.13 nM for the human CB1R in vitro. MK-0364 dose-dependently inhibited food intake and weight gain, with an acute minimum effective dose of 1 mg/kg in diet-induced obese (DIO) rats. CB1R mechanism-based effect was demonstrated for MK-0364 by its lack of efficacy in CB1R-deficient mice. Chronic treatment of DIO rats with MK-0364 dose-dependently led to significant weight loss with a minimum effective dose of 0.3 mg/kg (p.o.), or a plasma Cmax of 87 nM. Weight loss was accompanied by the loss of fat mass. Partial occupancy (30–40%) of brain CB1R by MK-0364 was sufficient to reduce body weight. The magnitude of weight loss was correlated with brain CB1R occupancy. The partial receptor occupancy requirement for efficacy was also consistent with the reduced food intake of the heterozygous mice carrying one disrupted allele of CB1R gene compared with the wild-type mice. These studies demonstrated that MK-0364 is a highly potent and selective CB1R inverse agonist and that it is orally active in rodent models of obesity.


Journal of Pharmacology and Experimental Therapeutics | 2007

Anti-obesity efficacy of a novel cannabinoid-1 receptor inverse agonist MK-0364 in rodents

Tung M. Fong; Xiao-Ming Guan; Donald J. Marsh; Chun-Pyn Shen; D. Sloan Stribling; Kim Rosko; Julie Z. Lao; Hong Yu; Yue Feng; Jing C. Xiao; Lex H.T. Van der Ploeg; Mark T. Goulet; Williams K. Hagmann; Linus S. Lin; Thomas J. Lanza; James P. Jewell; Ping Liu; Shrenik K. Shah; Hongbo Qi; Xinchun Tong; Junying Wang; Suoyu S. Xu; Barbara Francis; Alison M. Strack; D. Euan MacIntyre; Lauren P. Shearman

The cannabinoid-1 receptor (CB1R) has been implicated in the control of energy balance. To explore the pharmacological utility of CB1R inhibition for the treatment of obesity, we evaluated the efficacy of N-[(1S,2S)-3-(4-chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl]-2-methyl-2-{[5-(trifluoromethyl)pyridin-2-yl]oxy}propanamide (MK-0364) and determined the relationship between efficacy and brain CB1R occupancy in rodents. MK-0364 was shown to be a highly potent CB1R inverse agonist that inhibited the binding and functional activity of various agonists with a binding Ki of 0.13 nM for the human CB1R in vitro. MK-0364 dose-dependently inhibited food intake and weight gain, with an acute minimum effective dose of 1 mg/kg in diet-induced obese (DIO) rats. CB1R mechanism-based effect was demonstrated for MK-0364 by its lack of efficacy in CB1R-deficient mice. Chronic treatment of DIO rats with MK-0364 dose-dependently led to significant weight loss with a minimum effective dose of 0.3 mg/kg (p.o.), or a plasma Cmax of 87 nM. Weight loss was accompanied by the loss of fat mass. Partial occupancy (30–40%) of brain CB1R by MK-0364 was sufficient to reduce body weight. The magnitude of weight loss was correlated with brain CB1R occupancy. The partial receptor occupancy requirement for efficacy was also consistent with the reduced food intake of the heterozygous mice carrying one disrupted allele of CB1R gene compared with the wild-type mice. These studies demonstrated that MK-0364 is a highly potent and selective CB1R inverse agonist and that it is orally active in rodent models of obesity.


Regulatory Peptides | 1997

Evidence that elevated plasma corticosterone levels are the cause of reduced hypothalamic corticotrophin-releasing hormone gene expression in diabetes

Michael W. Schwartz; Alison M. Strack; Mary F. Dallman

Uncontrolled diabetes mellitus causes both a sustained activation of the hypothalamic-pituitary-adrenal (HPA) axis and reduced expression of corticotrophin-releasing hormone (CRH) mRNA in the hypothalamic paraventricular nucleus (PVN). To investigate the role of glucocorticoids in the regulation of CRH mRNA expression in the PVN of diabetic rats, we studied surgically adrenalectomized (ADX) and sham-operated male Sprague-Dawley rats 4 days after i.v. injection of streptozotocin (STZ; 65 mg/kg i.v.) or vehicle. Among sham-operated animals, AM plasma corticosterone levels were significantly increased in diabetic as compared to nondiabetic animals (1.46+/-0.54 vs. 0.22+/-0.05 microg/dl; P <0.05), and were positively correlated to both plasma ACTH levels (r = 0.74; P = 0.015) and adrenal gland weight (r = 0.70; P = 0.025). In contrast, CRH mRNA levels measured in the PVN by in situ hybridization were inversely related to the plasma corticosterone level (r = -0.68; P = 0.045). In a second experiment, both diabetic and nondiabetic ADX rats received a continuous subcutaneous infusion of either corticosterone at one of two doses or its vehicle for 4 days. Among vehicle-treated ADX animals, STZ diabetes raised hypothalamic CRH mRNA levels, in contrast to the tendency for diabetes to lower CRH mRNA in intact rats in the first experiment. Corticosterone administration lowered CRH mRNA comparably in both diabetic and nondiabetic ADX rats. In contrast, diabetes reduced arginine vasopressin (AVP) mRNA levels in the PVN of ADX rats and blunted the inhibitory effect of glucocorticoids on AVP mRNA levels in this setting. We conclude (1) glucocorticoids are necessary for the effect of diabetes to reduce hypothalamic CRH gene expression, since diabetes causes a paradoxical increase in CRH mRNA levels in adrenalectomized animals; (2) glucocorticoid inhibition of hypothalamic CRH gene expression is intact in diabetic rats; and (3) the activation of the HPA axis by diabetes is associated with a proportionate decrease in PVN CRH gene expression. These findings support a model in which hypothalamic factors additional to CRH activate the HPA axis in uncontrolled diabetes, and inhibit CRH gene expression indirectly by negative glucocorticoid feedback.


Neuroendocrinology | 1993

Lesions of the Hippocampal Efferent Pathway (Fimbria-Fornix) Do Not Alter Sensitivity of Adrenocorticotropin to Feedback Inhibition by Corticosterone in Rats

Margaret J. Bradbury; Alison M. Strack; Mary F. Dallman

The hypothalamic-pituitary-adrenal (HPA) axis controls the diurnal and stress-induced release of adrenal corticosteroids into the general blood circulation. In turn, corticosteroids inhibit the HPA axis under basal conditions and during stress through occupation of their receptors (types I and II) in the brain by closing a negative feedback loop. The primary site in the brain at which corticosteroids act to inhibit the HPA axis has not been identified. High concentrations of both types of receptors are found in neurons of the hippocampal formation, a structure which has been reported by some, but not others, to control activity within the HPA axis by serving as a major negative feedback site. In many of these past studies, blood was collected after extensive handling or exposure to ether, conditions which do not favor the detection of basal hormone concentrations. To address these controversies, we tested the feedback sensitivity of the anterior pituitary hormone responsible for corticosteroid production, adrenocorticotropin (ACTH), to corticosterone (B), the main corticosteroid in rats, in total fornix- and, as controls, cortex-lesioned rats. All rats were given vascular catheters to avoid any handling-induced differences in plasma B or ACTH when sampling blood. In some experiments, fornix- and cortex-lesioned rats were adrenalectomized and given 1 of 3 doses of exogenous B provided in a subcutaneous pellet to ensure that plasma B was equal in different lesion groups. We hypothesized that if the hippocampal formation were an important site of B-mediated inhibition of the HPA axis, fornix-lesioned rats would have higher plasma B as a result of increased endogenous secretion in the morning or the evening compared to cortex-lesioned rats in rats with adrenal glands. In addition, we hypothesized that adrenalectomized fornix-lesioned rats given the same low to moderate levels of exogenous constant B would have higher basal and stress-induced ACTH than cortex-lesioned rats. Diurnal plasma B was not affected by fornix lesions in intact rats. Moreover, basal ACTH measured in the morning and the evening and stress-induced ACTH was the same in adrenalectomized fornix- and cortex-lesioned rats with constant exogenous B. We conclude, therefore, that information about occupancy of B receptors in the hippocampus carried by the fornix primarily subserves functions which do not directly regulate activity in the HPA axis.


Stress | 1996

Clamped Corticosterone (B) Reveals the Effect of Endogenous B on Both Facilitated Responsivity to Acute Restraint and Metabolic Responses to Chronic Stress

Susan F. Akana; E S Hanson; Cydney Horsley; Alison M. Strack; Seema Bhatnagar; Margaret J. Bradbury; Milligan Ed; Mary F. Dallman

To determine the effects of both corticosterone (B) and chronic stressors on acute ACTH responses to restraint, young male rats were exposed to streptozotocin-induced diabetes, cold (5-7 degreesC) or intracerebroventricular (icv) neuropeptide Y (NPY) for 5 d and then exposed to restraint within 2 h after lights on. Two groups of rats were studied: intact and adrenalectomized replaced with B pellets that maintained plasma B in the normal mean 24-h range of intact rats. In addition to ACTH and B responses to restraint on d 5, body weight, food intake, fat depots, glucose and other hormones were measured to determine the role of stress-induced elevations in B on energy balance. ACTH responses to restraint were normal in intact rats subjected to diabetes or cold. By contrast, there was no ACTH or B response to restraint in NPY-infused intact rats. All 3 groups of chronically stimulated adrenalectomized rats with clamped B had facilitated ACTH responses to restraint compared to their treatment controls. Overall food intake increased in all groups of stressed rats; however, augmented intake occurred only during the light in intact rats and equally in the light and dark in B-clamped rats. White adipose depot weights were decreased by both diabetes and cold and increased by NPY in intact rats; the decreases with cold and increases with NPY were both blunted and changes in fat stores were not significant in adrenalectomized, B-clamped rats. We conclude that: 1. diabetes- and cold-induced facilitation of restraint-induced afferent input to hypothalamic control of the hypothalamo-pituitary-adrenal (HPA) axis is opposed in intact rats by the elevated feedback signal of B secretion; 2. NPY does not induce facilitation of afferent stress pathways; 3. chronic stimulation of the HPA axis induces acute hyperresponsiveness of hypothalamic neurons to restraint provided that the afferent input of this acute stimulus is not prevented by B feedback; 4. stimulus-induced elevations in B secretion result in day-time feeding; 5. insensitivity of both caloric efficiency and white fat stores to chronic stress in adrenalectomized, B-clamped rats results from loss of normally variable B levels.


The Journal of Comparative Neurology | 1997

Central amygdala Fos expression during hypotensive or febrile, nonhypotensive endotoxemia in conscious rats

Nancy C. Tkacs; Jianhua Li; Alison M. Strack

The distribution and time course of Fos expression in neurons in the central nucleus of the amygdala (CeA) were studied in endotoxemic rats in two separate experiments. In each case, the severity of the endotoxin (lipopolysaccharide; LPS) challenge was characterized by using physiological outcome variables, including blood pressure and heart rate. Throughout the rostrocaudal extent of the CeA, extensive Fos staining was found 3 hours after injection with a hypotensive dose of Salmonella enteritidis LPS. Hypotension alone has been reported to induce Fos in the CeA; therefore, the remaining experiments were performed by using a lower dose of Escherichia coli LPS that did not cause hypotension. The nonhypotensive dose of E. coli LPS also induced Fos in large numbers of neurons of the CeA, with peak staining at 2 hours and Fos staining persisting for 6 hours after LPS injection. Tachycardia and fever after LPS also persisted for at least 6 hours. CeA Fos staining during nonhypotensive endotoxemia was predominantly located in the lateral subnucleus, although Fos‐stained medial subnucleus neurons were also present. Additional forebrain regions that showed persistent Fos staining 6 hours after LPS included the parvocellular paraventricular nucleus of the hypothalamus, the bed nucleus of the stria terminalis, and the medial preoptic area. Forebrain regions that contained Fos‐stained nuclei at earlier time points, but not at 6 hours, included the supraoptic nucleus, magnocellular regions of the paraventricular nucleus of the hypothalamus, the subfornical organ, and the organum vasculosum of the lamina terminalis. Few CeA neurons showed Fos staining in rats that were restrained in a ventilated plastic tube. Neurons in the lateral septal nucleus and the medial amygdaloid nucleus, which have numerous Fos‐stained nuclei after stressors such as footshock or restraint, did not show Fos staining above control levels after LPS administration. Activation of CeA neurons after intravenous LPS may indicate persistent drive from vagal afferents and may implicate the CeA in the autonomic, neuroendocrine, and/or behavioral responses to this treatment. J. Comp. Neurol. 379:592–602, 1997.

Collaboration


Dive into the Alison M. Strack's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susan F. Akana

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

E S Hanson

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cydney Horsley

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge