Network


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

Hotspot


Dive into the research topics where Gregory J. Morton is active.

Publication


Featured researches published by Gregory J. Morton.


Nature | 2006

Central nervous system control of food intake and body weight

Gregory J. Morton; David E. Cummings; Denis G. Baskin; Gregory S. Barsh; Michael J. Schwartz

The capacity to adjust food intake in response to changing energy requirements is essential for survival. Recent progress has provided an insight into the molecular, cellular and behavioural mechanisms that link changes of body fat stores to adaptive adjustments of feeding behaviour. The physiological importance of this homeostatic control system is highlighted by the severe obesity that results from dysfunction of any of several of its key components. This new information provides a biological context within which to consider the global obesity epidemic and identifies numerous potential avenues for therapeutic intervention and future research.


Journal of Clinical Investigation | 2012

Obesity is associated with hypothalamic injury in rodents and humans

Joshua P. Thaler; Chun Xia Yi; Ellen A. Schur; Stephan J. Guyenet; Bang H. Hwang; Marcelo O. Dietrich; Xiaolin Zhao; David A. Sarruf; Vitaly Izgur; Kenneth R. Maravilla; Hong T. Nguyen; Jonathan D. Fischer; Miles E. Matsen; Brent E. Wisse; Gregory J. Morton; Tamas L. Horvath; Denis G. Baskin; Matthias H. Tschöp; Michael W. Schwartz

Rodent models of obesity induced by consuming high-fat diet (HFD) are characterized by inflammation both in peripheral tissues and in hypothalamic areas critical for energy homeostasis. Here we report that unlike inflammation in peripheral tissues, which develops as a consequence of obesity, hypothalamic inflammatory signaling was evident in both rats and mice within 1 to 3 days of HFD onset, prior to substantial weight gain. Furthermore, both reactive gliosis and markers suggestive of neuron injury were evident in the hypothalamic arcuate nucleus of rats and mice within the first week of HFD feeding. Although these responses temporarily subsided, suggesting that neuroprotective mechanisms may initially limit the damage, with continued HFD feeding, inflammation and gliosis returned permanently to the mediobasal hypothalamus. Consistent with these data in rodents, we found evidence of increased gliosis in the mediobasal hypothalamus of obese humans, as assessed by MRI. These findings collectively suggest that, in both humans and rodent models, obesity is associated with neuronal injury in a brain area crucial for body weight control.


Nature | 2001

Intracellular signalling: Key enzyme in leptin-induced anorexia

Kevin D. Niswender; Gregory J. Morton; Walter H. Stearns; Christopher J. Rhodes; Martin G. Myers; Michael W. Schwartz

Leptin is a key hormonal regulator of energy balance that acts upon hypothalamic neurons to reduce food intake, but the intracellular mechanisms involved are incompletely understood. Here we show that systemic administration of leptin in rats activates the enzyme phosphatidylinositol-3-OH kinase (PI(3)K) in the hypothalamus and that intracerebro-ventricular (i.c.v.) infusion of inhibitors of this enzyme prevents leptin-induced anorexia. Our results indicate that PI(3)K is a crucial enzyme in the signal-transduction pathway that links hypothalamic leptin to reduced food intake.


American Journal of Physiology-endocrinology and Metabolism | 2009

Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet

Kelly A. Posey; Deborah J. Clegg; Richard L. Printz; Jaeman Byun; Gregory J. Morton; Anuradha Vivekanandan-Giri; Subramaniam Pennathur; Denis G. Baskin; Jay W. Heinecke; Stephen C. Woods; Michael W. Schwartz; Kevin D. Niswender

Weight gain induced by an energy-dense diet is hypothesized to arise in part from defects in the neuronal response to circulating adiposity negative feedback signals, such as insulin. Peripheral tissue insulin resistance involves cellular inflammatory responses thought to be invoked by excess lipid. Therefore, we sought to determine whether similar signaling pathways are activated in the brain of rats fed a high-fat (HF) diet. The ability of intracerebroventricular (icv) insulin to reduce food intake and activate hypothalamic signal transduction is attenuated in HF-fed compared with low-fat (LF)-fed rats. This effect was accompanied by both hypothalamic accumulation of palmitoyl- and stearoyl-CoA and activation of a marker of inflammatory signaling, inhibitor of kappaB kinase-beta (IKKbeta). Hypothalamic insulin resistance and inflammation were observed with icv palmitate infusion or HF feeding independent of excess caloric intake. Last, we observed that central IKKbeta inhibition reduced food intake and was associated with increased hypothalamic insulin sensitivity in rats fed a HF but not a LF diet. These data collectively support a model of diet-induced obesity whereby dietary fat, not excess calories, induces hypothalamic insulin resistance by increasing the content of saturated acyl-CoA species and activating local inflammatory signals, which result in a failure to appropriately regulate food intake.


Disease Models & Mechanisms | 2010

Standard operating procedures for describing and performing metabolic tests of glucose homeostasis in mice

Julio E. Ayala; Varman T. Samuel; Gregory J. Morton; Silvana Obici; Colleen M. Croniger; Gerald I. Shulman; David H. Wasserman; Owen P. McGuinness

The Mouse Metabolic Phenotyping Center (MMPC) Consortium was established to address the need to characterize the growing number of mouse models of metabolic diseases, particularly diabetes and obesity. A goal of the MMPC Consortium is to propose standard methods for assessing metabolic phenotypes in mice. In this article, we discuss issues pertaining to the design and performance of various tests of glucose metabolism. We also propose guidelines for the description of methods, presentation of data and interpretation of results. The recommendations presented in this article are based on the experience of the MMPC Consortium and other investigators.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2008

Vascular Inflammation, Insulin Resistance, and Reduced Nitric Oxide Production Precede the Onset of Peripheral Insulin Resistance

Francis Kim; Matilda Pham; Ezekiel Maloney; Norma O. Rizzo; Gregory J. Morton; Brent E. Wisse; Elizabeth A. Kirk; Alan Chait; Michael W. Schwartz

Objectives—Obesity causes inflammation and insulin resistance in the vasculature as well as in tissues involved in glucose metabolism such as liver, muscle, and adipose tissue. To investigate the relative susceptibility of vascular tissue to these effects, we determined the time course over which inflammation and insulin resistance develops in various tissues of mice with diet-induced obesity (DIO) and compared these tissue-based responses to changes in circulating inflammatory markers. Methods and Results—Adult male C57BL/6 mice were fed either a control low-fat diet (LF; 10% saturated fat) or a high-fat diet (HF, 60% saturated fat) for durations ranging between 1 to 14 weeks. Cellular inflammation and insulin resistance were assessed by measuring phospho-I&kgr;Bα and insulin-induced phosphorylation of Akt, respectively, in extracts of thoracic aorta, liver, skeletal muscle, and visceral fat. As expected, HF feeding induced rapid increases of body weight, fat mass, and fasting insulin levels compared to controls, each of which achieved statistical significance within 4 weeks. Whereas plasma markers of inflammation became elevated relatively late in the course of DIO (eg, serum amyloid A [SAA], by Week 14), levels of phospho-I&kgr;Bα in aortic lysates were elevated by 2-fold within the first week. The early onset of vascular inflammation was accompanied by biochemical evidence of both endothelial dysfunction (reduced nitric oxide production; induction of intracellular adhesion molecule-1 and vascular cell adhesion molecule-1) and insulin resistance (impaired insulin-induced phosphorylation of Akt and eNOS). Although inflammation and insulin resistance were also detected in skeletal muscle and liver of HF-fed animals, these responses were observed much later (between 4 and 8 weeks of HF feeding), and they were not detected in visceral adipose tissue until 14 weeks. Conclusions—During obesity induced by HF feeding, inflammation and insulin resistance develop in the vasculature well before these responses are detected in muscle, liver, or adipose tissue. This observation suggests that the vasculature is more susceptible than other tissues to the deleterious effects of nutrient overload.


Journal of Clinical Investigation | 2005

Leptin action in the forebrain regulates the hindbrain response to satiety signals

Gregory J. Morton; James E. Blevins; Diana L. Williams; Kevin D. Niswender; Richard W. Gelling; Christopher J. Rhodes; Denis G. Baskin; Michael W. Schwartz

The capacity to adjust energy intake in response to changing energy requirements is a defining feature of energy homeostasis. Despite the identification of leptin as a key mediator of this process, the mechanism whereby changes of body adiposity are coupled to adaptive, short-term adjustments of energy intake remains poorly understood. To investigate the physiological role of leptin in the control of meal size and the response to satiety signals, and to identify brain areas mediating this effect, we studied Koletsky (fa(k)/fa(k)) rats, which develop severe obesity due to the genetic absence of leptin receptors. Our finding of markedly increased meal size and reduced satiety in response to the gut peptide cholecystokinin (CCK) in these leptin receptor-deficient animals suggests a critical role for leptin signaling in the response to endogenous signals that promote meal termination. To determine if the hypothalamic arcuate nucleus (ARC) (a key forebrain site of leptin action) mediates this leptin effect, we used adenoviral gene therapy to express either functional leptin receptors or a reporter gene in the area of the ARC of fa(k)/fa(k) rats. Restoration of leptin signaling to this brain area normalized the effect of CCK on the activation of neurons in the nucleus of the solitary tract and area postrema, key hindbrain areas for processing satiety-related inputs. This intervention also reduced meal size and enhanced CCK-induced satiety in fa(k)/fa(k) rats. These findings demonstrate that forebrain signaling by leptin, a long-term regulator of body adiposity, limits food intake on a meal-to-meal basis by regulating the hindbrain response to short-acting satiety signals.


Physiological Reviews | 2011

Leptin and the Central Nervous System Control of Glucose Metabolism

Gregory J. Morton; Michael W. Schwartz

The regulation of body fat stores and blood glucose levels is critical for survival. This review highlights growing evidence that leptin action in the central nervous system plays a key role in both processes. Investigation into underlying mechanisms has begun to clarify the physiological role of leptin in the control of glucose metabolism and raises interesting new possibilities for the treatment of diabetes and related disorders.


International Journal of Obesity | 2001

The NPY/AgRP neuron and energy homeostasis

Gregory J. Morton; Michael W. Schwartz

Kennedy hypothesized nearly 50 y ago that negative feedback regulation of body fat stores involves hormones that circulate in proportion to adiposity and enter the brain, where they exert inhibitory effects on food intake and energy balance. Recent studies implicate leptin and insulin as ‘adiposity signals’ to the brain that promote negative energy balance in two ways: by inhibiting ‘anabolic’ hypothalamic neuronal circuits that stimulate food intake and promote weight gain, and by activating ‘catabolic’ pathways that reduce food intake and body weight. Chief among candidate ‘anabolic’ effector pathways is the NPY/AgRP neuron, found only in the hypothalamic arcuate nucleus. These neurons make peptides that potently stimulate food intake not only by increasing neuropeptide Y (NPY) signaling, but by reducing melanocortin signaling via the release of agouti-related peptide (AgRP), an endogenous melanocortin 3/4 receptor antagonist. Since NPY/AgRP neurons express receptors for leptin and insulin and are inhibited by these hormones, they are activated by a decrease of leptin or insulin signaling. Fasting, uncontrolled diabetes, and genetic leptin deficiency are examples of conditions in which food intake increases via a mechanism hypothesized to involve NPY/AgRP neurons. Data are reviewed which illustrate the role of these neurons in adaptive and maladaptive states characterized by hyperphagia and weight gain.


PLOS Biology | 2005

Effects of hypothalamic neurodegeneration on energy balance

Allison W. Xu; Christopher B. Kaelin; Gregory J. Morton; Kayoko Ogimoto; Kimber L. Stanhope; James L. Graham; Denis G. Baskin; Peter J. Havel; Michael W. Schwartz; Gregory S. Barsh

Normal aging in humans and rodents is accompanied by a progressive increase in adiposity. To investigate the role of hypothalamic neuronal circuits in this process, we used a Cre-lox strategy to create mice with specific and progressive degeneration of hypothalamic neurons that express agouti-related protein (Agrp) or proopiomelanocortin (Pomc), neuropeptides that promote positive or negative energy balance, respectively, through their opposing effects on melanocortin receptor signaling. In previous studies, Pomc mutant mice became obese, but Agrp mutant mice were surprisingly normal, suggesting potential compensation by neuronal circuits or genetic redundancy. Here we find that Pomc-ablation mice develop obesity similar to that described for Pomc knockout mice, but also exhibit defects in compensatory hyperphagia similar to what occurs during normal aging. Agrp-ablation female mice exhibit reduced adiposity with normal compensatory hyperphagia, while animals ablated for both Pomc and Agrp neurons exhibit an additive interaction phenotype. These findings provide new insight into the roles of hypothalamic neurons in energy balance regulation, and provide a model for understanding defects in human energy balance associated with neurodegeneration and aging.

Collaboration


Dive into the Gregory J. Morton's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kayoko Ogimoto

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brent E. Wisse

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Thomas H. Meek

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong T. Nguyen

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge