Network


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

Hotspot


Dive into the research topics where Lora K. Heisler is active.

Publication


Featured researches published by Lora K. Heisler.


Neuron | 2006

Serotonin reciprocally regulates melanocortin neurons to modulate food intake

Lora K. Heisler; Erin E. Jobst; Gregory M. Sutton; Ligang Zhou; Erzsebet Borok; Zoë D. Thornton-Jones; Hongyan Liu; Jeffrey M. Zigman; Nina Balthasar; Toshiro Kishi; Charlotte E. Lee; Carl J. Aschkenasi; Chen Yu Zhang; Jia Yu; Olivier Boss; Kathleen G. Mountjoy; Peter G. Clifton; Bradford B. Lowell; Jeffrey M. Friedman; Tamas L. Horvath; Andrew A. Butler; Joel K. Elmquist; Michael A. Cowley

The neural pathways through which central serotonergic systems regulate food intake and body weight remain to be fully elucidated. We report that serotonin, via action at serotonin1B receptors (5-HT1BRs), modulates the endogenous release of both agonists and antagonists of the melanocortin receptors, which are a core component of the central circuitry controlling body weight homeostasis. We also show that serotonin-induced hypophagia requires downstream activation of melanocortin 4, but not melanocortin 3, receptors. These results identify a primary mechanism underlying the serotonergic regulation of energy balance and provide an example of a centrally derived signal that reciprocally regulates melanocortin receptor agonists and antagonists in a similar manner to peripheral adiposity signals.


Neuron | 2008

5-HT2CRs Expressed by Pro-Opiomelanocortin Neurons Regulate Energy Homeostasis

Yong Xu; Juli E. Jones; Daisuke Kohno; Kevin W. Williams; Charlotte E. Lee; Michelle J. Choi; Jason G. Anderson; Lora K. Heisler; Jeffrey M. Zigman; Bradford B. Lowell; Joel K. Elmquist

Summary Drugs activating 5-hydroxytryptamine 2C receptors (5-HT2CRs) potently suppress appetite, but the underlying mechanisms for these effects are not fully understood. To tackle this issue, we generated mice with global 5-HT2CR deficiency (2C null) and mice with 5-HT2CRs re-expression only in pro-opiomelanocortin (POMC) neurons (2C/POMC mice). We show that 2C null mice predictably developed hyperphagia, hyperactivity, and obesity and showed attenuated responses to anorexigenic 5-HT drugs. Remarkably, all these deficiencies were normalized in 2C/POMC mice. These results demonstrate that 5-HT2CR expression solely in POMC neurons is sufficient to mediate effects of serotoninergic compounds on food intake. The findings also highlight the physiological relevance of the 5-HT2CR-melanocortin circuitry in the long-term regulation of energy balance.


The Journal of Neuroscience | 2007

Serotonin Activates the Hypothalamic–Pituitary–Adrenal Axis via Serotonin 2C Receptor Stimulation

Lora K. Heisler; Nina Pronchuk; Katsunori Nonogaki; Ligang Zhou; Jacob Raber; Loraine Tung; Giles S. H. Yeo; Stephen O'Rahilly; William F. Colmers; Joel K. Elmquist; Laurence H. Tecott

The dynamic interplay between serotonin [5-hydroxytryptamine (5-HT)] neurotransmission and the hypothalamic–pituitary–adrenal (HPA) axis has been extensively studied over the past 30 years, but the underlying mechanism of this interaction has not been defined. A possibility receiving little attention is that 5-HT regulates upstream corticotropin-releasing hormone (CRH) signaling systems via activation of serotonin 2C receptors (5-HT2CRs) in the paraventricular nucleus of the hypothalamus (PVH). Through complementary approaches in wild-type rodents and 5-HT2CR-deficient mice, we determined that 5-HT2CRs are necessary for 5-HT-induced HPA axis activation. We used laser-capture PVH microdissection followed by microarray analysis to compare the expression of 13 5-HTRs. Only 5-HT2CR and 5-HT1DR transcripts were consistently identified as present in the PVH, and of these, the 5-HT2CR was expressed at a substantially higher level. The abundant expression of 5-HT2CRs in the PVH was confirmed with in situ hybridization histochemistry. Dual-neurohistochemical labeling revealed that approximately one-half of PVH CRH-containing neurons coexpressed 5-HT2CR mRNA. We observed that PVH CRH neurons consistently depolarized in the presence of a high-affinity 5-HT2CR agonist, an effect blocked by a 5-HT2CR antagonist. Supporting the importance of 5-HT2CRs in CRH neuronal activity, genetic inactivation of 5-HT2CRs produced a downregulation of CRH mRNA and blunted CRH and corticosterone release after 5-HT compound administration. These findings thus provide a mechanistic explanation for the longstanding observation of HPA axis stimulation in response to 5-HT and thereby give insight into the neural circuitry mediating the complex neuroendocrine responses to stress.


Nature Neuroscience | 2012

Unraveling the brain regulation of appetite: lessons from genetics.

Giles S. H. Yeo; Lora K. Heisler

Over the past 20 years, genetic studies have illuminated critical pathways in the hypothalamus and brainstem mediating energy homeostasis, such as the melanocortin, leptin, 5-hydroxytryptamine and brain-derived neurotrophic factor signaling axes. The identification of these pathways necessary for appropriate appetitive responses to energy state has yielded insight into normal homeostatic processes. Although monogenic alterations in each of these axes result in severe obesity, such cases remain rare. The major burden of disease is carried by those with common obesity, which has so far resisted yielding meaningful biological insights. Recent progress into the etiology of common obesity has been made with genome-wide association studies. Such studies now reveal more than 32 different candidate obesity genes, most of which are highly expressed or known to act in the CNS, emphasizing, as in rare monogenic forms of obesity, the role of the brain in predisposition to obesity.


Cell Metabolism | 2007

Serotonin 2C Receptor Agonists Improve Type 2 Diabetes via Melanocortin-4 Receptor Signaling Pathways

Ligang Zhou; Gregory M. Sutton; Justin J. Rochford; Robert K. Semple; Daniel D. Lam; Laura J. Oksanen; Zoë D. Thornton-Jones; Peter G. Clifton; Chen Yu Yueh; Mark L. Evans; Rory J. McCrimmon; Joel K. Elmquist; Andrew A. Butler; Lora K. Heisler

Summary The burden of type 2 diabetes and its associated premature morbidity and mortality is rapidly growing, and the need for novel efficacious treatments is pressing. We report here that serotonin 2C receptor (5-HT2CR) agonists, typically investigated for their anorectic properties, significantly improve glucose tolerance and reduce plasma insulin in murine models of obesity and type 2 diabetes. Importantly, 5-HT2CR agonist-induced improvements in glucose homeostasis occurred at concentrations of agonist that had no effect on ingestive behavior, energy expenditure, locomotor activity, body weight, or fat mass. We determined that this primary effect on glucose homeostasis requires downstream activation of melanocortin-4 receptors (MC4Rs), but not MC3Rs. These findings suggest that pharmacological targeting of 5-HT2CRs may enhance glucose tolerance independently of alterations in body weight and that this may prove an effective and mechanistically novel strategy in the treatment of type 2 diabetes.


Genes, Brain and Behavior | 2007

Serotonin 5‐HT2C receptors regulate anxiety‐like behavior

Lora K. Heisler; Ligang Zhou; P. Bajwa; J. Hsu; Laurence H. Tecott

Central serotonin (5‐hydroxytryptamine, 5‐HT) systems have been implicated in the pathophysiology and treatment of anxiety disorders, which are among the world’s most prevalent psychiatric conditions. Here, we report that the 5‐HT2C receptor (5‐HT2CR) subtype is critically involved in regulating behaviors characteristic of anxiety using male 5‐HT2CR knockout (KO) mice. Specific neural substrates underlying the 5‐HT2CR KO anxiolytic phenotype were investigated, and we report that 5‐HT2CR KO mice display a selective blunting of extended amygdala corticotropin‐releasing hormone neuronal activation in response to anxiety stimuli. These findings illustrate a mechanism through which 5‐HT2CRs affect anxiety‐related behavior and provide insight into the neural circuitry mediating the complex psychological process of anxiety.


Pharmacology, Biochemistry and Behavior | 2010

Brain serotonin system in the coordination of food intake and body weight.

Daniel D. Lam; Alastair S. Garfield; Oliver J. Marston; Jill Shaw; Lora K. Heisler

An inverse relationship between brain serotonin and food intake and body weight has been known for more than 30 years. Specifically, augmentation of brain serotonin inhibits food intake, while depletion of brain serotonin promotes hyperphagia and weight gain. Through the decades, serotonin receptors have been identified and their function in the serotonergic regulation of food intake clarified. Recent refined genetic studies now indicate that a primary mechanism through which serotonin influences appetite and body weight is via serotonin 2C receptor (5-HT(2C)R) and serotonin 1B receptor (5-HT(1B)R) influencing the activity of endogenous melanocortin receptor agonists and antagonists at the melanocortin 4 receptor (MC4R). However, other mechanisms are also possible and the challenge of future research is to delineate them in the complete elucidation of the complex neurocircuitry underlying the serotonergic control of appetite and body weight.


The Journal of Physiology | 2009

Pharmacological targeting of the serotonergic system for the treatment of obesity.

Alastair S. Garfield; Lora K. Heisler

The attenuation of food intake as induced by an increase in serotonergic (5‐hydroxytryptamine, 5‐HT) efficacy has been a target of antiobesity pharmacotherapies. However, the induction of tolerance and/or side‐effects limited the clinical utility of the earliest serotonin‐related medications. With the global prevalence of obesity rising, there has been renewed interest in the manipulation of the serotonergic system as a point of pharmacological intervention. The serotonin2C receptor (5‐HT2CR), serotonin1B (rodent)/serotonin1Dβ (human) receptor (5‐HT1B/1DβR) and serotonin6 receptor (5‐HT6R) represent the most promising serotonin receptor therapeutic targets. Canonical serotonin receptor compounds have given way to a myriad of novel receptor‐selective ligands, many of which have observable anorectic effects. Here we review serotonergic compounds reducing ingestive behaviour and discuss their clinical potential for the treatment of obesity.


Cell Metabolism | 2008

Identification of Adropin as a Secreted Factor Linking Dietary Macronutrient Intake with Energy Homeostasis and Lipid Metabolism

K. Ganesh Kumar; James L. Trevaskis; Daniel D. Lam; Gregory M. Sutton; Robert A. Koza; Vladimir N. Chouljenko; Konstantin G. Kousoulas; Pamela M. Rogers; Robert A. Kesterson; Marie Thearle; Anthony W. Ferrante; Randall L. Mynatt; Thomas P. Burris; Jesse Z. Dong; Heather A. Halem; Michael D. Culler; Lora K. Heisler; Jacqueline M. Stephens; Andrew A. Butler

Obesity and nutrient homeostasis are linked by mechanisms that are not fully elucidated. Here we describe a secreted protein, adropin, encoded by a gene, Energy Homeostasis Associated (Enho), expressed in liver and brain. Liver Enho expression is regulated by nutrition: lean C57BL/6J mice fed high-fat diet (HFD) exhibited a rapid increase, while fasting reduced expression compared to controls. However, liver Enho expression declines with diet-induced obesity (DIO) associated with 3 months of HFD or with genetically induced obesity, suggesting an association with metabolic disorders in the obese state. In DIO mice, transgenic overexpression or systemic adropin treatment attenuated hepatosteatosis and insulin resistance independently of effects on adiposity or food intake. Adropin regulated expression of hepatic lipogenic genes and adipose tissue peroxisome proliferator-activated receptor gamma, a major regulator of lipogenesis. Adropin may therefore be a factor governing glucose and lipid homeostasis, which protects against hepatosteatosis and hyperinsulinemia associated with obesity.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Identification of neuronal subpopulations that project from hypothalamus to both liver and adipose tissue polysynaptically

Sarah Stanley; Shirly Pinto; Jeremy P. Segal; Cristian A. Pérez; Agnes Viale; Jeff DeFalco; Xiaoli Cai; Lora K. Heisler; Jeffrey M. Friedman

The autonomic nervous system regulates fuel availability and energy storage in the liver, adipose tissue, and other organs; however, the molecular components of this neural circuit are poorly understood. We sought to identify neural populations that project from the CNS indirectly through multisynaptic pathways to liver and epididymal white fat in mice using pseudorabies virus strains expressing different reporters together with BAC transgenesis and immunohistochemistry. Neurons common to both circuits were identified in subpopulations of the paraventricular nucleus of the hypothalamus (PVH) by double labeling with markers expressed in viruses injected in both sites. The lateral hypothalamus and arcuate nucleus of the hypothalamus and brainstem regions (nucleus of the solitary tract and A5 region) also project to both tissues but are labeled at later times. Connections from these same sites to the PVH were evident after direct injection of virus into the PVH, suggesting that these regions lie upstream of the PVH in a common pathway to liver and adipose tissue (two metabolically active organs). These common populations of brainstem and hypothalamic neurons express neuropeptide Y and proopiomelanocortin in the arcuate nucleus, melanin-concentrating hormone, and orexin in the lateral hypothalamus and in the corticotrophin-releasing hormone and oxytocin in the PVH. The delineation of this circuitry will facilitate a functional analysis of the possible role of these potential command-like neurons to modulate autonomic outflow and coordinate metabolic responses in liver and adipose tissue.

Collaboration


Dive into the Lora K. Heisler's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel D. Lam

Technische Universität München

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ligang Zhou

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jill Shaw

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge