Network


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

Hotspot


Dive into the research topics where Kathleen G. Mountjoy is active.

Publication


Featured researches published by Kathleen G. Mountjoy.


Cell | 2005

Divergence of Melanocortin Pathways in the Control of Food Intake and Energy Expenditure

Nina Balthasar; Louise T. Dalgaard; Charlotte E. Lee; Jia Yu; Hisayuki Funahashi; Todd D. Williams; Manuel Ferreira; Vinsee Tang; Robert A. McGovern; Christopher D. Kenny; Lauryn M. Christiansen; Elizabeth Edelstein; Brian Choi; Olivier Boss; Carl J. Aschkenasi; Chen Yu Zhang; Kathleen G. Mountjoy; Toshiro Kishi; Joel K. Elmquist; Bradford B. Lowell

Activation of melanocortin-4-receptors (MC4Rs) reduces body fat stores by decreasing food intake and increasing energy expenditure. MC4Rs are expressed in multiple CNS sites, any number of which could mediate these effects. To identify the functionally relevant sites of MC4R expression, we generated a loxP-modified, null Mc4r allele (loxTB Mc4r) that can be reactivated by Cre-recombinase. Mice homozygous for the loxTB Mc4r allele do not express MC4Rs and are markedly obese. Restoration of MC4R expression in the paraventricular hypothalamus (PVH) and a subpopulation of amygdala neurons, using Sim1-Cre transgenic mice, prevented 60% of the obesity. Of note, increased food intake, typical of Mc4r null mice, was completely rescued while reduced energy expenditure was unaffected. These findings demonstrate that MC4Rs in the PVH and/or the amygdala control food intake but that MC4Rs elsewhere control energy expenditure. Disassociation of food intake and energy expenditure reveals unexpected divergence in melanocortin pathways controlling energy balance.


The Journal of Comparative Neurology | 2003

Expression of melanocortin 4 receptor mRNA in the central nervous system of the rat

Toshiro Kishi; Carl J. Aschkenasi; Charlotte E. Lee; Kathleen G. Mountjoy; Clifford B. Saper; Joel K. Elmquist

The melanocortin 4 receptor (MC4‐R) plays a pivotal role in maintaining energy homeostasis in rodents and humans. For example, MC4‐R deletion or mutation results in obesity, hyperphagia, and insulin resistance. Additionally, subsets of leptin‐induced autonomic responses can be blocked by melanocortin receptor antagonism, suggesting that MC4‐R–expressing neurons are downstream targets of leptin. However, the critical autonomic control sites expressing MC4‐Rs are still unclear. In the present study, we systematically examined the distribution of MC4‐R mRNA in the adult rat central nervous system, including the spinal cord, by using in situ hybridization histochemistry (ISHH) with a novel cRNA probe. Autonomic control sites expressing MC4‐R mRNA in the hypothalamus included the anteroventral periventricular, ventromedial preoptic, median preoptic, paraventricular, dorsomedial, and arcuate nuclei. The subfornical organ, dorsal hypothalamic, perifornical, and posterior hypothalamic areas were also observed to express MC4‐R mRNA. Within extrahypothalamic autonomic control sites, MC4‐R–specific hybridization was evident in the infralimbic and insular cortices, bed nucleus of the stria terminalis, central nucleus of the amygdala, periaqueductal gray, lateral parabrachial nucleus, nucleus of the solitary tract, dorsal motor nucleus of the vagus (DMV), and intermediolateral nucleus of the spinal cord (IML). By using dual‐label ISHH, we confirmed that the cells expressing MC4‐R mRNA in the IML and DMV were autonomic preganglionic neurons as cells in both sites coexpressed choline acetyltransferase mRNA. The distribution of MC4‐R mRNA is consistent with the proposed roles of central melanocortin systems in feeding and autonomic regulation. J. Comp. Neurol. 457:213–235, 2003.


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.


American Journal of Physiology-endocrinology and Metabolism | 1998

Melanin-concentrating hormone: a functional melanocortin antagonist in the hypothalamus

David S. Ludwig; Kathleen G. Mountjoy; Jeffrey B. Tatro; Jennifer Gillette; Robert C. Frederich; Jeffrey S. Flier; Eleftheria Maratos-Flier

Melanin-concentrating hormone (MCH) and alpha-melanocyte-stimulating hormone (alpha-MSH) demonstrate opposite actions on skin coloration in teleost fish. Both peptides are present in the mammalian brain, although their specific physiological roles remain largely unknown. In this study, we examined the interactions between MCH and alpha-MSH after intracerebroventricular administration in rats. MCH increased food intake in a dose-dependent manner and lowered plasma glucocorticoid levels through a mechanism involving ACTH. In contrast, alpha-MSH decreased food intake and increased glucocorticoid levels. MCH, at a twofold molar excess, antagonized both actions of alpha-MSH. alpha-MSH, at a threefold molar excess, blocked the orexigenic properties of MCH. MCH did not block alpha-MSH binding or the ability of alpha-MSH to induce cAMP in cells expressing either the MC3 or MC4 receptor, the principal brain alpha-MSH receptor subtypes. These data suggest that MCH and alpha-MSH exert opposing and antagonistic influences on feeding behavior and the stress response and may function in a coordinate manner to regulate metabolism through a novel mechanism mediated in part by an MCH receptor.Melanin-concentrating hormone (MCH) and α-melanocyte-stimulating hormone (α-MSH) demonstrate opposite actions on skin coloration in teleost fish. Both peptides are present in the mammalian brain, although their specific physiological roles remain largely unknown. In this study, we examined the interactions between MCH and α-MSH after intracerebroventricular administration in rats. MCH increased food intake in a dose-dependent manner and lowered plasma glucocorticoid levels through a mechanism involving ACTH. In contrast, α-MSH decreased food intake and increased glucocorticoid levels. MCH, at a twofold molar excess, antagonized both actions of α-MSH. α-MSH, at a threefold molar excess, blocked the orexigenic properties of MCH. MCH did not block α-MSH binding or the ability of α-MSH to induce cAMP in cells expressing either the MC3 or MC4 receptor, the principal brain α-MSH receptor subtypes. These data suggest that MCH and α-MSH exert opposing and antagonistic influences on feeding behavior and the stress response and may function in a coordinate manner to regulate metabolism through a novel mechanism mediated in part by an MCH receptor.


Molecular and Cellular Endocrinology | 1997

OBESITY, DIABETES AND FUNCTIONS FOR PROOPIOMELANOCORTIN-DERIVED PEPTIDES

Kathleen G. Mountjoy; Jencia Wong

Melanocortin peptides (adrenocorticotropin (ACTH), alpha-,beta-, and gamma-melanocyte stimulating hormone (MSH), and fragments thereof) derived from proopiomelanocortin (POMC) have a diverse array of biological activities, many of which have yet to be fully elucidated. The recent cloning of a family of five distinct melanocortin receptors through which these peptides act has provided the tools to further our understanding of melanocortin peptide functions. Early work on melanocortin peptides focused on their roles in pigmentation, adrenocortical function, the immune, central and peripheral nervous systems. Although melanocortin peptides have long been known to affect lipolysis, characterisation of the melanocortin receptors has opened up several lines of evidence for important roles in the development of obesity, insulin resistance and type II diabetes. We present here a review of the current evidence for melanocortin peptides playing such a role, and based on this evidence, a model for melanocortin peptides and their receptors in maintaining energy balance.


Biochemical Journal | 2010

Functions for pro-opiomelanocortin-derived peptides in obesity and diabetes.

Kathleen G. Mountjoy

Melanocortin peptides, derived from POMC (pro-opiomelanocortin) are produced in the ARH (arcuate nucleus of the hypothalamus) neurons and the neurons in the commissural NTS (nucleus of the solitary tract) of the brainstem, in anterior and intermediate lobes of the pituitary, skin and a wide range of peripheral tissues, including reproductive organs. A hypothetical model for functional roles of melanocortin receptors in maintaining energy balance was proposed in 1997. Since this time, there has been an extraordinary amount of knowledge gained about POMC-derived peptides in relation to energy homoeostasis. Development of a Pomc-null mouse provided definitive proof that POMC-derived peptides are critical for the regulation of energy homoeostasis. The melanocortin system consists of endogenous agonists and antagonists, five melanocortin receptor subtypes and receptor accessory proteins. The melanocortin system, as is now known, is far more complex than most of us could have imagined in 1997, and, similarly, the importance of this system for regulating energy homoeostasis in the general human population is much greater than we would have predicted. Of the known factors that can cause human obesity, or protect against it, the melanocortin system is by far the most significant. The present review is a discussion of the current understanding of the roles and mechanism of action of POMC, melanocortin receptors and AgRP (agouti-related peptide) in obesity and Type 2 diabetes and how the central and/or peripheral melanocortin systems mediate nutrient, leptin, insulin, gut hormone and cytokine regulation of energy homoeostasis.


Developmental Brain Research | 1998

Melanocortin-4 receptor mRNA expression in the developing autonomic and central nervous systems

Kathleen G. Mountjoy; J. Martin Wild

MC4-R mRNA expression was investigated in fetal rats (E14-E20) using in situ hybridisation. The autonomic nervous system showed the highest levels of MC4-R mRNA expression. In the spinal cord, dense signal was seen over the future intermediolateral cell column and dorsal horn. In the brain, MC4-R was expressed by E14 in diencephalon neuroepithelia, telencephalon, lamina terminalis and spinal trigeminal nucleus and was expressed by E19 throughout many regions of the brain.


Brain Research | 2001

Differential expression of cocaine- and amphetamine-regulated transcript and agouti related-protein in chronically food-restricted sheep

Belinda A. Henry; Alexandra Rao; Bettina Anastasia Ikenasio; Kathleen G. Mountjoy; Alan J. Tilbrook; Iain J. Clarke

Recently, much attention has focused on the role of the melanocortin system in the regulation of energy homeostasis, especially the satiety effects of the pro-opiomelanocortin (POMC)-derived peptide alpha-melanocyte stimulating hormone (alpha-MSH). We have found that POMC mRNA levels are similar in fat and thin sheep and the current study sought to further characterize the effects of nutritional status on the melanocortin system. To this end, we studied the expression of agouti-related peptide (AGRP) (an endogenous antagonist of alpha-MSH) and cocaine- and amphetamine-regulated transcript (CART), which is co-localized within POMC cells of the arcuate nucleus (ARC) in rodents. Twelve ovariectomized ewes were randomly divided into two groups and fed a maintenance (n=6) or restricted diet (n=6). At the time of experimentation, the animals had significantly (P<0.0001) different bodyweights (53.4+/-2.2 kg, ad libitum vs. 30.4+/-1.2 kg, food-restricted), which was largely due to altered body fat deposits. In situ hybridization was used to study the expression of POMC, AGRP and CART. The expression of POMC in the ARC was similar in ad libitum and food-restricted animals but the expression of AGRP was profoundly increased in the food-restricted group. The expression of CART was abundant throughout the hypothalamus but was not found in the ARC. In food-restricted animals, the expression of CART was lower in the retrochiasmatic nucleus (P<0.01), paraventricular nucleus (P<0.001), the dorsomedial nucleus and the lateral hypothalamic area (P<0.05), but was higher (P<0.01) in the posterior hypothalamic area. Thus, long-term changes in nutritional status have profound effects on the expression of AGRP and CART in the hypothalamus.


Diabetes Research and Clinical Practice | 2002

Melanocortin-3 receptor gene variants in a Maori kindred with obesity and early onset type 2 diabetes

Jencia Wong; Donald R. Love; Cam Kyle; Andre Daniels; Marie White; Alistair W. Stewart; Audrey H. Schnell; Robert C. Elston; Ian Holdaway; Kathleen G. Mountjoy

Genetic studies suggest a diabetes susceptibility locus on human chromosome 20, near the melanocortin receptor-3 (MC3-R) gene. We examined the MC3-R as a candidate gene for type 2 diabetes in 12 members of a large Maori kindred with multiple affected members. The coding region of the MC3-R gene was sequenced for both diabetic and non-diabetic individuals. Two separate single base pair substitutions were found in the MC3-R coding sequence and these resulted in amino acid changes, Lysine6Threonine and Isoleucine81Valine. Neither of these MC3-R variants tracked with the presence of diabetes. Furthermore, the variant and wild type MC3-R showed similar functional coupling to adenylyl cyclase. A polymorphic marker (D20S32e) close to the human MC3-R (hMC3-R) coding sequence was investigated in a 60-member pedigree for association with diabetes and other metabolic parameters. There was an association between D20S32e genotype and fasting insulin (P=0.0085) and the insulin resistance index, HOMA-R (P=0.0042). An association was also found between genotype and HDL levels during oral glucose tolerance testing (P=0.0037). These associations were independent of BMI, age, gender and diabetes. Our data do not support a role for variations in the coding region of the hMC3-R in the development of type 2 diabetes in this Maori kindred, but suggest that a locus on chromosome 20 q, close to D20S32e, may contribute to both insulin secretion and action in the Maori kindred.


Endocrinology | 1999

Agouti Antagonism of Melanocortin-4 Receptor: Greater Effect with Desacetyl-α-Melanocyte-Stimulating Hormone (MSH) than withα -MSH

Kathleen G. Mountjoy; Derril H. Willard; William O. Wilkison

Desacetyl-alpha-MSH is more abundant than alpha-MSH in the brain, the fetus, human blood, and amniotic fluid, but there is little information on its ability to interact with melanocortin receptors. The aim of this study is to compare and contrast the ability of desacetyl-alpha-MSH and alpha-MSH to couple melanocortin receptors stably expressed in HEK293 cells, to the protein kinase A (PKA) signaling pathway. Desacetyl-alpha-MSH activated mouse MC1, MC3, MC4 and MC5 receptors with EC50s = 0.13, 0.96, 0.53, and 0.84 nM, and alpha-MSH activated these receptors with EC50s = 0.17, 0.88, 1.05, and 1.34 nM, respectively. Mouse agouti protein competitively antagonized alpha-MSH and desacetyl-alpha-MSH coupling to the MC1-R similarly. In contrast, mouse agouti protein antagonized desacetyl-alpha-MSH much more effectively and potently than alpha-MSH coupling the MC4-R to the PKA signaling pathway. Furthermore, mouse agouti protein (10 nM) significantly reduced (1.4-fold) the maximum response of mMC4-R to desacetyl-alpha-MSH and 100 nM mouse agouti significantly increased (4.8-fold) the EC50. Minimal antagonism of alpha-MSH coupling mMC4-R to the PKA signaling pathway was observed with 10 nM mouse agouti, whereas both 50 and 100 nM mouse agouti appeared to reduce the maximum reponse (1.1- and 1.3-fold, respectively) and increase the EC50 (2.5- and 3.4-fold respectively). Mouse agouti protein did not significantly antagonize either alpha-MSH or desacetyl-alpha-MSH coupling mouse MC3 and MC5 receptors. Understanding the similarities and differences in activation of melanocortin receptors by desacetyl-alpha-MSH and alpha-MSH will contribute to delineating the functional roles for these endogenous melanocortin peptides.

Collaboration


Dive into the Kathleen G. Mountjoy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joel K. Elmquist

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rikus Botha

University of Auckland

View shared research outputs
Top Co-Authors

Avatar

Carl J. Aschkenasi

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge