@phdthesis{Appl2007, author = {Appl, Thomas}, title = {Neurochemical and functional characterisation of the Melanin-concentrating hormone system in the rat brain}, url = {http://nbn-resolving.de/urn:nbn:de:kobv:517-opus-14604}, school = {Universit{\"a}t Potsdam}, year = {2007}, abstract = {The central melanin-concentrating hormone (MCH) system has been intensively studied for its involvement in the regulation of feeding behaviour and body weight regulation. The importance of the neuropeptide MCH in the control of energy balance has been underlined by MCH knock out and Melanin-concentrating hormone receptor subtype 1 (MCHR-1) knock-out animals. The anorectic and anti-obesity effects of selective MCHR-1 antagonists have confirmed the notion that pharmacological blockade of MCHR-1 is a potential therapeutic approach for obesity. First aim of this work is to study the neurochemical "equipment" of MCHR-1 immunoreactive neurons by double-labelling immunohistochemistry within the rat hypothalamus. Of special interest is the neuroanatomical identification of other hypothalamic neuropeptides that are co-distributed with MCHR-1. A second part of this study deals with the examination of neuronal activation patterns after pharmacological or physiological, feeding-related stimuli and was introduced to further understand central regulatory mechanisms of the MCH system. In the first part of work, I wanted to neurochemically characterize MCHR-1 immunoreactive neurons in the rat hypothalamus for colocalisation with neuropeptides of interest. Therefore I performed an immunohistochemical colocalisation study using a specific antibody against MCHR-1 in combination with antibodies against hypothalamic neuropeptides. I showed that MCHR-1 immunoreactivity (IR) was co-localised with orexin A in the lateral hypothalamus, and with adrenocorticotropic hormone and neuropeptide Y in the arcuate nucleus. Additionally, MCHR-1 IR was co-localised with the neuropeptides vasopressin and oxytocin in magnocellular neurons of the supraoptic and paraventricular hypothalamic nucleus and corticotrophin releasing hormone in the parvocellular division of the paraventricular hypothalamic nucleus. Moreover, for the first time MCHR-1 immunoreactivity was found in both the adenohypophyseal and neurohypophyseal part of the rat pituitary. These results provide the neurochemical basis for previously described potential physiological actions of MCH at its target receptor. In particular, the MCHR-1 may be involved not only in food intake regulation, but also in other physiological actions such as fluid regulation, reproduction and stress response, possibly through here examined neuropeptides. Central activation patterns induced by pharmacological or physiological stimulation can be mapped using c-Fos immunohistochemistry. In the first experimental design, central administration (icv) of MCH in the rat brain resulted in acute and significant increase of food and water intake, but this animal treatment did not induce a specific c-Fos induction pattern in hypothalamic nuclei. In contrast, sub-chronic application of MCHR-1 antagonist promoted a significant decrease in food- and water intake during an eight day treatment period. A qualitative analysis of c-Fos immunohistochemistry of sections derived from MCHR-1 antagonist treated animals showed a specific neuronal activation in the paraventricular nucleus, the supraoptic nucleus and the dorsomedial hypothalamus. These results could be substantiated by quantitative evaluation of an automated, software-supported analysis of the c-Fos signal. Additionally, I examined the activation pattern of rats in a restricted feeding schedule (RFS) to identify pathways involved in hunger and satiety. Animals were trained for 9 days to feed during a three hour period. On the last day, food restricted animals was also allowed to feed for the three hours, while food deprived (FD) animals did not receive food. Mapping of neuronal activation showed a clear difference between stareved (FD) and satiated (FR) rats. FD animals showed significant induction of c-Fos in forebrain regions, several hypothalamic nuclei, amygdaloid thalamus and FR animals in the supraoptic nucleus and the paraventricular nucleus of the hypothalamus, and the nucleus of the solitary tract. In the lateral hypothalamus of FD rats, c-Fos IR showed strong colocalisation for Orexin A, but no co-staining for MCH immunoreactivity. However, a large number of c-Fos IR neurons within activated regions of FD and FR animals was co-localised with MCHR-1 within selected regions. To conclude, the experimental set-up of scheduled feeding can be used to induce a specific hunger or satiety activation pattern within the rat brain. My results show a differential activation by hunger signals of MCH neurons and furthermore, demonstrates that MCHR-1 expressing neurons may be essential parts of downstream processing of physiological feeding/hunger stimuli. In the final part of my work, the relevance of here presented studies is discussed with respect to possible introduction of MCHR-1 antagonists as drug candidates for the treatment of obesity.}, language = {en} }