1
|
Jászberényi M, Thurzó B, Jayakumar AR, Schally AV. The Aggravating Role of Failing Neuropeptide Networks in the Development of Sporadic Alzheimer's Disease. Int J Mol Sci 2024; 25:13086. [PMID: 39684795 DOI: 10.3390/ijms252313086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease imposes an increasing burden on aging Western societies. The disorder most frequently appears in its sporadic form, which can be caused by environmental and polygenic factors or monogenic conditions of incomplete penetrance. According to the authors, in the majority of cases, Alzheimer's disease represents an aggravated form of the natural aging of the central nervous system. It can be characterized by the decreased elimination of amyloid β1-42 and the concomitant accumulation of degradation-resistant amyloid plaques. In the present paper, the dysfunction of neuropeptide regulators, which contributes to the pathophysiologic acceleration of senile dementia, is reviewed. However, in the present review, exclusively those neuropeptides or neuropeptide families are scrutinized, and the authors' investigations into their physiologic and pathophysiologic activities have made significant contributions to the literature. Therefore, the pathophysiologic role of orexins, neuromedins, RFamides, corticotrope-releasing hormone family, growth hormone-releasing hormone, gonadotropin-releasing hormone, ghrelin, apelin, and natriuretic peptides are discussed in detail. Finally, the therapeutic potential of neuropeptide antagonists and agonists in the inhibition of disease progression is discussed here.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
| | - Balázs Thurzó
- Department of Pathophysiology, University of Szeged, P.O. Box 427, H-6701 Szeged, Hungary
- Emergency Patient Care Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Arumugam R Jayakumar
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V Schally
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
2
|
Keskin M, Avul S, Beyaz A, Koca N. The association of Neuromedin U levels and non-alcoholic fatty liver disease: A comparative analysis. Heliyon 2024; 10:e27291. [PMID: 38486771 PMCID: PMC10937677 DOI: 10.1016/j.heliyon.2024.e27291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024] Open
Abstract
This comprehensive study delves into the potential link between Neuromedin U (NmU) serum levels and the development of non-alcoholic fatty liver disease (NAFLD), a condition of increasing global prevalence and significant public health concern. The research provides a nuanced understanding of the disease's etiology by examining a cohort of 112 participants, including individuals with and without NAFLD. The study meticulously considers a spectrum of variables such as demographic factors, body composition metrics, and blood parameters. Advanced diagnostic tools like Fibroscan® are employed to ascertain NAFLD presence, ensuring accurate and reliable results. The investigation reveals a noteworthy correlation between NAFLD and several risk factors, notably obesity, increased waist and neck circumferences, hypertriglyceridemia, and insulin resistance. These findings underscore the multifactorial nature of NAFLD and its intricate connection with metabolic syndromes. Intriguingly, the study observes lower NmU levels in individuals diagnosed with NAFLD. However, the role of NmU as an independent risk factor for NAFLD remains inconclusive, warranting further investigation. Although triglyceride level was observed to be an independent risk factor for NAFLD, this relationship was not associated with NmU. This research contributes significantly to the existing knowledge on NAFLD, highlighting the disease's complexity and the interplay of various risk factors. It also opens up new avenues for future research, particularly in exploring the role of NmU within the metabolic pathways associated with NAFLD. The insights gained from this study could guide the development of novel diagnostic and therapeutic strategies for NAFLD, addressing a crucial need in contemporary healthcare. In conclusion, the findings of this study not only enhance the understanding of NAFLD's pathophysiology but also emphasize the importance of comprehensive risk factor analysis in the management and prevention of this growing health concern.
Collapse
Affiliation(s)
- Murat Keskin
- Department of Gastroenterology, Medicana Private Hospital, Bursa, Turkey
| | - Sercan Avul
- Department of Internal Medicine, Birecik State Hospital, Şanlıurfa, Turkey
| | - Aylin Beyaz
- Department of Biochemistry, Kagizman State Hospital, Kars, Turkey
| | - Nizameddin Koca
- Department of Internal Medicine, University of Health Sciences, Bursa Faculty of Medicine, Bursa City Training & Research Hospital, Bursa, Turkey
| |
Collapse
|
3
|
Engin A. Misalignment of Circadian Rhythms in Diet-Induced Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:27-71. [PMID: 39287848 DOI: 10.1007/978-3-031-63657-8_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronize them with daily cycles. While the central clock in the suprachiasmatic nucleus (SCN) is mainly synchronized by the light/dark cycles, the peripheral clocks react to other stimuli, including the feeding/fasting state, nutrients, sleep-wake cycles, and physical activity. During the disruption of circadian rhythms due to genetic mutations or social and occupational obligations, incorrect arrangement between the internal clock system and environmental rhythms leads to the development of obesity. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition leads to uncoupling of the peripheral clocks from the central pacemaker and to the development of metabolic disorders. The strong coupling of the SCN to the light-dark cycle creates a situation of misalignment when food is ingested during the "wrong" time of day. Food-anticipatory activity is mediated by a self-sustained circadian timing, and its principal component is a food-entrainable oscillator. Modifying the time of feeding alone greatly affects body weight, whereas ketogenic diet (KD) influences circadian biology, through the modulation of clock gene expression. Night-eating behavior is one of the causes of circadian disruption, and night eaters have compulsive and uncontrolled eating with severe obesity. By contrast, time-restricted eating (TRE) restores circadian rhythms through maintaining an appropriate daily rhythm of the eating-fasting cycle. The hypothalamus has a crucial role in the regulation of energy balance rather than food intake. While circadian locomotor output cycles kaput (CLOCK) expression levels increase with high-fat diet-induced obesity, peroxisome proliferator-activated receptor-alpha (PPARα) increases the transcriptional level of brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like 1 (BMAL1) in obese subjects. In this context, effective timing of chronotherapies aiming to correct SCN-driven rhythms depends on an accurate assessment of the SCN phase. In fact, in a multi-oscillator system, local rhythmicity and its disruption reflects the disruption of either local clocks or central clocks, thus imposing rhythmicity on those local tissues, whereas misalignment of peripheral oscillators is due to exosome-based intercellular communication.Consequently, disruption of clock genes results in dyslipidemia, insulin resistance, and obesity, while light exposure during the daytime, food intake during the daytime, and sleeping during the biological night promote circadian alignment between the central and peripheral clocks. Thus, shift work is associated with an increased risk of obesity, diabetes, and cardiovascular diseases because of unusual eating times as well as unusual light exposure and disruption of the circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
4
|
Elhadi K, Daiwile AP, Cadet JL. Modeling methamphetamine use disorder and relapse in animals: short- and long-term epigenetic, transcriptional., and biochemical consequences in the rat brain. Neurosci Biobehav Rev 2023; 155:105440. [PMID: 38707245 PMCID: PMC11068368 DOI: 10.1016/j.neubiorev.2023.105440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 05/07/2024]
Abstract
Methamphetamine use disorder (MUD) is a neuropsychiatric disorder characterized by binge drug taking episodes, intervals of abstinence, and relapses to drug use even during treatment. MUD has been modeled in rodents and investigators are attempting to identify its molecular bases. Preclinical experiments have shown that different schedules of methamphetamine self-administration can cause diverse transcriptional changes in the dorsal striatum of Sprague-Dawley rats. In the present review, we present data on differentially expressed genes (DEGs) identified in the rat striatum following methamphetamine intake. These include genes involved in transcription regulation, potassium channel function, and neuroinflammation. We then use the striatal data to discuss the potential significance of the molecular changes induced by methamphetamine by reviewing concordant or discordant data from the literature. This review identified potential molecular targets for pharmacological interventions. Nevertheless, there is a need for more research on methamphetamine-induced transcriptional consequences in various brain regions. These data should provide a more detailed neuroanatomical map of methamphetamine-induced changes and should better inform therapeutic interventions against MUD.
Collapse
Affiliation(s)
- Khalid Elhadi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Atul P. Daiwile
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, 21224
| |
Collapse
|
5
|
Tian R, Asadollahpour Nanaie H, Wang X, Dalai B, Zhao M, Wang F, Li H, Yang D, Zhang H, Li Y, Wang T, Luan T, Wu J. Genomic adaptation to extreme climate conditions in beef cattle as a consequence of cross-breeding program. BMC Genomics 2023; 24:186. [PMID: 37024818 PMCID: PMC10080750 DOI: 10.1186/s12864-023-09235-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Understanding the evolutionary forces related to climate changes that have been shaped genetic variation within species has long been a fundamental pursuit in biology. In this study, we generated whole-genome sequence (WGS) data from 65 cross-bred and 45 Mongolian cattle. Together with 62 whole-genome sequences from world-wide cattle populations, we estimated the genetic diversity and population genetic structure of cattle populations. In addition, we performed comparative population genomics analyses to explore the genetic basis underlying variation in the adaptation to cold climate and immune response in cross-bred cattle located in the cold region of China. To elucidate genomic signatures that underlie adaptation to cold climate, we performed three statistical measurements, fixation index (FST), log2 nucleotide diversity (θπ ratio) and cross population composite likelihood ratio (XP-CLR), and further investigated the results to identify genomic regions under selection for cold adaptation and immune response-related traits. RESULTS By generating WGS data, we investigated the population genetic structure and phylogenetic relationship of studied cattle populations. The results revealed clustering of cattle groups in agreement with their geographic distribution. We detected noticeable genetic diversity between indigenous cattle ecotypes and commercial populations. Analysis of population structure demonstrated evidence of shared genetic ancestry between studied cross-bred population and both Red-Angus and Mongolian breeds. Among all studied cattle populations, the highest and lowest levels of linkage disequilibrium (LD) per Kb were detected in Holstein and Rashoki populations (ranged from ~ 0.54 to 0.73, respectively). Our search for potential genomic regions under selection in cross-bred cattle revealed several candidate genes related with immune response and cold shock protein on multiple chromosomes. We identified some adaptive introgression genes with greater than expected contributions from Mongolian ancestry into Molgolian x Red Angus composites such as TRPM8, NMUR1, PRKAA2, SMTNL2 and OXR1 that are involved in energy metabolism and metabolic homeostasis. In addition, we detected some candidate genes probably associated with immune response-related traits. CONCLUSION The study identified candidate genes involved in responses to cold adaptation and immune response in cross-bred cattle, including new genes or gene pathways putatively involved in these adaptations. The identification of these genes may clarify the molecular basis underlying adaptation to extreme environmental climate and as such they might be used in cattle breeding programs to select more efficient breeds for cold climate regions.
Collapse
Affiliation(s)
- Rugang Tian
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China.
| | - Hojjat Asadollahpour Nanaie
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiao Wang
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Baolige Dalai
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Meng Zhao
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Feng Wang
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Hui Li
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Ding Yang
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Hao Zhang
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Yuan Li
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Tingyue Wang
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Hohhot, 010031, China
| | - Tu Luan
- Faculty of Biosciences, Norwegian University of Life Sciences, As, Norway
| | - Jianghong Wu
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao, China.
| |
Collapse
|
6
|
Assar DH, Mokhbatly AAA, ELazab MFA, Ghazy EW, Gaber AA, Elbialy ZI, Hassan AA, Nabil A, Asa SA. Silver nanoparticles induced testicular damage targeting NQO1 and APE1 dysregulation, apoptosis via Bax/Bcl-2 pathway, fibrosis via TGF-β/α-SMA upregulation in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:26308-26326. [PMID: 36367645 PMCID: PMC9995601 DOI: 10.1007/s11356-022-23876-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
In medicine, silver nanoparticles (AgNPs) are employed often. They do, however, have negative impacts, particularly on the reproductive organs. This research aimed to assess AgNP impact on the testis and the possible intracellular mechanisms to induce testicular deteriorations in rats at various concentrations and different time intervals. Sprague Dawley rats (n = 40) were allocated into four equal groups: the control one, and three other groups injected intra-peritoneally with AgNP solution 0.25, 0.5, and 1 mg/kg b.w. respectively for 15 and 30 days. Our findings revealed that AgNPs reduced body and testicular weights, estradiol (E2) and testosterone (T) hormone levels, and sperm parameters while elevating the nitric oxide and malondialdehyde levels with inhibition of reduced glutathione contents in testicular tissue. Interestingly, AgNPs significantly upregulated the testicular inducible nitric oxide synthase, B cell lymphoma 2 (Bcl-2)-associated X, transforming growth factor, and alpha-smooth muscle actin (α-SMA) expression levels. However, apurinic/apyrimidinic endo deoxyribonuclease 1 (APE1), NAD (P) H quinone dehydrogenase 1 (NQO1), and Bcl-2 expression levels were all downregulated indicating exhaustion of body antioxidant and repairing defense mechanisms in testicles in comparison with the control rats. Various histological alterations were also detected which dramatically increased in rats sacrificed after 30 days such as loss of the lining cells of seminiferous tubules with no spermatozoa and tubular irregularities associated with thickening of their basement membranes. Immunolabeling implicated in the apoptotic pathway revealed a negative expression of Bcl-2 and marked immunoreactivity for caspase-3 after 30 days of AgNP treatment in comparison to the control rats. To our knowledge, there have been no previous publications on the role of the α-SMA, APE1, and NQO1 genes in the molecular pathogenesis of AgNP testicular cytotoxicity following AgNP acute and chronic exposure.
Collapse
Affiliation(s)
- Doaa H. Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Abd-Allah A. Mokhbatly
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Mohamed F. Abou ELazab
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Emad W. Ghazy
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Ahmed A. Gaber
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Zizy I. Elbialy
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Ayman A. Hassan
- High Technological Institute of Applied Health Sciences, Egypt Liver Research Institute and Hospital (ELRIAH), Sherbin, ElMansora Egypt
| | - Ahmed Nabil
- Beni-Suef University, Beni-Suef, Egypt, Egypt Liver Research Institute and Hospital (ELRIAH), Sherbin, ElMansora Egypt
| | - Samah Abou Asa
- Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| |
Collapse
|
7
|
Yokogi K, Goto Y, Otsuka M, Ojima F, Kobayashi T, Tsuchiba Y, Takeuchi Y, Namba M, Kohno M, Tetsuka M, Takeuchi S, Matsuyama M, Aizawa S. Neuromedin U-deficient rats do not lose body weight or food intake. Sci Rep 2022; 12:17472. [PMID: 36302800 PMCID: PMC9614009 DOI: 10.1038/s41598-022-21764-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/30/2022] [Indexed: 01/12/2023] Open
Abstract
Studies in genetically modified mice establish that essential roles of endogenous neuromedin U (NMU) are anorexigenic function and metabolic regulation, indicating that NMU is expected to be a potential target for anti-obesity agents. However, in central administration experiments in rats, inconsistent results have been obtained, and the essential role of NMU energy metabolism in rats remain unclear. This study aims to elucidate the role of endogenous NMU in rats. We generated NMU knockout (KO) rats that unexpectedly showed no difference in body weight, adiposity, circulating metabolic markers, body temperature, locomotor activity, and food consumption in both normal and high fat chow feeding. Furthermore, unlike reported in mice, expressions of Nmu and NMU receptor type 2 (Nmur2) mRNA were hardly detectable in the rat hypothalamic nuclei regulating feeding and energy metabolism, including the arcuate nucleus and paraventricular nucleus, while Nmu was expressed in pars tuberalis and Nmur2 was expressed in the ependymal cell layer of the third ventricle. These results indicate that the species-specific expression pattern of Nmu and Nmur2 may allow NMU to have distinct functions across species, and that endogenous NMU does not function as an anorexigenic hormone in rats.
Collapse
Affiliation(s)
- Kyoka Yokogi
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| | - Yuki Goto
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| | - Mai Otsuka
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| | - Fumiya Ojima
- grid.415086.e0000 0001 1014 2000Department of Natural Sciences and Biology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192 Japan
| | - Tomoe Kobayashi
- grid.415729.c0000 0004 0377 284XDivision of Molecular Genetics, Shigei Medical Research Institute, 2117 Yamada, Minami-ku, Okayama 701-0202 Japan
| | - Yukina Tsuchiba
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| | - Yu Takeuchi
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| | - Masumi Namba
- grid.415729.c0000 0004 0377 284XDivision of Molecular Genetics, Shigei Medical Research Institute, 2117 Yamada, Minami-ku, Okayama 701-0202 Japan
| | - Mayumi Kohno
- grid.415729.c0000 0004 0377 284XDivision of Molecular Genetics, Shigei Medical Research Institute, 2117 Yamada, Minami-ku, Okayama 701-0202 Japan
| | - Minami Tetsuka
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| | - Sakae Takeuchi
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| | - Makoto Matsuyama
- grid.415729.c0000 0004 0377 284XDivision of Molecular Genetics, Shigei Medical Research Institute, 2117 Yamada, Minami-ku, Okayama 701-0202 Japan
| | - Sayaka Aizawa
- grid.261356.50000 0001 1302 4472Department of Biology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Kitaku, Okayama 700-8530 Japan
| |
Collapse
|
8
|
Yamaguchi Y, Murai I, Takeda M, Doi S, Seta T, Hanada R, Kangawa K, Okamura H, Miyake T, Doi M. <i>Nmu</i>/<i>Nms</i>/<i>Gpr176</i> Triple-Deficient Mice Show Enhanced Light-Resetting of Circadian Locomotor Activity. Biol Pharm Bull 2022; 45:1172-1179. [DOI: 10.1248/bpb.b22-00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yoshiaki Yamaguchi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Iori Murai
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Momoko Takeda
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Shotaro Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Takehito Seta
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Reiko Hanada
- Department of Neurophysiology, Faculty of Medicine, Oita University
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute
| | - Hitoshi Okamura
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Takahito Miyake
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Masao Doi
- Department of Systems Biology, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
9
|
Takayama K. Peptide Tool-Driven Functional Elucidation of Biomolecules Related to Endocrine System and Metabolism. Chem Pharm Bull (Tokyo) 2022; 70:413-419. [PMID: 35650039 DOI: 10.1248/cpb.c22-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The enhancement of basic research based on biomolecule-derived peptides has the potential to elucidate their biological function and lead to the development of new drugs. In this review, two biomolecules, namely "neuromedin U (NMU)" and "myostatin," are discussed. NMU, a neuropeptide first isolated from the porcine spinal cord, non-selectively activates two types of receptors (NMUR1 and NMUR2) and displays a variety of physiological actions, including appetite suppression. The development of receptor-selective regulators helps elucidate each receptor's detailed biological roles. A structure-activity relationship (SAR) study was conducted to achieve this purpose using the amidated C-terminal core structure of NMU for receptor activation. Through obtaining receptor-selective hexapeptide agonists, molecular functions of the core structure were clarified. Myostatin is a negative regulator of skeletal muscle growth and has attracted attention as a target for treating atrophic muscle disorders. Although the protein inhibitors, such as antibodies and receptor-decoys have been developed, the inhibition by smaller molecules, including peptides, is less advanced. Focusing on the inactivation mechanism by prodomain proteins derived from myostatin-precursor, a first mid-sized α-helical myostatin-inhibitory peptide (23-mer) was identified from the mouse sequence. The detailed SAR study based on this peptide afforded the structural requirements for effective inhibition. The subsequent computer simulation proposed the docking mode at the activin type I receptor binding site of myostatin. The resulting development of potent inhibitors suggested the existence of a more appropriate binding mode linked to their β-sheet forming properties, suggesting that further investigations might be needed.
Collapse
Affiliation(s)
- Kentaro Takayama
- Department of Environmental Biochemistry, Kyoto Pharmaceutical University
| |
Collapse
|
10
|
Qi X, Liu P, Wang Y, Xue J, An Y, Zhao C. Insights Into the Research Status of Neuromedin U: A Bibliometric and Visual Analysis From 1987 to 2021. Front Med (Lausanne) 2022; 9:773000. [PMID: 35273971 PMCID: PMC8901607 DOI: 10.3389/fmed.2022.773000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Neuromedin U (NMU) is a regulatory peptide that is widely distributed throughout the body and performs a variety of physiological functions through its corresponding receptors. In recent years, NMU has become the focus of attention in various fields of research as its diverse and essential functions have gradually been elucidated. However, there have been no bibliometrics studies on the development trend and knowledge structure of NMU research. Therefore, in this study, we used VOSviewer software to statistically analyze scientific data from articles related to NMU to track the developmental footprint of this research field, including relevant countries, institutions, authors, and keywords. We retrieved a total of 338 papers related to NMU, written by 1,661 authors from 438 organizations of 41 countries that were published in 332 journals. The first study on NMU was reported by a group in Japan in 1985. Subsequently, nine articles on NMU were published from 1987 to 2006. A small leap in this field could be detected in 2009, with 30 articles published worldwide. Among the various countries in which this research has been performed, Japan and the United States have made the most outstanding contributions. Miyazato M, Kangawa K, and Mori K from the Department of Biochemistry, National Retrain and Cardiovascular Center Research Institute in Japan were the most productive authors who have the highest number of citations. Keyword analysis showed six clusters: central-nervous-system, homeostasis, energy metabolism, cancer, immune inflammation, and food intake. The three most highly cited articles were associated with inflammation. Overall, this study demonstrates the research trends and future directions of NMU, providing an objective description of the contributions in this field along with reference value for future research.
Collapse
Affiliation(s)
- Xueping Qi
- Shanxi Medical University, Taiyuan, China
- Department of Otolaryngology, Head and Neck Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
- Key Research Laboratory of Airway Neuroimmunology, Taiyuan, China
| | | | - Yanjie Wang
- Department of Otolaryngology, Head and Neck Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
- Key Research Laboratory of Airway Neuroimmunology, Taiyuan, China
| | - Jinmei Xue
- Department of Otolaryngology, Head and Neck Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
- Key Research Laboratory of Airway Neuroimmunology, Taiyuan, China
| | - Yunfang An
- Department of Otolaryngology, Head and Neck Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
- Key Research Laboratory of Airway Neuroimmunology, Taiyuan, China
| | - Changqing Zhao
- Department of Otolaryngology, Head and Neck Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
- Key Research Laboratory of Airway Neuroimmunology, Taiyuan, China
| |
Collapse
|
11
|
McArthur E, Rinker DC, Capra JA. Quantifying the contribution of Neanderthal introgression to the heritability of complex traits. Nat Commun 2021; 12:4481. [PMID: 34294692 PMCID: PMC8298587 DOI: 10.1038/s41467-021-24582-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 06/24/2021] [Indexed: 11/15/2022] Open
Abstract
Eurasians have ~2% Neanderthal ancestry, but we lack a comprehensive understanding of the genome-wide influence of Neanderthal introgression on modern human diseases and traits. Here, we quantify the contribution of introgressed alleles to the heritability of more than 400 diverse traits. We show that genomic regions in which detectable Neanderthal ancestry remains are depleted of heritability for all traits considered, except those related to skin and hair. Introgressed variants themselves are also depleted for contributions to the heritability of most traits. However, introgressed variants shared across multiple Neanderthal populations are enriched for heritability and have consistent directions of effect on several traits with potential relevance to human adaptation to non-African environments, including hair and skin traits, autoimmunity, chronotype, bone density, lung capacity, and menopause age. Integrating our results, we propose a model in which selection against introgressed functional variation was the dominant trend (especially for cognitive traits); however, for a few traits, introgressed variants provided beneficial variation via uni-directional (e.g., lightening skin color) or bi-directional (e.g., modulating immune response) effects.
Collapse
Affiliation(s)
- Evonne McArthur
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37235, USA
| | - David C Rinker
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - John A Capra
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37235, USA.
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA.
- Bakar Computational Health Sciences Institute and Department of Epidemiology and Statistics, University of California San Francisco, San Francisco, CA, 94107, USA.
| |
Collapse
|
12
|
Neuromedin U, a Key Molecule in Metabolic Disorders. Int J Mol Sci 2021; 22:ijms22084238. [PMID: 33921859 PMCID: PMC8074168 DOI: 10.3390/ijms22084238] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
Obesity is now a public health concern. The leading cause of obesity is an energy imbalance between ingested and expended calories. The mechanisms of feeding behavior and energy metabolism are regulated by a complex of various kinds of molecules, including anorexigenic and orexigenic neuropeptides. One of these neuropeptides, neuromedin U (NMU), was isolated in the 1980s, and its specific receptors, NMUR1 and NMUR2, were defined in 2000. A series of subsequent studies has revealed many of the physiological roles of the NMU system, including in feeding behavior, energy expenditure, stress responses, circadian rhythmicity, and inflammation. Particularly over the past decades, many reports have indicated that the NMU system plays an essential and direct role in regulating body weight, feeding behavior, energy metabolism, and insulin secretion, which are tightly linked to obesity pathophysiology. Furthermore, another ligand of NMU receptors, NMS (neuromedin S), was identified in 2005. NMS has physiological functions similar to those of NMU. This review summarizes recent observations of the NMU system in relation to the pathophysiology of obesity in both the central nervous systems and the peripheral tissues.
Collapse
|
13
|
Malendowicz LK, Rucinski M. Neuromedins NMU and NMS: An Updated Overview of Their Functions. Front Endocrinol (Lausanne) 2021; 12:713961. [PMID: 34276571 PMCID: PMC8283259 DOI: 10.3389/fendo.2021.713961] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/21/2021] [Indexed: 11/19/2022] Open
Abstract
More than 35 years have passed since the identification of neuromedin U (NMU). Dozens of publications have been devoted to its physiological role in the organism, which have provided insight into its occurrence in the body, its synthesis and mechanism of action at the cellular level. Two G protein-coupled receptors (GPCRs) have been identified, with NMUR1 distributed mainly peripherally and NMUR2 predominantly centrally. Recognition of the role of NMU in the control of energy homeostasis of the body has greatly increased interest in this neuromedin. In 2005 a second, structurally related peptide, neuromedin S (NMS) was identified. The expression of NMS is more restricted, it is predominantly found in the central nervous system. In recent years, further peptides related to NMU and NMS have been identified. These are neuromedin U precursor related peptide (NURP) and neuromedin S precursor related peptide (NSRP), which also exert biological effects without acting via NMUR1, or NMUR2. This observation suggests the presence of another, as yet unrecognized receptor. Another unresolved issue within the NMU/NMS system is the differences in the effects of various NMU isoforms on diverse cell lines. It seems that development of highly specific NMUR1 and NMUR2 receptor antagonists would allow for a more detailed understanding of the mechanisms of action of NMU/NMS and related peptides in the body. They could form the basis for attempts to use such compounds in the treatment of disorders, for example, metabolic disorders, circadian rhythm, stress, etc.
Collapse
|
14
|
Anan M, Higa R, Shikano K, Shide M, Soda A, Carrasco Apolinario ME, Mori K, Shin T, Miyazato M, Mimata H, Hikida T, Hanada T, Nakao K, Kangawa K, Hanada R. Cocaine has some effect on neuromedin U expressing neurons related to the brain reward system. Heliyon 2020; 6:e03947. [PMID: 32462086 PMCID: PMC7240118 DOI: 10.1016/j.heliyon.2020.e03947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/20/2019] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Neuromedin U (NMU) is a bioactive neuropeptide, highly distributed in the gastrointestinal tract and the central nervous system. NMU has various physiological functions related to feeding behavior, energy metabolism, stress responses, circadian rhythmicity and inflammation. Recently, several reports indicate that the central NMU system plays an important role in the reward systems in the brain. However, the underlying molecular mechanisms are not yet fully defined. In this study, we found that some of cocaine-induced c-Fos immunoreactive cells were co-localized with NMU in the nucleus accumbens (NAc), caudate putamen (CPu), and basolateral amygdala (BLA), which are key brain regions associated with the brain reward system, in wild type mice. Whereas, a treatment with cocaine did not influence the kinetics of NMU or NMU receptors mRNA expression in these brain regions, and NMU-knockout mice did not show any higher preference for cocaine compared with their control mice. These results indicate that cocaine has some effect on NMU expressing neurons related to the brain reward system, and this suggests NMU system may have a role on the brain reward systems activated by cocaine.
Collapse
Affiliation(s)
- Madoka Anan
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Ryoko Higa
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kenshiro Shikano
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masahito Shide
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Akinobu Soda
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | | | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Toshitaka Shin
- Department of Urology, Faculty of Medicine, Oita University, Oita, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Hiromitsu Mimata
- Department of Urology, Faculty of Medicine, Oita University, Oita, Japan
| | - Takatoshi Hikida
- Laboratory for Advanced Brain Functions, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Toshikatsu Hanada
- Department of Cell Biology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Reiko Hanada
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
15
|
Vallöf D, Kalafateli AL, Jerlhag E. Brain region-specific neuromedin U signalling regulates alcohol-related behaviours and food intake in rodents. Addict Biol 2020; 25:e12764. [PMID: 31069918 PMCID: PMC7187236 DOI: 10.1111/adb.12764] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/21/2022]
Abstract
Albeit neuromedin U (NMU) attenuates alcohol‐mediated behaviours, its mechanisms of action are poorly defined. Providing that the behavioural effects of alcohol are processed within the nucleus accumbens (NAc) shell, anterior ventral tegmental area (aVTA), and laterodorsal tegmental area (LDTg), we assessed the involvement of NMU signalling in the aforementioned areas on alcohol‐mediated behaviours in rodents. We further examined the expression of NMU and NMU receptor 2 (NMUR2) in NAc and the dorsal striatum of high compared with low alcohol‐consuming rats, as this area is of importance in the maintenance of alcohol use disorder (AUD). Finally, we investigated the involvement of NAc shell, aVTA and LDTg in the consumption of chow and palatable peanut butter, to expand the link between NMU and reward‐related behaviours. We demonstrated here, that NMU into the NAc shell, but not aVTA or LDTg, blocked the ability of acute alcohol to cause locomotor stimulation and to induce memory retrieval of alcohol reward, as well as reduced peanut butter in mice. In addition, NMU into NAc shell decreased alcohol intake in rats. On a molecular level, we found increased NMU and decreased NMUR2 expression in the dorsal striatum in high compared with low alcohol‐consuming rats. Both aVTA and LDTg, rather than NAc shell, were identified as novel sites of action for NMU's anorexigenic properties in mice based on NMU's ability to selectively reduce chow intake when injected to these areas. Collectively, these data indicate that NMU signalling in different brain areas selectively modulates different behaviours.
Collapse
Affiliation(s)
- Daniel Vallöf
- Institute of Neuroscience and Physiology, Department of PharmacologyThe Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Aimilia Lydia Kalafateli
- Institute of Neuroscience and Physiology, Department of PharmacologyThe Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of PharmacologyThe Sahlgrenska Academy at the University of Gothenburg Gothenburg Sweden
| |
Collapse
|
16
|
Nakahara K, Maruyama K, Ensho T, Mori K, Miyazato M, Kangawa K, Uemura R, Sakoda H, Nakazato M, Murakami N. Neuromedin U suppresses prolactin secretion via dopamine neurons of the arcuate nucleus. Biochem Biophys Res Commun 2019; 521:521-526. [PMID: 31677791 DOI: 10.1016/j.bbrc.2019.10.156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 10/22/2019] [Indexed: 11/15/2022]
Abstract
Neuromedin U (NMU) has a precursor that contains one additional peptide consisting of 33 or 36 amino acid residues. Recently, we identified this second peptide from rat brain and designated it neuromedin U precursor-related peptide (NURP), showing it to stimulate prolactin release from the pituitary when injected via the intracerebroventricular (icv) route. Here, we examined whether NMU, like NURP, also stimulates prolactin release. Unlike NURP, icv injection of NMU significantly decreased the secretion of prolactin from the pituitary. This suppression of prolactin release by NMU was observed in hyper-prolactin states such as lactation, stress, pseudopregnancy, domperidone (dopamine antagonist) administration, and icv injection of NURP. Immunohistochemical analysis revealed that icv injection of NMU induced cFos expression in dopaminergic neurons of the arcuate nucleus, but not the substantia nigra. Mice with double knockout of NMU and neuromedin S (NMS), the latter also binding to NMU receptors, showed a significant increase of the plasma prolactin level after domperidone treatment relative to wild-type mice. These results suggest that NMU and NURP may play important reciprocal roles in physiological prolactin secretion.
Collapse
Affiliation(s)
- Keiko Nakahara
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Keisuke Maruyama
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Takuya Ensho
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Ryoko Uemura
- Department of Veterinary Domestic Animal Health, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan
| | - Hideyuki Sakoda
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Noboru Murakami
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki, 889-2192, Japan.
| |
Collapse
|
17
|
Garczyk S, Klotz N, Szczepanski S, Denecke B, Antonopoulos W, von Stillfried S, Knüchel R, Rose M, Dahl E. Oncogenic features of neuromedin U in breast cancer are associated with NMUR2 expression involving crosstalk with members of the WNT signaling pathway. Oncotarget 2018; 8:36246-36265. [PMID: 28423716 PMCID: PMC5482652 DOI: 10.18632/oncotarget.16121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/07/2017] [Indexed: 11/25/2022] Open
Abstract
Neuromedin U (NMU) has been shown driving the progression of various tumor entities, including breast cancer. However, the expression pattern of NMU and its receptors in breast cancer tissues as well as systematic insight into mechanisms and downstream targets of the NMU-driven signaling pathways are still elusive. Here, NMU expression was found up-regulated in all breast cancer subtypes when compared to healthy breast tissue. Using an in silico dataset comprising 1,195 samples, high NMU expression was identified as an indicator of poor outcome in breast tumors showing strong NMUR2 expression. Next, the biological impact of NMU on breast cancer cells in relation to NMUR2 expression was analyzed. Ectopic NMU expression reduced colony growth while promoting a motile phenotype in NMUR2-positive SKBR3 but not NMUR2-negative Hs578T cells. To uncover signaling pathways and key molecules affected by NMU in SKBR3 cells, Affymetrix microarray analysis was applied. Forced NMU expression affected molecules involved in WNT receptor signaling among others. As such we demonstrated enhanced activation of the WNT/planar cell polarity (PCP) effector RAC1 and down-regulation of canonical WNT targets such as MYC. In summary, NMU might contribute to progression of NMUR2-positive breast cancer representing a potential druggable target for future personalized strategies.
Collapse
Affiliation(s)
- Stefan Garczyk
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Natalie Klotz
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Sabrina Szczepanski
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Bernd Denecke
- IZKF Aachen, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Wiebke Antonopoulos
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Saskia von Stillfried
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Ruth Knüchel
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Michael Rose
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| | - Edgar Dahl
- Molecular Oncology Group, Institute of Pathology, Medical Faculty of the RWTH Aachen University, D-52074 Aachen, Germany
| |
Collapse
|
18
|
Teranishi H, Hayashi M, Higa R, Mori K, Miyazawa T, Hino J, Amano Y, Tozawa R, Ida T, Hanada T, Miyazato M, Hanada R, Kangawa K, Nakao K. Role of neuromedin U in accelerating of non-alcoholic steatohepatitis in mice. Peptides 2018; 99:134-141. [PMID: 29017855 DOI: 10.1016/j.peptides.2017.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/16/2022]
Abstract
Neuromedin U (NMU), a neuropeptide originally isolated from porcine spinal cord, has multiple physiological functions and is involved in obesity and inflammation. Excessive fat accumulation in the liver leads to non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), which is closely associated with obesity. NAFLD and NASH develop and progress via complex pathophysiological processes, and it remains unclear to what extend the NMU system contributes to the risk of obesity-related disorders such as NAFLD and NASH. Here, we demonstrate that the NMU system plays a role in NAFLD/NASH pathogenesis. In the normal mouse liver, NMU mRNA was not detectable, and expression of the mRNA encoding neuromedin U receptor 1 (NMUR1), the peripheral receptor of NMU, was low. However, the expression of both was significantly increased in the livers of NASH mice. Furthermore, overproduction of NMU induced the mouse liver by hydrodynamic injection, exacerbated NASH pathogenesis. These data indicate a novel role for the peripheral NMU system, providing new insights into the pathogenesis of NAFLD/NASH.
Collapse
Affiliation(s)
- Hitoshi Teranishi
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masafumi Hayashi
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Ryoko Higa
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Cell Biology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Takashi Miyazawa
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yuichiro Amano
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Ryuichi Tozawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takanori Ida
- Interdisciplinary Research Organization, University of Miyazaki, Miyazaki, Japan
| | - Toshikatsu Hanada
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Cell Biology, Faculty of Medicine, Oita University, Oita, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Reiko Hanada
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan.
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
19
|
Ensho T, Maruyama K, Mori K, Miyazato M, Kangawa K, Nakahara K, Murakami N. Neuromedin U precursor-related peptide (NURP) exerts neuromedin U-like sympathetic nerve action in the rat. Biochem Biophys Res Commun 2017; 492:412-418. [PMID: 28843854 DOI: 10.1016/j.bbrc.2017.08.084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 08/21/2017] [Indexed: 11/26/2022]
Abstract
It has been suggested that novel peptide that is produced from the neuromedin U (NMU) precursor may exist, as this precursor contains multiple consensus sequences for proteolytic processing. Recently, we identified two mature novel peptides comprising 33 and 36 residues in the rat brain, which were designated neuromedin U precursor-related peptide (NURP) 33 and 36. In the present study, we compared the roles of NURP33 and 36 with that of NMU, as neither activates the NMU receptors. Immunoreactivity for NMU and NURPs was widely present in the central nervous system and showed a similar distribution. Intracerebroventricular (icv) injection of NURP33 in rats increased locomotor activity, energy expenditure, heart rate and back surface temperature (BS-T), similarly to NMU or NURP36. NMU treatment reduced food intake, but NURP33 did not. Pretreatment with the β3 blocker, SR59230A, and the cyclooxygenase blocker, indomethacin, inhibited the NURP33- or NMU-induced increase of BS-T. In addition, icv injection of NURP33 or NMU increased the expression of mRNA for cyclooxygenase 2 in the hypothalamus and for uncoupling protein 1 in the brown adipose tissue. These results suggest that although NURP33 and 36 do not activate the NMU receptors, they might exert NMU-like sympathetic nerve action in the brain.
Collapse
Affiliation(s)
- Takuya Ensho
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Keisuke Maruyama
- Basic and Clinical Research Project for CNS Drugs, Medical Innovation Center, Kyoto University, Kyoto 606-8507, Japan
| | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita 565-8565, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita 565-8565, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita 565-8565, Japan
| | - Keiko Nakahara
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Noboru Murakami
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Japan.
| |
Collapse
|
20
|
Kono T, Ida T, Kawahara N, Watanabe F, Biswas G, Sato T, Mori K, Miyazato M. Identification and immunoregulatory function of neuromedin U (Nmu) in the Japanese pufferfish Takifugu rubripes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 73:246-256. [PMID: 28286258 DOI: 10.1016/j.dci.2017.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/07/2017] [Accepted: 03/07/2017] [Indexed: 06/06/2023]
Abstract
In this study, immunoregulatory function of neuromedin U (Nmu) in the teleost fish Fugu (Takifugu rubripes) was characterized. Three splicing variants of nmu mRNA encoding preproNMUs consisting of 164 (Nmu1), 139 (Nmu2), and 129 (Nmu3) amino acid residues were found in Fugu.The biologically active C-terminal region of Fugu Nmu showed high homology among fish and other vertebrate NMUs. The genomic organization of Fugu nmu differed from those of zebrafish and mammals. However, in phylogenetic analysis, Fugu Nmu formed a cluster with NMUs of other vertebrates, in addition to neuromedin S. The splicing variants of mRNA were identified in various tissues. Nmu-21 and Nmu-9 were purified as endogenous peptides from Fugu intestine. The synthetic Nmu-21 peptide activated phagocytic cells, and elevated the expression of cytokine mRNA in peripheral blood leukocytes.
Collapse
Affiliation(s)
- Tomoya Kono
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen kibanadai-nishi, Miyazaki 889-2192, Japan.
| | - Takanori Ida
- Division of Searching and Identification of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan; Division of Research & Inspection for Infectious Diseases, Center for Animal Disease Control, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Natsumi Kawahara
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen kibanadai-nishi, Miyazaki 889-2192, Japan
| | - Fumiya Watanabe
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen kibanadai-nishi, Miyazaki 889-2192, Japan
| | - Gouranga Biswas
- ICAR-Central Institute of Brackishwater Aquaculture, Kakdwip Research Centre, Kakdwip, South 24 Parganas, West Bengal 743347, India
| | - Takahiro Sato
- Molecular Genetics, Institute of Life Sciences, Kurume University, Fukuoka 839-0864, Japan
| | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka 565-8565, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka 565-8565, Japan
| |
Collapse
|
21
|
King AN, Barber AF, Smith AE, Dreyer AP, Sitaraman D, Nitabach MN, Cavanaugh DJ, Sehgal A. A Peptidergic Circuit Links the Circadian Clock to Locomotor Activity. Curr Biol 2017; 27:1915-1927.e5. [PMID: 28669757 PMCID: PMC5698909 DOI: 10.1016/j.cub.2017.05.089] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/19/2017] [Accepted: 05/30/2017] [Indexed: 01/29/2023]
Abstract
The mechanisms by which clock neurons in the Drosophila brain confer an ∼24-hr rhythm onto locomotor activity are unclear, but involve the neuropeptide diuretic hormone 44 (DH44), an ortholog of corticotropin-releasing factor. Here we identified DH44 receptor 1 as the relevant receptor for rest:activity rhythms and mapped its site of action to hugin-expressing neurons in the subesophageal zone (SEZ). We traced a circuit that extends from Dh44-expressing neurons in the pars intercerebralis (PI) through hugin+ SEZ neurons to the ventral nerve cord. Hugin neuropeptide, a neuromedin U ortholog, also regulates behavioral rhythms. The DH44 PI-Hugin SEZ circuit controls circadian locomotor activity in a daily cycle but has minimal effect on feeding rhythms, suggesting that the circadian drive to feed can be separated from circadian locomotion. These findings define a linear peptidergic circuit that links the clock to motor outputs to modulate circadian control of locomotor activity.
Collapse
Affiliation(s)
- Anna N King
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Annika F Barber
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amelia E Smith
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Austin P Dreyer
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Divya Sitaraman
- Department of Psychological Sciences, University of San Diego, San Diego, CA 92110, USA
| | - Michael N Nitabach
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA; Department of Genetics, Yale University, New Haven, CT 06520, USA; Kavli Institute for Neuroscience, Yale University, New Haven, CT 06520, USA
| | - Daniel J Cavanaugh
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Amita Sehgal
- Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Postnatal developmental of Neuromedin S and its receptor in the male Xiaomeishan pig reproductive axis. Anim Reprod Sci 2017; 181:115-124. [DOI: 10.1016/j.anireprosci.2017.03.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/31/2017] [Indexed: 11/18/2022]
|
23
|
Vallöf D, Ulenius L, Egecioglu E, Engel JA, Jerlhag E. Central administration of the anorexigenic peptide neuromedin U decreases alcohol intake and attenuates alcohol-induced reward in rodents. Addict Biol 2017; 22:640-651. [PMID: 26769653 PMCID: PMC6680249 DOI: 10.1111/adb.12355] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/26/2015] [Accepted: 12/01/2015] [Indexed: 01/24/2023]
Abstract
By investigating the neurochemical mechanisms through which alcohol activates the brain reward systems, novel treatment strategies for alcohol use disorder (AUD), a chronic relapsing disease, can be developed. In contrast to the common view of the function of gut-brain peptides, such as neuromedin U (NMU), to regulate food intake and appetite, a novel role in reinforcement mediation has been implied. The anorexigenic effects of NMU are mediated via NMU2 receptors, preferably in the arcuate nucleus and paraventricular nucleus. The expression of NMU2 receptors is also expressed in several reward-related areas in the brain, suggesting a role in reward regulation. The present experiments were therefore set up to investigate the effect of intracerebroventricular administration of NMU on alcohol-mediated behaviors in rodents. We found that central administration of NMU attenuated alcohol-induced locomotor stimulation, accumbal dopamine release and the expression of conditioned place preference in mice. In addition, NMU dose dependently decreased alcohol intake in high, but not in low, alcohol-consuming rats. Central NMU administration did not alter the blood alcohol concentrations nor change the corticosterone levels in rodents. Given that AUD is a major health-care challenge causing an enormous cost to society and novel treatment strategies are warranted, our data suggest that NMU analogues deserve to be evaluated as novel treatment of AUD in humans.
Collapse
Affiliation(s)
- Daniel Vallöf
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Lisa Ulenius
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Emil Egecioglu
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Jörgen A. Engel
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
| |
Collapse
|
24
|
Gajjar S, Patel BM. Neuromedin: An insight into its types, receptors and therapeutic opportunities. Pharmacol Rep 2017; 69:438-447. [PMID: 31994106 DOI: 10.1016/j.pharep.2017.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 12/26/2016] [Accepted: 01/18/2017] [Indexed: 12/17/2022]
Abstract
Neuropeptides are small protein used by neurons in signal communications. Neuromedin U was the first neuropeptide discovered from the porcine spinal and showed its potent constricting activities on uterus hence was entitled with neuromedin U. Following neuromedin U another of its isoform was discovered neuromedin S which was observed in suprachiasmatic nucleus hence was entitled neuromedin S. Neuromedin K and neuromedin L are of kanassin class which belong to tachykinin family. Bombesin family consists of neuromedin B and neuromedin C. All these different neuromedins have various physiological roles like constrictive effects on the smooth muscles, control of blood pressure, pain sensations, hunger, bone metastasis and release and regulation of hormones. Over the years various newer physiological roles have been observed thus opening ways for various novel therapeutic treatments. This review aims to provide an overview of important different types of neuromedin, their receptors, signal transduction mechanism and implications for various diseases.
Collapse
|
25
|
Abstract
The biological clocks of the circadian timing system coordinate cellular and physiological processes and synchronizes these with daily cycles, feeding patterns also regulates circadian clocks. The clock genes and adipocytokines show circadian rhythmicity. Dysfunction of these genes are involved in the alteration of these adipokines during the development of obesity. Food availability promotes the stimuli associated with food intake which is a circadian oscillator outside of the suprachiasmatic nucleus (SCN). Its circadian rhythm is arranged with the predictable daily mealtimes. Food anticipatory activity is mediated by a self-sustained circadian timing and its principal component is food entrained oscillator. However, the hypothalamus has a crucial role in the regulation of energy balance rather than food intake. Fatty acids or their metabolites can modulate neuronal activity by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. The timing of three-meal schedules indicates close association with the plasma levels of insulin and preceding food availability. Desynchronization between the central and peripheral clocks by altered timing of food intake and diet composition can lead to uncoupling of peripheral clocks from the central pacemaker and to the development of metabolic disorders. Metabolic dysfunction is associated with circadian disturbances at both central and peripheral levels and, eventual disruption of circadian clock functioning can lead to obesity. While CLOCK expression levels are increased with high fat diet-induced obesity, peroxisome proliferator-activated receptor (PPAR) alpha increases the transcriptional level of brain and muscle ARNT-like 1 (BMAL1) in obese subjects. Consequently, disruption of clock genes results in dyslipidemia, insulin resistance and obesity. Modifying the time of feeding alone can greatly affect body weight. Changes in the circadian clock are associated with temporal alterations in feeding behavior and increased weight gain. Thus, shift work is associated with increased risk for obesity, diabetes and cardio-vascular diseases as a result of unusual eating time and disruption of circadian rhythm.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
26
|
Vallöf D, Vestlund J, Engel JA, Jerlhag E. The Anorexigenic Peptide Neuromedin U (NMU) Attenuates Amphetamine-Induced Locomotor Stimulation, Accumbal Dopamine Release and Expression of Conditioned Place Preference in Mice. PLoS One 2016; 11:e0154477. [PMID: 27139195 PMCID: PMC4854378 DOI: 10.1371/journal.pone.0154477] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/14/2016] [Indexed: 12/28/2022] Open
Abstract
Amphetamine dependence, besides its substantial economical consequence, is a serious cause of mortality and morbidity. By investigations of the neurochemical correlates through which addictive drugs, such as amphetamine, activate the mesoaccumbal dopamine system unique targets for treatment of drug addiction can be identified. This reward link consists of a dopamine projection from the ventral tegmental area to the nucleus accumbens (NAc) suggesting that these brain areas are important for reward. The physiological function of gut-brain peptides has expanded beyond food intake modulation and involves regulation of drug reinforcement. A novel candidate for reward regulation is the anorexigenic peptide neuromedin U (NMU). We therefore investigated the effects of intracerebroventricular (icv) administration of NMU on amphetamine’s well-documented effects on the mesoaccumbal dopamine system, i.e. locomotor stimulation and accumbal dopamine release in mice. In addition, the effect of accumbal NMU administration on locomotor activity was examined. The effect of NMU, icv or intra-NAc, on the expression of conditioned place preference (CPP) was elucidated. Firstly, we showed that icv administration of NMU attenuate the amphetamine-induced locomotor stimulation, accumbal dopamine release and expression of CPP in mice. Secondly, we found that a lower dose of NMU (icv) reduce the amphetamine-induced locomotor stimulation in mice. Thirdly, we demonstrated that NMU administration into the NAc block the ability of amphetamine to cause a locomotor stimulation in mice. However, accumbal NMU administration did not attenuate the amphetamine-induced expression of CPP in mice. Our novel data suggest that central NMU signalling is involved in development of amphetamine dependence.
Collapse
Affiliation(s)
- Daniel Vallöf
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jesper Vestlund
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jörgen A Engel
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Institute of Neuroscience and Physiology, Department of Pharmacology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
27
|
Nakahara K, Akagi A, Shimizu S, Tateno S, Qattali AW, Mori K, Miyazato M, Kangawa K, Murakami N. Involvement of endogenous neuromedin U and neuromedin S in thermoregulation. Biochem Biophys Res Commun 2016; 470:930-5. [PMID: 26826380 DOI: 10.1016/j.bbrc.2016.01.155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 01/24/2016] [Indexed: 11/25/2022]
Abstract
We investigated the possible involvement of neuromedin U (NMU) and neuromedin S (NMS) in thermoregulation in rats. Intracerebroventricular (icv) injection of NMU or NMS increased the back surface temperature (BS-T) in a dose-dependent manner during both the light and dark periods. Pre-treatment with the β3 blocker SR59230A, and the cyclooxygenase blocker indomethacin, inhibited the increase in BS-T induced by NMS. Icv injection of NMS and NMU increased the expression of mRNAs for prostaglandin E synthase and cyclooxygenase 2 (COX2) in the hypothalamus, and that of mRNA for uncoupling protein 1 (UCP1) in the brown adipose tissue. Comparison of thermogenesis in terms of body temperature under normal and cold conditions revealed that NMS-KO and double-KO mice had a significantly low BS-T during the active phase, whereas NMU-KO mice did not. Exposure to low temperature decreased the BS temperature in all KO mice, but BS-T was lower in NMS-KO and double-KO mouse than in NMU-KO mice. Calorie and oxygen consumption was also significantly lower in all KO mice than in wild-type mice during the dark period. These results suggest that NMU and NMS are involved in thermoregulation via the prostaglandin E2 and β3 adrenergic receptors, but that endogenous NMS might play a more predominant role than NMU.
Collapse
Affiliation(s)
- Keiko Nakahara
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| | - Ai Akagi
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| | - Seiya Shimizu
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| | - Satoshi Tateno
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| | - Abdul Wahid Qattali
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| | - Kenji Mori
- Department of Biochemistry, National Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cardiovascular Center Research Institute, Fujishirodai 5-7-1, Suita, Osaka 565-8565, Japan
| | - Noboru Murakami
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan.
| |
Collapse
|
28
|
Martinez VG, O'Driscoll L. Neuromedin U: a multifunctional neuropeptide with pleiotropic roles. Clin Chem 2015; 61:471-82. [PMID: 25605682 DOI: 10.1373/clinchem.2014.231753] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Neuromedin U (NmU) belongs to the neuromedin family, comprising a series of neuropeptides involved in the gut-brain axis and including neuromedins B and C (bombesin-like), K (neurokinin B), L (neurokinin A or neurotensin), N, S, and U. CONTENT Although initially isolated from porcine spinal cord on the basis of their ability to induce uterine smooth muscle contraction, these peptides have now been found to be expressed in several different tissues and have been ascribed numerous functions, from appetite regulation and energy balance control to muscle contraction and tumor progression. NmU has been detected in several species to date, particularly in mammals (pig, rat, rabbit, dog, guinea pig, human), but also in amphibian, avian, and fish species. The NmU sequence is highly conserved across different species, indicating that this peptide is ancient and plays an important biological role. Here, we summarize the main structural and functional characteristics of NmU and describe its many roles, highlighting the jack-of-all-trades nature of this neuropeptide. SUMMARY NmU involvement in key processes has outlined the possibility that this neuropeptide could be a novel target for the treatment of obesity and cancer, among other disorders. Although the potential for NmU as a therapeutic target is obvious, the multiple functions of this molecule should be taken into account when designing an approach to targeting NmU and/or its receptors.
Collapse
Affiliation(s)
- Vanesa G Martinez
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
29
|
Takayama K, Mori K, Taketa K, Taguchi A, Yakushiji F, Minamino N, Miyazato M, Kangawa K, Hayashi Y. Discovery of Selective Hexapeptide Agonists to Human Neuromedin U Receptors Types 1 and 2. J Med Chem 2014; 57:6583-93. [DOI: 10.1021/jm500599s] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kentaro Takayama
- Department
of Medicinal Chemistry, Tokyo University of Pharmacy and Life Sciences, Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | - Koji Taketa
- Department
of Medicinal Chemistry, Tokyo University of Pharmacy and Life Sciences, Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Akihiro Taguchi
- Department
of Medicinal Chemistry, Tokyo University of Pharmacy and Life Sciences, Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Fumika Yakushiji
- Department
of Medicinal Chemistry, Tokyo University of Pharmacy and Life Sciences, Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | - Yoshio Hayashi
- Department
of Medicinal Chemistry, Tokyo University of Pharmacy and Life Sciences, Horinouchi, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
30
|
Aizawa S, Sakata I, Nagasaka M, Higaki Y, Sakai T. Negative regulation of neuromedin U mRNA expression in the rat pars tuberalis by melatonin. PLoS One 2013; 8:e67118. [PMID: 23843987 PMCID: PMC3699551 DOI: 10.1371/journal.pone.0067118] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/15/2013] [Indexed: 11/19/2022] Open
Abstract
The pars tuberalis (PT) is part of the anterior pituitary gland surrounding the median eminence as a thin cell layer. The characteristics of PT differ from those of the pars distalis (PD), such as cell composition and gene expression, suggesting that the PT has a unique physiological function compared to the PD. Because the PT highly expresses melatonin receptor type 1, it is considered a mediator of seasonal and/or circadian signals of melatonin. Expression of neuromedin U (NMU) that is known to regulate energy balance has been previously reported in the rat PT; however, the regulatory mechanism of NMU mRNA expression and secretion in the PT are still obscure. In this study, we examined both the diurnal change of NMU mRNA expression in the rat PT and the effects of melatonin on NMU in vivo. In situ hybridization and quantitative PCR analysis of laser microdissected PT samples revealed that NMU mRNA expression in the PT has diurnal variation that is high during the light phase and low during the dark phase. Furthermore, melatonin administration significantly suppressed NMU mRNA expression in the PT in vivo. On the other hand, 48 h fasting did not have an effect on PT-NMU mRNA expression, and the diurnal change of NMU mRNA expression was maintained. We also found the highest expression of neuromedin U receptor type 2 (NMUR2) mRNA in the third ventricle ependymal cell layer, followed by the arcuate nucleus and the spinal cord. These results suggest that NMU mRNA expression in the PT is downregulated by melatonin during the dark phase and shows diurnal change. Considering that NMU mRNA in the PT showed the highest expression level in the brain, PT-NMU may act on NMUR2 in the brain, especially in the third ventricle ependymal cell layer, with a circadian rhythm.
Collapse
Affiliation(s)
- Sayaka Aizawa
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Sakuraku, Saitama, Japan
| | - Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Sakuraku, Saitama, Japan
| | - Mai Nagasaka
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Sakuraku, Saitama, Japan
| | - Yuriko Higaki
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Sakuraku, Saitama, Japan
| | - Takafumi Sakai
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, Sakuraku, Saitama, Japan
| |
Collapse
|
31
|
Kitazawa T, Itoh K, Yaosaka N, Maruyama K, Matsuda K, Teraoka H, Kaiya H. Ghrelin does not affect gastrointestinal contractility in rainbow trout and goldfish in vitro. Gen Comp Endocrinol 2012; 178:539-45. [PMID: 22776445 DOI: 10.1016/j.ygcen.2012.06.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/24/2012] [Indexed: 12/15/2022]
Abstract
Ghrelin has been identified in rainbow trout and goldfish, and it has been shown to regulate growth hormone release and food intake in these species as seen in mammals. The aim of this study was to investigate the functional role of ghrelin in regulation of gastrointestinal contractility in both fishes. Neither rainbow trout ghrelin nor rat ghrelin affected the contractility of gastrointestinal strips of rainbow trout. Similarly, goldfish ghrelin-17 and rat ghrelin did not cause marked contraction in the goldfish intestinal bulb. Detail examinations using the goldfish intestine revealed that human neurotensin, substance-P, goldfish neuromedine-U and carbachol showed apparent contractile activities in the intestinal strips. Electrical field stimulation (EFS, 1-20 Hz) caused a frequency-dependent contraction of the intestinal bulb. Atropine partially inhibited and tetrodotoxin abolished the EFS-induced contraction. Pretreatments with goldfish ghrelin-17 and rat ghrelin did not modify the EFS-induced contraction. The mRNAs of two types of growth hormone secretagogue receptor (GHS-R), GHS-R1a-1 and GHS-R1a-2, were detected in the goldfish intestine, and the expression level of GHS-R1a-2 was 4-times higher than that of GHS-R1a-1. The expression levels of GHS-R1a-1 and GHS-R1a-2 in four regions of the goldfish intestine (intestinal bulb, intestine-1, intestine-2 and intestine-3) were almost the same. In conclusion, ghrelin does not affect gastrointestinal contractility of the rainbow trout and goldfish, although GHSR-like receptor/GHS-R1a is expressed entire intestine. These results suggest diversity of ghrelin function in vertebrates.
Collapse
Affiliation(s)
- Takio Kitazawa
- Department of Veterinary Science, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
32
|
Yang G, Su J, Yao Y, Lei Z, Zhang G, Liu Y, Liu J, Li X. Distribution of neuromedin S and its receptor NMU2R in pigs. Res Vet Sci 2012; 92:180-6. [DOI: 10.1016/j.rvsc.2010.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Revised: 11/12/2010] [Accepted: 11/23/2010] [Indexed: 11/24/2022]
|
33
|
Rahman AA, Shahid IZ, Pilowsky PM. Differential cardiorespiratory and sympathetic reflex responses to microinjection of neuromedin U in rat rostral ventrolateral medulla. J Pharmacol Exp Ther 2012; 341:213-24. [PMID: 22262923 DOI: 10.1124/jpet.111.191254] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
The rostral ventrolateral medulla (RVLM) regulates sympathetic vasomotor outflow and reflexes. Intracerebroventricular neuromedin U (NMU) increases sympathetic nerve activity (SNA), mean arterial pressure (MAP), and heart rate (HR), but the central nuclei that mediate these effects are unknown. In urethane-anesthetized, vagotomized, and artificially ventilated male Sprague-Dawley rats (n = 36) the effects of bilateral microinjection of NMU (50 nl, each side) into RVLM on cardiorespiratory variables, somatosympathetic reflex, arterial baroreflex, and chemoreflex were investigated. Microinjection of NMU into RVLM elicited a hypertension, tachycardia, and an increase in splanchnic SNA (SSNA) and lumbar SNA (LSNA) at lower doses (25 and 50 pmol). At higher dose (100 pmol), NMU caused a biphasic response, a brief hypertension and sympathoexcitation followed by prolonged hypotension and sympathoinhibition. The peak excitatory and inhibitory response was found at 100 pmol NMU with an increase in MAP, HR, SSNA, and LSNA of 36 mm Hg, 20 beats per minute, 34%, and 89%, respectively, and a decrease of 33 mm Hg, 25 beats per minute, 42%, and 52%, respectively, from baseline. NMU, in the RVLM, also increased phrenic nerve amplitude and the expiratory period and reduced the inspiratory period. NMU (100 pmol) attenuated the somatosympathetic reflex and the sympathoexcitatory and respiratory responses to hypoxia and hypercapnia. After NMU injection in RVLM, the maximum gain of the SSNA baroreflex function curve was increased, but that of the LSNA was reduced. The present study provides functional evidence for a complex differential modulatory activity of NMU on the cardiovascular and reflex responses that are integrated in the RVLM.
Collapse
Affiliation(s)
- Ahmed A Rahman
- The Australian School of Advanced Medicine, L1, F10A, Macquarie University, Sydney, NSW 2109, Australia
| | | | | |
Collapse
|
34
|
Mori M, Mori K, Ida T, Sato T, Kojima M, Miyazato M, Kangawa K. Different distribution of neuromedin S and its mRNA in the rat brain: NMS peptide is present not only in the hypothalamus as the mRNA, but also in the brainstem. Front Endocrinol (Lausanne) 2012; 3:152. [PMID: 23264767 PMCID: PMC3524995 DOI: 10.3389/fendo.2012.00152] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/16/2012] [Indexed: 11/24/2022] Open
Abstract
Neuromedin S (NMS) is a neuropeptide identified as another endogenous ligand for two orphan G protein-coupled receptors, FM-3/GPR66 and FM-4/TGR-1, which have also been identified as types 1 and 2 receptors for neuromedin U structurally related to NMS. Although expression of NMS mRNA is found mainly in the brain, spleen, and testis, the distribution of its peptide has not yet been investigated. Using a newly prepared antiserum, we developed a highly sensitive radioimmunoassay for rat NMS. NMS peptide was clearly detected in the rat brain at a concentration of 68.3 ± 3.4 fmol/g wet weight, but it was hardly detected in the spleen and testis. A high content of NMS peptide was found in the hypothalamus, midbrain, and pons-medulla oblongata, whereas abundant expression of NMS mRNA was detected only in the hypothalamus. These differing distributions of the mRNA and peptide suggest that nerve fibers originating from hypothalamic NMS neurons project into the midbrain, pons, or medulla oblongata. In addition, abundant expression of type 2 receptor mRNA was detected not only in the hypothalamus, but also in the midbrain and pons-medulla oblongata. These results suggest novel, unknown physiological roles of NMS within the brainstem.
Collapse
Affiliation(s)
- Miwa Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research InstituteOsaka, Japan
- Miwa Mori and Kenji Mori have contributed equally to this work
| | - Kenji Mori
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research InstituteOsaka, Japan
- Miwa Mori and Kenji Mori have contributed equally to this work
| | - Takanori Ida
- Interdisciplinary Research Organization, University of MiyazakiMiyazaki, Japan
| | - Takahiro Sato
- Molecular Genetics, Institute of Life Sciences, Kurume UniversityFukuoka, Japan
| | - Masayasu Kojima
- Molecular Genetics, Institute of Life Sciences, Kurume UniversityFukuoka, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research InstituteOsaka, Japan
- *Correspondence: Mikiya Miyazato, Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan. e-mail:
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research InstituteOsaka, Japan
| |
Collapse
|
35
|
Sakamoto T, Nakahara K, Maruyama K, Katayama T, Mori K, Miyazato M, Kangawa K, Murakami N. Neuromedin S regulates cardiovascular function through the sympathetic nervous system in mice. Peptides 2011; 32:1020-6. [PMID: 21356261 DOI: 10.1016/j.peptides.2011.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 02/21/2011] [Accepted: 02/21/2011] [Indexed: 11/20/2022]
Abstract
Intracerebroventricular (icv) injection of neuromedin S (NMS) in mice increased the heart rate in a dose-dependent manner. On the other hand, genetically NMS deficient mice (NMS-KO mice) exhibited a decreased heart rate and significant extension of the QRS and PR interval in the electrocardiogram complex. Although treatment with a parasympathetic nerve blocker, methylscopolamine, and a sympathetic nerve blocker, timolol, respectively increased and decreased the heart rate in both NMS-KO and wild-type mice, the extent of the decrease induced by timolol was smaller in NMS-KO than in wild-type mice. In addition, pretreatment with timolol completely inhibited the NMS-induced heart rate increase in wild-type mice. No expression of mRNA for NMS or the NMS receptor was evident in the heart by RT-PCR analysis. These results suggest that endogenous NMS may regulate cardiovascular function by activating the sympathetic nervous system.
Collapse
Affiliation(s)
- Takumi Sakamoto
- Department of Veterinary Physiology, Faculty of Agriculture, Miyazaki University, Miyazaki 889-2192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Ahnaou A, Drinkenburg WHIM. Neuromedin U(2) receptor signaling mediates alteration of sleep-wake architecture in rats. Neuropeptides 2011; 45:165-74. [PMID: 21296417 DOI: 10.1016/j.npep.2011.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 01/07/2011] [Indexed: 10/18/2022]
Abstract
Growing evidence indicates that neuromedin U (NmU) neuropeptide system plays an integral role in mediating the stress response through the corticotrophin-releasing factor (CRF) pathways. Stress is often associated with alteration in sleep-wake architecture both in human and laboratory animals. Here, we investigated whether activation of the NmU₂ receptor, a major high affinity receptor for NmU predominantly expressed in the brain, affects sleep behavior in rats. Effects of single (acute) intracebroventricular (icv) infusion of 2.5 nmol of the full agonists porcine NmU8 and rat NmU23 were assessed on sleep-wake architecture in freely moving rats, which were chronically implanted with EEG and EMG electrodes. In addition, repeated once daily administration of NmU8 at 2.5 nmol during 8 consecutive days (sub-chronic) was studied. Acute icv infusion of NmU23 elicited a robust alteration in sleep-wake architecture, namely enhanced wakefulness and suppressed sleep during the first 4h after administration. Acute infusion NmU8 had no effect on spontaneous sleep-wake architecture. However, sub-chronic icv infusion of NmU8 increased the amount of rapid eye movement (REM) sleep and intermediate stage (IS), while decreased light sleep. Additionally, NmU8 increased transitions from sleep states towards wakefulness suggesting a disruption in sleep continuity. The present results show that central-activation of NmU₂ receptor markedly reduced sleep duration and disrupted the mechanisms underlying NREM-REM sleep transitions. Given that sleep-wakefulness cycle is strongly influenced by stress and the role of NmU/NmU₂ receptor signaling in stress response, the disruption in sleep pattern associated with peptides species may support at least some signs of stress.
Collapse
Affiliation(s)
- A Ahnaou
- Janssen Pharmaceutical Companies of Johnson & Johnson, Dept. of Neurosciences, A Division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium.
| | | |
Collapse
|
37
|
Maruyama K, Kaiya H, Miyazato M, Konno N, Wakasugi T, Uchiyama M, Shioda S, Murakami N, Matsuda K. Isolation and characterisation of two cDNAs encoding the neuromedin U receptor from goldfish brain. J Neuroendocrinol 2011; 23:282-91. [PMID: 21182546 DOI: 10.1111/j.1365-2826.2010.02106.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intracerebroventricular administration of neuromedin U (NMU) exerts an anorexigenic effect in a goldfish model. However, little is known about the NMU receptor and its signalling system in fish. In the present study, we isolated and cloned two cDNAs encoding different proteins comprising 429 and 388 amino acid residues from the goldfish brain based on the nucleotide sequences of human NMU receptor 1 (NMU-R1) and receptor 2 (NMU-R2). Hydropathy and phylogenetic analyses suggested that these two proteins were orthologues of NMU-R1 and -R2 of goldfish. We established two human embryonic kidney 293 cell lines stably expressing putative NMU-R1 and -R2, respectively, and showed that NMU induced an increase in intracellular calcium concentration ([Ca(2+)](i)) in these cells. We examined the presence of NMU-R1 and -R2 in the goldfish brain by western blotting analysis using affinity-purified antisera raised against peptide fragments derived from these receptors. NMU-R1-specific and NMU-R2-specific antisera detected a 49-kDa and 45-kDa immunopositive bands, respectively, in the brain extract. The mass of each band corresponded to that of the deduced respective primary structures. Reverse transcriptase-polymerase chain reaction analysis showed that NMU-R1 and -R2 transcripts were detected in several tissues. In particular, both mRNAs were strongly expressed in the goldfish brain. By contrast, NMU-R2 mRNA was also expressed in the gut. These results indicate for the first time that NMU-R orthologues exist in goldfish, and suggest physiological roles of NMU and its receptor system in fish.
Collapse
Affiliation(s)
- K Maruyama
- Laboratory of Regulatory Biology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Budhiraja S, Chugh A. Neuromedin U: physiology, pharmacology and therapeutic potential. Fundam Clin Pharmacol 2009; 23:149-57. [PMID: 19645813 DOI: 10.1111/j.1472-8206.2009.00667.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neuromedin U (NmU), a multifunctional neuropeptide, belongs to a family of neuropeptides, the neuromedins. It is ubiquitously distributed with highest levels found in the gastrointestinal tract and pituitary. The conservation of structural elements of NmU across species, the widespread distribution of NmU and its receptors throughout the body point to a fundamental role in key physiological processes. Two G protein coupled receptors for NmU have been cloned NmU R1 and NmU R2. NmU R1 is expressed pre-dominantly in the periphery especially the gastrointestinal tract whereas NmU R2 is expressed pre-dominantly in the central nervous system. Current evidence suggests a role of NmU in pain, in regulation of feeding and energy homeostasis, stress, cancer, immune mediated inflammatory diseases like asthma, inflammatory diseases, maintaining the biological clock, in the regulation of smooth muscle contraction in the gastrointestinal and genitourinary tract, and in the control of blood flow and blood pressure. With the development of drugs selectively acting on receptors and knockout animal models, exact pathophysiological roles of NmU will become clearer.
Collapse
Affiliation(s)
- S Budhiraja
- Department of Pharmacology, Pt. B. D. Sharma, PGIMS, Rohtak-124001, Haryana, India.
| | | |
Collapse
|
39
|
Yayou K, Kitagawa S, Ito S, Kasuya E, Sutoh M. Effects of intracerebroventricular administration of neuromedin U or neuromedin S in steers. Gen Comp Endocrinol 2009; 163:324-8. [PMID: 19442664 DOI: 10.1016/j.ygcen.2009.04.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 04/24/2009] [Accepted: 04/29/2009] [Indexed: 11/26/2022]
Abstract
Although neuromedin U (NMU) and neuromedin S (NMS) are reported to modulate stress responses mainly through corticotropin-releasing hormone system in rodents, the in vivo effects of centrally administered NMU or NMS on stress regulation have not been fully elucidated in cattle. We examined adrenocorticotropic hormone levels, body temperature, and behavioral responses to intracerebroventricularly (ICV) administered rat NMU or rat NMS in steers. ICV NMU and NMS (0.2, 2, and 20 nmol/200 microl) evoked a dose-related increase in plasma cortisol concentrations (CORT). There was a significant time-treatment interaction for the time course of CORT (p<0.001). ICV NMU evoked a dose-related increase in rectal temperature (RT). There was a significant time-treatment interaction for the change in RT from pre-injection value (p<0.05). There was a significant difference among treatments in the percentage of time spent lying (Friedman's test, chi(2)=15.6, p<0.01) and in the total number of head shaking (Friedman's test, chi(2)=14.49, p<0.01). A high dose of NMS tended to shorten the duration of lying and increase the number of head shaking. These findings indicate that both central NMU and NMS might participate in controlling the hypothalamo-pituitary-adrenal axis, that central NMU might participate in controlling body temperature, and that central NMS is likely to be involved in behavioral activation in cattle.
Collapse
Affiliation(s)
- K Yayou
- Laboratory of Neurobiology, National Institute of Agrobiological Sciences, Tsukuba 305-8602, Japan.
| | | | | | | | | |
Collapse
|
40
|
Abstract
Neuromedin U (NMU) is known to have potent actions on appetite and energy expenditure. Deletion of the NMU gene in mice leads to an obese phenotype, characterized by hyperphagia and decreased energy expenditure. Conversely, transgenic mice that overexpress proNMU exhibit reduced body weight and fat storage. Here, we show that central administration of NMU or the related peptide neuromedin S (NMS) dose-dependently decreases food intake, increases metabolic rate, and leads to significant weight loss in mice. The effects of NMU and NMS on both feeding and metabolism are almost completely lost in mice lacking the putative CNS receptor for NMU and NMS, NMUr2. However, NMUr2 knockout mice do not exhibit overt differences in body weight or energy expenditure compared with wild-type mice, suggesting that the dramatic phenotype of the NMU gene knockout mouse is not due simply to the loss of NMU/NMUr2 signaling. Putative proteolytic cleavage sites indicate that an additional peptide is produced from the NMU precursor protein, which is extremely well conserved between human, mouse, and rat. Here, we demonstrate that this peptide, proNMU(104-136), has a pronounced effect on energy balance in mice. Specifically, central administration of proNMU(104-136) causes a significant but transient ( approximately 4 h) increase in feeding, yet both food intake and body weight are decreased over the following 24 h. proNMU(104-136) administration also significantly increased metabolic rate. These results suggest that proNMU(104-136) is a novel modulator of energy balance and may contribute to the phenotype exhibited by NMU knockout mice.
Collapse
Affiliation(s)
- David A Bechtold
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | |
Collapse
|
41
|
Affiliation(s)
- Colleen M Novak
- Mayo Clinic, Endocrine Research Unit, 200 1st Street Southwest, Saint Marys Hospital, Joseph 5-194, Rochester, Minnesota 55901, USA.
| |
Collapse
|
42
|
Mitchell JD, Maguire JJ, Davenport AP. Emerging pharmacology and physiology of neuromedin U and the structurally related peptide neuromedin S. Br J Pharmacol 2009; 158:87-103. [PMID: 19519756 DOI: 10.1111/j.1476-5381.2009.00252.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Neuromedin U (NMU) has been paired with the G-protein-coupled receptors (GPRs) NMU(1) (formerly designated as the orphan GPR66 or FM-3) and NMU(2) (FM-4 or hTGR-1). Recently, a structurally related peptide, neuromedin S (NMS), which shares an amidated C-terminal heptapeptide motif, has been identified in both rat and human, and has been proposed as a second ligand for these receptors. Messenger RNA encoding NMU receptor subtypes shows differential expression: NMU(1) is predominantly expressed in peripheral tissues, particularly the gastrointestinal tract, whereas NMU(2) is abundant within the brain and spinal cord. NMU peptide parallels receptor distribution with highest expression in the gastrointestinal tract and specific structures within the brain, reflecting its major role in the regulation of energy balance. The NMU knockout mouse has an obese phenotype and, in agreement, the Arg165Trp amino acid variant of NMU-25 in humans, which is functionally inactive, co-segregated with childhood-onset obesity. Emerging physiological roles for NMU include vasoconstriction mediated predominantly via NMU(1) with nociception and bone remodelling via NMU(2). The NMU system has also been implicated in the pathogenesis of septic shock and cancers including bladder carcinoma and acute myeloid leukaemia. Intriguingly, NMS is more potent at NMU(2) receptors in vivo where it has similar central actions in suppression of feeding and regulation of circadian rhythms to NMU. Taken together with its vascular actions, NMU may be a functional link between energy balance and the cardiovascular system and may provide a future target for therapies directed against the disorders that comprise metabolic syndrome.
Collapse
Affiliation(s)
- J D Mitchell
- Clinical Pharmacology Unit, University of Cambridge, Level 6 Centre for Clinical Investigation, Cambridge, UK
| | | | | |
Collapse
|
43
|
Mangold C, Ksiazek I, Yun SW, Berger E, Binkert C. Distribution of neuromedin U binding sites in the rat CNS revealed by in vitro receptor autoradiography. Neuropeptides 2008; 42:377-86. [PMID: 18547640 DOI: 10.1016/j.npep.2008.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 04/28/2008] [Accepted: 04/29/2008] [Indexed: 10/22/2022]
Abstract
Neuromedin U (NMU), a neuropeptide implicated in feeding, inflammation, pain control and anxiety-related behaviours, is widely distributed in peripheral organs and the CNS. These effects are thought to be mediated by its receptors NMU(1) and NMU(2). Since its precise sites of interaction in the CNS were to date unknown, we studied the distribution of in vitro binding sites for (125)I-NMU-23 in the rat CNS by receptor autoradiography. High-density specific binding was found in discrete areas of the brain and spinal cord, namely in the limbic system (hippocampal formation, septohippocampal nucleus, indusium griseum, hypothalamus, amygdaloid nuclei), superior colliculus, dorsal raphé, and substantia gelatinosa of the spinal cord. Our findings provide further supportive evidence for a multifunctional role for the peptide in the brain and spinal cord.
Collapse
Affiliation(s)
- C Mangold
- Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | | | | | | | | |
Collapse
|
44
|
Kosiol C, Vinař T, da Fonseca RR, Hubisz MJ, Bustamante CD, Nielsen R, Siepel A. Patterns of positive selection in six Mammalian genomes. PLoS Genet 2008; 4:e1000144. [PMID: 18670650 PMCID: PMC2483296 DOI: 10.1371/journal.pgen.1000144] [Citation(s) in RCA: 426] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 06/27/2008] [Indexed: 01/28/2023] Open
Abstract
Genome-wide scans for positively selected genes (PSGs) in mammals have provided insight into the dynamics of genome evolution, the genetic basis of differences between species, and the functions of individual genes. However, previous scans have been limited in power and accuracy owing to small numbers of available genomes. Here we present the most comprehensive examination of mammalian PSGs to date, using the six high-coverage genome assemblies now available for eutherian mammals. The increased phylogenetic depth of this dataset results in substantially improved statistical power, and permits several new lineage- and clade-specific tests to be applied. Of approximately 16,500 human genes with high-confidence orthologs in at least two other species, 400 genes showed significant evidence of positive selection (FDR<0.05), according to a standard likelihood ratio test. An additional 144 genes showed evidence of positive selection on particular lineages or clades. As in previous studies, the identified PSGs were enriched for roles in defense/immunity, chemosensory perception, and reproduction, but enrichments were also evident for more specific functions, such as complement-mediated immunity and taste perception. Several pathways were strongly enriched for PSGs, suggesting possible co-evolution of interacting genes. A novel Bayesian analysis of the possible "selection histories" of each gene indicated that most PSGs have switched multiple times between positive selection and nonselection, suggesting that positive selection is often episodic. A detailed analysis of Affymetrix exon array data indicated that PSGs are expressed at significantly lower levels, and in a more tissue-specific manner, than non-PSGs. Genes that are specifically expressed in the spleen, testes, liver, and breast are significantly enriched for PSGs, but no evidence was found for an enrichment for PSGs among brain-specific genes. This study provides additional evidence for widespread positive selection in mammalian evolution and new genome-wide insights into the functional implications of positive selection.
Collapse
Affiliation(s)
- Carolin Kosiol
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Tomáš Vinař
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | | | - Melissa J. Hubisz
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
| | - Carlos D. Bustamante
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Rasmus Nielsen
- Institute of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Adam Siepel
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
45
|
Brighton PJ, Wise A, Dass NB, Willars GB. Paradoxical behavior of neuromedin U in isolated smooth muscle cells and intact tissue. J Pharmacol Exp Ther 2008; 325:154-64. [PMID: 18180374 DOI: 10.1124/jpet.107.132803] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neuromedin U (NmU) is a neuropeptide showing high levels of structural conservation across different species. Since its discovery in 1985, NmU has been implicated in numerous physiological roles, including smooth muscle contraction, energy homeostasis, stress, intestinal ion transport, pronociception, and circadian rhythm. Two G-protein-coupled receptors have been identified for NmU and cloned from humans, rats, and mice. Recombinantly expressed NmU receptors couple to both Galpha(q/11) and Galpha(i) G-proteins, and NmU binds essentially irreversibly, preventing signaling to repetitive applications of NmU. However, it is unclear whether these properties reflect those of endogenously expressed NmU receptors or how these properties influence the functional consequences of NmU receptor signaling. Here, we have explored the signaling by rat NmU receptors expressed endogenously in cultured rat colonic smooth muscle cells and explore the functional consequence of this signaling by investigating the NmU-mediated contraction of ex vivo rat colonic smooth muscle preparations. We demonstrate that endogenous rat NmU receptors couple to both Galpha(q/11) and Galpha(i) G-proteins. Furthermore, we show complex patterns of Ca(2+) signaling, including oscillations, and provide evidence of essentially irreversible binding of NmU to smooth muscle cells. Challenge of either circular or longitudinal rat isolated colonic smooth muscle preparations with NmU resulted in robust contractions. Stimulation was direct, and paradoxically, repetitive applications of NmU mediated repetitive contractions with no evidence of desensitization, highlighting a major discrepancy in the behavior of NmU in single cells and in intact tissues. The reason for this discrepancy is presently unknown.
Collapse
Affiliation(s)
- Paul J Brighton
- Department of Cell Physiology and Pharmacology, Medical Sciences Building, University of Leicester, University Road, Leicester LE1 9HN, UK
| | | | | | | |
Collapse
|
46
|
Kotz CM, Teske JA, Billington CJ. Neuroregulation of nonexercise activity thermogenesis and obesity resistance. Am J Physiol Regul Integr Comp Physiol 2008; 294:R699-710. [PMID: 18160530 DOI: 10.1152/ajpregu.00095.2007] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
High levels of spontaneous physical activity in lean people and the nonexercise activity thermogenesis (NEAT) derived from that activity appear to protect lean people from obesity during caloric challenge, while obesity in humans is characterized by dramatically reduced spontaneous physical activity. We have similarly demonstrated that obesity-resistant rats have significantly greater spontaneous physical activity than obesity-prone rats, and that spontaneous physical activity predicts body weight gain. Although the energetic cost of activity varies between types of activity and may be regulated, individual level of spontaneous physical activity is important in determining propensity for obesity. We review the current status of knowledge about the brain mechanisms involved in controlling the level of spontaneous physical activity and the NEAT so generated. Focus is on potential neural mediators of spontaneous physical activity and NEAT, including orexin A (also known as hypocretin 1), agouti-related protein, ghrelin, and neuromedin U, in addition to brief mention of neuropeptide Y, corticotrophin releasing hormone, cholecystokinin, estrogen, leptin, and dopamine effects on spontaneous physical activity. We further review evidence that strain differences in orexin stimulation pathways for spontaneous physical activity and NEAT appear to track with the body weight phenotype, thus providing a potential mechanistic explanation for reduced activity and weight gain.
Collapse
Affiliation(s)
- Catherine M Kotz
- Veterans Affairs Medical Center, GRECC (11G) One Veterans Drive, Minneapolis, MN 55417, USA.
| | | | | |
Collapse
|
47
|
Abstract
Neuromedin S, a novel neuropeptide of 36 amino acids, was isolated from rat brain as an endogenous ligand for the orphan G protein-coupled receptors FM-3/GPR66 and FM-4/TGR-1, identified to date as type-1 and type-2 neuromedin U (NMU) receptors, respectively. The peptide was designated neuromedin S (NMS) because it is specifically expressed in the suprachiasmatic nucleus of the hypothalamus. NMS is structurally related to NMU; these peptides share a C-terminal core structure. In this review, we will outline the recent discoveries regarding the structure, cognate receptors, distribution, and possible physiological functions of NMS.
Collapse
|
48
|
Jászberényi M, Bagosi Z, Thurzó B, Földesi I, Telegdy G. Endocrine and behavioral effects of neuromedin S. Horm Behav 2007; 52:631-9. [PMID: 17900576 DOI: 10.1016/j.yhbeh.2007.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 07/13/2007] [Accepted: 07/30/2007] [Indexed: 11/28/2022]
Abstract
The present experiments focused on the effects of neuromedin S on hypothalamic-pituitary-adrenal (HPA) activation and behavior. The peptide (0.25-1 nmol) was administered intracerebroventricularly to rats, the behavior of which was monitored by means of telemetry, open field observations and an elevated plus-maze (EPM) test. Autonomic functions such as the temperature and the heart rate were recorded by telemetry. The action on the HPA axis was assessed via measurements of the plasma corticosterone and ACTH levels. To reveal the transmission of the endocrine responses, animals were pretreated with corticotrophin releasing hormone receptor (CRHR) antagonists (1 nmol). In the open field test, the animals were pretreated with either a CRHR(1) antagonist (antalarmin) or haloperidol (10 microg/kg), while in the EPM test they were pretreated with antalarmin or diazepam (1 mg/kg). The dopamine release from striatal and amygdala slices after peptide treatment was measured with a superfusion apparatus. Neuromedin S exerted dose-dependent effects on the HPA system, which were inhibited by antalarmin. It also activated grooming and decreased the entries to and time spent in the open arms during the EPM test. The grooming response was abolished by haloperidol and antalarmin pretreatment, while diazepam and antalarmin showed a tendency to attenuate the response evoked in the EPM test. In the superfusion studies, neuromedin S enhanced the dopamine release from the amygdala slices. These results demonstrate that neuromedin S stimulates the HPA axis through the CRHR(1) pathway and evokes stereotyped behavior and anxiety through mesolimbic dopamine and corticotrophin releasing hormone release.
Collapse
Affiliation(s)
- Miklós Jászberényi
- Department of Pathophysiology, University of Szeged, H-6701, Szeged, Semmelweis u. 1., PO Box 427, Hungary
| | | | | | | | | |
Collapse
|
49
|
Vigo E, Roa J, Pineda R, Castellano JM, Navarro VM, Aguilar E, Pinilla L, Tena-Sempere M. Novel role of the anorexigenic peptide neuromedin U in the control of LH secretion and its regulation by gonadal hormones and photoperiod. Am J Physiol Endocrinol Metab 2007; 293:E1265-73. [PMID: 17726140 DOI: 10.1152/ajpendo.00425.2007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuromedin U (NMU) is a widely spread neuropeptide, with predominant expression at the gastrointestinal tract and brain, putatively involved in the regulation of a diversity of biological functions, including food intake, energy balance and circadian rhythms; all closely related to reproduction. Yet, the implication of NMU in the control of the gonadotropic axis remains scarcely studied. We report herein analyses on the hypothalamic expression and function of NMU in different physiological and experimental states of the rat reproductive system. Expression of NMU mRNA at the hypothalamus was persistently detected along female postnatal development, with maximum levels in adulthood that fluctuated across the cycle and were modulated by ovarian steroids. Acute central administration of NMU evoked increases of serum LH levels in pubertal female rats, while repeated injection of NMU tended to advance vaginal opening. Likewise, central injection of NMU increased serum LH concentrations in cycling female rats, with peak responses in estrus. In contrast, NMU significantly inhibited preelevated LH secretion in gonadectomized and kisspeptin-treated rats. Finally, in noncycling females due to photoperiodic manipulation (constant light), hypothalamic NMU mRNA levels were markedly depressed, but relative LH responses to exogenous NMU were significantly augmented. All together, our present data support a predominant stimulatory role of NMU in the control of the female gonadotropic axis, which appears under the influence of developmental, hormonal, and photoperiodic cues, and might contribute to the joint regulation of energy balance, biological rhythms, and reproduction.
Collapse
Affiliation(s)
- Eva Vigo
- Physiology Section, Department of Cell Biology, Physiology and Immunology. Faculty of Medicine, University of Córdoba, Avda Menéndez Pidal s/n, 14004, Cordoba, Spain
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Novak CM, Zhang M, Levine JA. Sensitivity of the hypothalamic paraventricular nucleus to the locomotor-activating effects of neuromedin U in obesity. Brain Res 2007; 1169:57-68. [PMID: 17706946 PMCID: PMC2735201 DOI: 10.1016/j.brainres.2007.06.055] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 06/15/2007] [Accepted: 06/25/2007] [Indexed: 11/25/2022]
Abstract
Obesity is associated with a decrease in energy expenditure relative to energy intake. The decrease in physical activity associated with obesity in several species, including humans, contributes to decreased energy expenditure. Several hormones and neuropeptides that affect appetite also modulate physical activity, including neuromedin U (NMU), a peptide found in the gut and brain. We have demonstrated that NMU microinjected into the hypothalamic paraventricular nucleus (PVN) in rats increases the energy expenditure associated with physical activity, called non-exercise activity thermogenesis (NEAT). Here we examined whether obesity in rats is related to decreased sensitivity of the PVN to the locomotor-activating effect of NMU. Diet-induced obese (DIO) rats and lean, diet-resistant (DR) rats were given PVN microinjections of increasing doses of NMU both before and after 1 month on a high-fat diet. We found that NMU increases physical activity, energy expenditure, and NEAT in a dose-dependent manner in both DR and DIO rats, both before and after 1 month on the high-fat diet. Before high-fat feeding, the obesity-prone and lean rats showed similar levels of physical activity after intra-PVN microinjections of NMU. After 1 month of the high-fat diet, however, the obesity-resistant rats showed significantly more NMU-induced physical activity compared to the obese DIO rats. Taken together with previous studies, these results suggest that obesity may represent a state associated with decreased central sensitivity to neuropeptides such as NMU that increase physical activity and therefore energy expenditure.
Collapse
Affiliation(s)
- Colleen M Novak
- Mayo Clinic, Endocrine Research Unit, St Marys Hospital, Joseph 5-194, 200 1st St. SW, Rochester, MN 55905, USA.
| | | | | |
Collapse
|