594 results on '"Neuropeptide FF"'
Search Results
2. Neuropeptide FF Promotes Neuronal Survival and Enhances Synaptic Protein Expression Following Ischemic Injury.
- Author
-
Choi, In-Ae, Yun, Ji Hee, Lee, Jongmin, and Choi, Dong-Hee
- Subjects
- *
NEUROPLASTICITY , *RECOMBINANT proteins , *ISCHEMIC stroke , *SIRTUINS , *LACTATE dehydrogenase , *PEROXISOME proliferator-activated receptors - Abstract
This study explores the neuroprotective effects of neuropeptide FF (NPFF, FLFQPQRFamide) in the context of ischemic injury. Based on transcriptomic analysis in stroke models treated with 5-Aza-dC and task-specific training, we identified significant gene expression changes, particularly involving NPFF. To further explore NPFF's role in promoting neuronal recovery, recombinant NPFF protein (rNPFF) was used in primary mixed cortical cultures subjected to oxygen-glucose deprivation and reoxygenation. Our results demonstrated that rNPFF significantly reduced lactate dehydrogenase release, indicating decreased cellular damage. It also significantly increased the expression of TUJ1 and MAP2, markers of neuronal survival and dendritic integrity. Additionally, rNPFF significantly upregulated key synaptic proteins, including GAP43, PSD95, and synaptophysin, which are essential for synaptic repair and plasticity. Post-injury rNPFF treatment led to a significant upregulation of pro-brain-derived neurotrophic factor (BDNF) and mature BDNF, which play critical roles in neuronal survival, growth, and synaptic plasticity. Moreover, rNPFF activated the protein kinase Cε isoform, Sirtuin 1, and peroxisome proliferator-activated receptor gamma pathways, which are crucial for regulating cellular stress responses, synaptic plasticity, and energy homeostasis, further promoting neuronal survival and recovery. These findings suggest that rNPFF may play a pivotal role in enhancing neuronal survival and synaptic plasticity after ischemic injury, highlighting its potential as a therapeutic target for stroke recovery. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Renal autocrine neuropeptide FF (NPFF) signaling regulates blood pressure
- Author
-
Hewang Lee, Bibhas Amatya, Van Anthony M. Villar, Laureano D. Asico, Jin Kwon Jeong, Jun Feranil, Shaun C. Moore, Xiaoxu Zheng, Michael Bishop, Jerald P. Gomes, Jacob Polzin, Noah Smeriglio, Pedro A. S. Vaz de Castro, Ines Armando, Robin A. Felder, Ling Hao, and Pedro A. Jose
- Subjects
Blood pressure ,Brain ,Dopamine D1-like receptor ,Kidney ,Neuropeptide FF ,Medicine ,Science - Abstract
Abstract The kidney and brain play critical roles in the regulation of blood pressure. Neuropeptide FF (NPFF), originally isolated from the bovine brain, has been suggested to contribute to the pathogenesis of hypertension. However, the roles of NPFF and its receptors, NPFF-R1 and NPFF-R2, in the regulation of blood pressure, via the kidney, are not known. In this study, we found that the transcripts and proteins of NPFF and its receptors, NPFF-R1 and NPFF-R2, were expressed in mouse and human renal proximal tubules (RPTs). In mouse RPT cells (RPTCs), NPFF, but not RF-amide-related peptide-2 (RFRP-2), decreased the forskolin-stimulated cAMP production in a concentration- and time-dependent manner. Furthermore, dopamine D1-like receptors colocalized and co-immunoprecipitated with NPFF-R1 and NPFF-R2 in human RPTCs. The increase in cAMP production in human RPTCs caused by fenoldopam, a D1-like receptor agonist, was attenuated by NPFF, indicating an antagonistic interaction between NPFF and D1-like receptors. The renal subcapsular infusion of NPFF in C57BL/6 mice decreased renal sodium excretion and increased blood pressure. The NPFF-mediated increase in blood pressure was prevented by RF-9, an antagonist of NPFF receptors. Taken together, our findings suggest that autocrine NPFF and its receptors in the kidney regulate blood pressure, but the mechanisms remain to be determined.
- Published
- 2024
- Full Text
- View/download PDF
4. Hypothalamic NPFFR2 attenuates central insulin signaling and its knockout diminishes metabolic dysfunction in mouse models of diabetes mellitus.
- Author
-
Lin, Ya-Tin, Wu, Kuan-Hsuan, Jhang, Jie-Jhu, Jhang, Jie-Lan, Yu, Zachary, Tsai, Sze-Chi, Chen, Jin-Chung, Hsu, Po-Hung, and Li, Hui-Yun
- Abstract
The hypothalamus is a crucial brain region that mediates the effects of insulin and leptin signals on peripheral metabolic functions. Previous research has shown that insulin signals in the hypothalamus act via multiple neuronal circuits and anabolic/catabolic pathways that converge on the vagus nerve and sympathetic fibers to coordinate energy metabolism in peripheral organs. Additionally, neuropeptide FF (NPFF) has been identified as a regulator of feeding behaviors and energy homeostasis in the hypothalamus, but the mechanisms underlying its involvement in metabolic control remain unclear. This study aims to explore the underlying mechanisms of NPFF in modulating metabolic disorders. In this study, we investigated the physiological role of NPFF in insulin-related energy homeostasis and metabolic health. First, we evaluated the effects of NPFF and its receptors on central insulin signaling using mouse hypothalamic cell lines and Npffr2 -overexpressing mice. To further explore the effects of NPFFR2 on insulin-related metabolic disorders, such as diabetes mellitus, we used Npffr2 -deleted mice in combination with the streptozotocin (STZ)-induced type 1 diabetes and high-fat diet/STZ-induced type 2 diabetic mouse models. The impacts of central NPFFR2 were demonstrated specifically through Npffr2 overexpression in the hypothalamic arcuate nucleus, which subsequently induced type 2 diabetes. We found that stimulating NPFFR2 in the hypothalamus blocked hypothalamic insulin activity. Npffr2 deletion improved central and peripheral metabolic symptoms in both mouse models of diabetes mellitus, exerting effects on central and systemic insulin resistance, feeding behaviors, glucose and insulin intolerance, lipid metabolism, liver steatosis, and inflammation of white adipose tissues. The overexpression of ARC Npffr2 augmented the metabolic dysregulation in the mouse model of type 2 diabetes. Our findings demonstrate that hypothalamic NPFFR2 negatively regulates insulin signaling in the central nervous system and plays an important role in maintaining systemic metabolic health, thereby providing valuable insights for potential clinical interventions targeting these health challenges. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Neuropeptide FF prevented histamine- or chloroquine-induced acute itch behavior through non-NPFF receptors mechanism in male mice.
- Author
-
Tang, Honghai, Zhang, Ting, Feng, Jiamin, Zhang, Mengna, Xu, Biao, Zhang, Qinqin, Li, Ning, Zhang, Nan, and Fang, Quan
- Abstract
The neuropeptide FF (NPFF) system regulates various physiological and pharmacological functions, particularly pain modulation. However, the modulatory effect of NPFF system on itch remains unclear. To investigate the modulatory effect and functional mechanism induced by NPFF system on acute itch, we examined the effects of supraspinal administration of NPFF and related peptides on acute itch induced by intradermal (i.d.) injection of histamine or chloroquine in male mice. Our results indicated that intracerebroventricular (i.c.v.) administration of NPFF dose-dependently prevented histamine- or chloroquine-induced acute itch behaviors. In addition, the modulatory effect of NPFF was not affected by the selective NPFF receptor antagonist RF9. Furthermore, we investigated the effects of NPVF and dNPA, the selective agonists of NPFF 1 and NPFF 2 receptors respectively, on the acute itch. The results demonstrated that both NPFF agonists effectively prevented acute itch induced by histamine or chloroquine in a manner similar to NPFF, and their effects were also not modified by RF9. To further investigate the possible mechanism of the NPFF receptors agonists, the NPFF-derived analogues [Phg
8 ]-NPFF and NPFF(1–7)-NH 2 that could not activate NPFF receptors in cAMP assays were subsequently tested in the acute itch model. Interestingly, [Phg8 ]-NPFF, but not NPFF(1–7)-NH 2 , prevented acute itch behavior after i.c.v. administration. In conclusion, our findings reveal that NPFF and related peptides prevent histamine- and chloroquine-induced acute itch through a NPFF receptor-independent mechanism. And it was revealed that the C-terminal phenyl structure of NPFF may play a crucial role in these modulatory effects on acute itch. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
6. Neuropeptide FF indirectly affects testicular morphogenesis and functions in medaka.
- Author
-
Soma Tomihara, Kana Ikegami, Rinko Shimomai, and Chie Umatani
- Subjects
- *
TESTIS physiology , *ORYZIAS latipes , *ENDOCRINE system , *FOLLICLE-stimulating hormone , *SPERMATOGENESIS - Abstract
Testicular morphogenesis and functions are considered to be under the control of neural and endocrine systems. However, the available literature is mainly limited to mammals, and it remains unclear how they are regulated in teleost species. Here, we demonstrated that neuropeptide FF (NPFF) in the brain is responsible for the follicle-stimulating hormone expression in the pituitary, which facilitates the testicular morphogenesis and androgen synthesis, and subsequently contributes to successful spermatogenesis. The present findings give us important insights into the neuroendocrine regulatory mechanisms underlying the testicular morphogenesis and functions in teleosts. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Neuropeptide FF-related gene in fish (Larimichthys polyactis): identification, characterization, and potential anti-inflammatory function.
- Author
-
Xiao, Ran, Guo, Zuting, Zheng, Li-bing, Huang, Wei, Chi, Chang-feng, and Lou, Bao
- Abstract
Background: Neuropeptide FF (NPFF), an octapeptide of the RFamide-related peptides (FaRPs), is involved in regulatory function in various biological processes. The regulatory role of NPFF in the immune and inflammatory response was currently being revealed. Methods: Neuropeptide FF-related gene (termed LpNPFF) and its two receptors, NPFF receptor 1 (LpNPFFR1) and NPFF receptor 2 (LpNPFFR2) were identified by PCR and Semi-quantitative RT-PCR assay. Effect of LpNPFF on the production of nitric oxide (NO) in macrophage RAW264.7 cell was divided into PBS group, lipopolysaccharide (LPS) group, LPS treated with LpNPFF group, and LPS treated with LpNPFF and receptor antagonist RF9 group. Then specimens were measured by color reaction at 570 nm absorbance value. Results: Sequence analysis showed that LpNPFF cDNA consists of 835 nucleotides with a 5′- untranslated region (UTR) of 150 base pair (bp), an open reading frame (ORF) of 384 bp and a 3′-UTR of 300 bp (Accession No. MT012894). The ORF encodes 127 amino acid (aa) residues with a hydrophobic signal peptide at N-terminus and two presumptive peptides with -PQRFa structure, LpNPFF (1) and LpNPFF (2). LpNPFFR1 and LpNPFFR2 encode 427 and 444 aa residues respectively, which both have seven hydrophobic TMDs and identified as G protein coupled receptors (GPCRs). Results of tissue distribution showed that LpNPFF and receptors were highly expressed in the brain and gonad. Furtherly, in vitro assay found LpNPFF could inhibit NO production in RAW 264.7 macrophages under inflammatory stress with LPS, while its receptor antagonist RF9 caused the evoke of NO generation. Conclusions: These results contribute to the further study of neuropeptide evolution in marine organisms, and also provide a new research idea for exploring the related functions of NPFF gene. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. NPFF Decreases Activity of Human Arcuate NPY Neurons: A Study in Embryonic-Stem-Cell-Derived Model.
- Author
-
Torz, Lola, Niss, Kristoffer, Lundh, Sofia, Rekling, Jens C., Quintana, Carlos Damian, Frazier, Signe Emilie Dannulat, Mercer, Aaron J., Cornea, Anda, Bertelsen, Charlotte Vinther, Gerstenberg, Marina Kjærgaard, Hansen, Ann Maria Kruse, Guldbrandt, Mette, Lykkesfeldt, Jens, John, Linu Mary, Villaescusa, J. Carlos, and Petersen, Natalia
- Subjects
- *
NEURONS , *NEUROPEPTIDE Y , *FOOD consumption , *HYPOTHALAMUS , *FOOD supply - Abstract
Restoring the control of food intake is the key to obesity management and prevention. The arcuate nucleus (ARC) of the hypothalamus is extensively being studied as a potential anti-obesity target. Animal studies showed that neuropeptide FF (NPFF) reduces food intake by its action in neuropeptide Y (NPY) neurons of the hypothalamic ARC, but the detailed mode of action observed in human neurons is missing, due to the lack of a human-neuron-based model for pharmacology testing. Here, we validated and utilized a human-neural-stem-cell-based (hNSC) model of ARC to test the effects of NPFF on cellular pathways and neuronal activity. We found that in the human neurons, decreased cAMP levels by NPFF resulted in a reduced rate of cytoplasmic calcium oscillations, indicating an inhibition of ARC NPY neurons. This suggests the therapeutic potential of NPFFR2 in obesity. In addition, we demonstrate the use of human-stem-cell-derived neurons in pharmacological applications and the potential of this model to address functional aspects of human hypothalamic neurons. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. Lack of neuropeptide FF signalling in mice leads to reduced repetitive behavior, altered drinking behavior, and fuel type selection.
- Author
-
Zhang, Lei, Koller, Julia, Ip, Chi Kin, Gopalasingam, Gopana, Bajaj, Nikita, Lee, Nicola J., Enriquez, Ronaldo F., and Herzog, Herbert
- Abstract
Although best known for their involvement in modulating nociception, Neuropeptide FF (NPFF) group peptides have been suggested to fulfil a variety of biological functions such as feeding, anxiety behaviors and thermogenesis. However, evidence supporting these functions of NPFF is mostly pharmacological, leaving the physiological relevance unaddressed. Here we examined the physiological impact of lack of NPFF signalling in both genders using a Npff−/− mouse model. NPFF expression in the mouse is restricted to the spinal cord and brainstem while its cognate receptor NPFFR2 has wider distribution throughout the brain. Both male and female Npff−/− mice showed reduced repetitive behaviors evidenced in the marble burying test and self‐grooming test. A decrease in anxiety‐related behaviors in the Npff−/− mice was also observe in the open field test and to a lesser degree in an elevated plus maze test. Moreover, both male and female Npff−/− mice exhibited increased water intake resulting from increases in drinking size, rather than number of drinking events. During a fasting‐refeeding challenge, Npff−/− mice of both genders displayed alterations in reparatory exchange ratio that reflect a greater fuel type flexibility. Npff−/− mice were otherwise wild‐type‐like regarding body weight, body composition, feeding behaviors, locomotion or energy expenditure. Together, these findings reveal the important physiological roles of NPFF signalling in the regulation of anxiety‐related and repetitive behaviors, fluid homeostasis and oxidative fuel selection, highlighting the therapeutical potential of the NPFF system in a number of behavioral and metabolic disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Expression analysis of neuropeptide FF receptors on neuroendocrine-related neurons in the rat brain using highly sensitive in situ hybridization.
- Author
-
Higo, Shimpei, Kanaya, Moeko, and Ozawa, Hitoshi
- Subjects
- *
IN situ hybridization , *KISSPEPTIN neurons , *DOPAMINERGIC neurons , *NEURONS , *PARAVENTRICULAR nucleus , *HYPOTHALAMUS , *SOMATOSENSORY cortex - Abstract
RF-amide peptides, a family of peptides characterized by a common carboxy-terminal Arg-Phe-NH2 motif, play various physiological roles in the brain including the modulation of neuroendocrine signaling. Neuropeptide FF (NPFF) receptors exhibit a high affinity for all RF-amide peptides, which suggests that the neurons expressing these NPFF receptors may have multiple functions in the brain. However, the distribution of the neurons expressing NPFF receptors in the rat brain remains poorly understood. This study aimed to determine the detailed histological distribution of mRNA that encodes the neuropeptide FF receptors (Npffr1 and Npffr2) in the rat brain using in situ hybridization. Neurons with strong Npffr1 expression were observed in the lateral septal nucleus and several hypothalamic areas related to neuroendocrine functions, including the paraventricular nucleus (PVN) and arcuate nucleus, whereas Npffr2-expressing neurons were observed mainly in brain regions involved in somatosensory pathways, such as several subnuclei of the thalamus. Npffr1 expression was observed in 70% of corticotropin-releasing hormone neurons, but in only a small population of oxytocin and vasopressin neurons in the PVN. Npffr1 expression was also observed in the dopaminergic neurons in the periventricular nucleus and the dorsal arcuate nucleus, and in the kisspeptin neurons in the anteroventral periventricular nucleus. These results suggest that NPFFR1-mediated signaling may be involved in neuroendocrine functions, such as in reproduction and stress response. In conjunction with a detailed histological map of NPFFRs, this study provides useful data for future neuroendocrine research. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Neuropeptide FF modulates neuroendocrine and energy homeostasis through hypothalamic signaling
- Author
-
Ya-Tin Lin and Jin-Chung Chen
- Subjects
anxiety ,depression ,energy homeostasis ,food intake ,hypothalamic–pituitary–adrenal axis ,hypothalamus ,neuroendocrine ,neuropeptide ff ,neuropeptide ff receptor 2 ,obesity ,paraventricular nucleus ,thermogenesis ,Physiology ,QP1-981 - Abstract
Neuropeptide FF (NPFF) is known as a morphine-modulating peptide and was first isolated in 1985. It has been characterized as an RF-amide peptide. The traditional role of NPFF is mediation of the pain response, and it displays both anti-opioid and pro-opioid actions through central nervous system. In the recent decade, additional evidence has revealed some untraditional features of NPFF, such as regulation of the neuroendocrine system, energy homeostasis, anti-inflammation, pain transmission, and peripheral modulation of adipose tissue macrophages. Neuropeptide FF receptor 2 (NPFFR2) is a physiological receptor of NPFF, and the actions of NPFF may occur through downstream NPFFR2 signaling. NPFF and NPFFR2 increase the neuronal activity in various areas of the hypothalamus to modulate the hypothalamic–pituitary–adrenal axis, the autonomic nervous system, food intake, and energy balance. These underlying cellular mechanisms have been explored in the past few years. Here, we review the impact of NPFF and related RF-amide peptides on hypothalamic function. The interaction of NPFF with NPFFR2 in the hypothalamus is emphasized, and NPFF-NPFFR2 system may represent an important therapeutic target in hypothalamic-related disorders in the future.
- Published
- 2019
- Full Text
- View/download PDF
12. Central and peripheral modulation of gastrointestinal transit in mice by DN‐9, a multifunctional opioid/NPFF receptor agonist.
- Author
-
Xu, Biao, Guo, Yuanyuan, Zhang, Mengna, Zhang, Run, Chen, Dan, Zhang, Qinqin, Xiao, Jian, Xu, Kangtai, Li, Ning, Qiu, Yu, Zhu, Hanwen, Niu, Jiandong, Zhang, Xiaoyu, and Fang, Quan
- Subjects
- *
OPIOID receptors , *OPIOIDS , *INTRAPERITONEAL injections , *MICE - Abstract
Background: The nonapeptide DN‐9 functions as a multifunctional agonist to opioid and neuropeptide FF (NPFF) receptors and exhibits antinociceptive effects at the central and peripheral levels. Methods: The effects of DN‐9 on small and colonic intestinal transit were evaluated using the upper gastrointestinal (GI) transit test and colonic bead expulsion assay, respectively. Opioid and NPFF receptor antagonists were used to investigate the mechanisms of DN‐9‐induced GI inhibition. Furthermore, the agonism of the DN‐9 analog [Phg9]‐DN‐9 to opioid and NPFF receptors was tested by the cAMP assay. Key results: Intracerebroventricular administration of DN‐9 dose‐dependently slowed upper GI transit and colonic expulsion via mu‐ and kappa‐opioid receptors in the brain, independent of the delta‐opioid receptor. Similarly, intraperitoneal injection of DN‐9 dose‐dependently inhibited GI propulsion via the peripheral opioid receptors. DN‐9‐induced GI transit inhibitions were significantly aggravated by the NPFF receptor antagonist RF9. Moreover, the DN‐9 analog [Phg9]‐DN‐9, an agonist at mu‐, delta‐, and kappa‐opioid receptors but not NPFF receptors, inhibited GI more potently than DN‐9. In addition, intracerebroventricular NPFF significantly attenuated the central inhibitory effects induced by [Phg9]‐DN‐9 and morphine. However, central and peripheral injections of NPFF or RF9 almost had no significant effects on GI transit by itself. Conclusion and Inferences: Intracerebroventricular and intraperitoneal administrations of DN‐9 inhibit GI transit via opioid receptors in mice by central and peripheral mechanisms, respectively. In addition, the NPFF agonism of DN‐9 possesses antiopioid effects on GI transit, which might explain the reduced constipation at the antinociceptive doses. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
13. NPFF increases fusogenic proteins syncytin 1 and syncytin 2 via GCM1 in first trimester primary human cytotrophoblast cells.
- Author
-
Zhu, Hua, Peng, Bo, Klausen, Christian, Yi, Yuyin, Li, Yan, Xiong, Siyuan, Dadelszen, Peter, and Leung, Peter C. K.
- Abstract
Neuropeptide FF (NPFF) is well‐known for its roles in the central nervous system. Despite studies demonstrating that NPFF receptor 2 (NPFFR2) mRNA is highest in placenta, nothing is known about NPFF‐NPFFR2 functions in placental development. Here, we investigated the effects of NPFF‐NPFFR2 on expression of syncytial [human chorionic gonadotropin (hCG) β] and fusogenic [syncytin 1, syncytin 2, and glial cells missing 1 (GCM1)] genes in first trimester primary human cytotrophoblast cells. By analyzing two publicly available microarray data sets, we found that NPFF is consistently expressed throughout gestation whereas NPFFR2 increases in first trimester and is elevated in placenta samples from women with preeclampsia. Immunohistochemistry showed that NPFFR2, syncytin 1/2, and GCM1 each displayed unique patterns of expression among different trophoblast populations in first trimester placenta. Treatment of primary human cytotrophoblast cells with NPFF increased the mRNA and protein levels of hCG β, syncytin 1, syncytin 2, and GCM1; and knockdown of NPFFR2 abolished these effects. Interestingly, GCM1 mediated NPFF‐induced upregulation of syncytin 1 and syncytin 2, but not hCG β, in primary human cytotrophoblasts. Our results demonstrate that NPFF acts via NPFFR2 to enhance production of hCG β and promote GCM1‐dependent expression of syncytin 1 and 2 in human cytotrophoblasts. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. The blockade of neuropeptide FF receptor 1 and 2 differentially contributed to the modulating effects on fentanyl-induced analgesia and hyperalgesia in mice.
- Author
-
Chen, Dan, Zhang, Mengna, Zhang, Qinqin, Wu, Shuyuan, Yu, Bowen, Zhang, Xiaodi, Hu, Xuanran, Zhang, Shichao, Yang, Zhenyun, Kuang, Junzhe, Xu, Biao, and Fang, Quan
- Subjects
- *
OPIOID receptors , *HYPERALGESIA , *ANALGESIA , *INTRATHECAL injections , *MALE models , *MICE - Abstract
Neuropeptide FF (NPFF) plays a critical role in various physiological processes through the activation of neuropeptide FF receptor 1 and 2 (NPFFR1 and NPFFR2). Numerous evidence has indicated that NPFF exhibits opposite opioid-modulating effects on opioid-induced analgesia after supraspinal and spinal administrations, while the detailed role of NPFFR1 and NPFFR2 remains unclear. In this study, we employed pharmacological and genetic inhibition of NPFFR to investigate the modulating roles of central NPFFR1 and NPFFR2 in opioid-induced analgesia and hyperalgesia, using a male mouse model of acute fentanyl-induced analgesia and secondary hyperalgesia. Our findings revealed that intrathecal (i.t.) injection of the nonselective NPFFR antagonist RF9 significantly enhanced fentanyl-induced analgesia, whereas intracerebroventricular (i.c.v.) injection did not show the same effect. Moreover, NPFFR2 deficient (npffr2 −/−) mice exhibited stronger analgesic responses to fentanyl compared to wild type (WT) or NPFFR1 knockout (npffr1 −/−) mice. Intrathecal injection of RF9 in npffr1 −/− mice also significantly enhanced fentanyl-induced analgesia. These results indicate a crucial role of spinal NPFFR2 in the enhancement of opioid analgesia. Contrastingly, hyperalgesia induced by fentanyl was markedly reversed in npffr1 −/− mice but remained unaffected in npffr2 −/− mice. Similarly, i.c.v. injection of the selective NPFFR1 antagonist RF3286 effectively prevented fentanyl-induced hyperalgesia in WT or npffr2 −/− mice. Notably, co-administration of i.c.v. RF3286 and i.t. RF9 augmented fentanyl-induced analgesia while reducing hyperalgesia. Collectively, these findings highlight the modulating effects of blocking spinal NPFFR2 and supraspinal NPFFR1 on fentanyl-induced analgesia and hyperalgesia, respectively, which shed a light on understanding the pharmacological function of NPFF system in future studies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Modulatory role of neuropeptide FF system in macrophages.
- Author
-
Wang, Yaxing, Zuo, Zhuo, Shi, Jiajia, Fang, Yanwei, Yin, Zhongqian, Wang, Zhe, Yang, Zhouqi, Jia, Bin, and Sun, Yulong
- Subjects
- *
MACROPHAGES , *PAIN perception , *ADIPOSE tissues , *INFLAMMATION , *ADIPOSE tissue physiology - Abstract
Neuropeptide FF (NPFF) is an octapeptide that regulates various cellular processes, especially pain perception. Recently, there has been a growing interest in understanding the modulation of NPFF in neuroendocrine inflammation. This review aims to provide a thorough overview of the regulation of NPFF in macrophage-mediated biological processes. We delve into the impact of NPFF on macrophage polarization, self-renewal modulation, and the promotion of mitophagy, facilitating the transition from thermogenic fat to fat-storing adipose tissue. Additionally, we explore the NPFF-dependent regulation of the inflammatory response mediated by macrophages, its impact on the differentiation of macrophages, and its capacity to induce alterations in the transcriptome of macrophages. We also address the potential of NPFF as a therapeutic molecule in the field of neuroendocrine inflammation. Overall, our work offers an understanding of the influence of NPFF on macrophage, facilitating the exploration of its pharmacological significance in future studies. • NPFF changes the transcriptome of macrophages. • NPFF affects the polarization of macrophages and the self-renewal of macrophages. • NPFF regulates the macrophage-mediated inflammatory response. • NPFF alters macrophage differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Neuropeptide FF and Neuropeptide VF and their control in the regulation of energy homeostasis and glucose metabolism
- Author
-
Koller, Julia and Koller, Julia
- Abstract
Despite the growing obesity epidemic and its adverse health effects, such as diabetes and cardiovascular diseases, effective treatments are still missing. This is partly due to our insufficient understanding of the regulatory mechanisms underlying energy homeostasis. This work used transgenic and knockout mouse models combined with neuronal manipulation and pathway tracing to investigate the role of the Neuropeptide FF (NPFF) and Neuropeptide VF (NPVF) in the regulation of energy balance and glucose homeostasis. This study reveals that NPFF signalling plays important roles in fluid and glucose homeostasis, since Npff-/- mice exhibit an increased water intake and improved glucose tolerance, the latter being due to higher insulin responsiveness. Coherently, chemogenetic activation of brainstem NPFF neurons via DREADD technology impaired glucose tolerance, whereas inhibition improved it. In addition, the lack of NPFF signalling leads to a rise in energy expenditure and brown adipose tissue thermogenesis, however, shows impaired high fat diet-induced thermogenesis at thermoneutrality. Together this indicates a critical role of NPFF signalling in the thermos-sensory and dietary regulation of thermogenesis. Consistent with a role of NPFF in the control of energy metabolism, targeted stimulation of NPFF neurons induces a torpor-like state, characterized by strong hypothermia and hypometabolic reactions. Lack of NPVF shows strong gender-specific effects on glucose homeostasis and energy metabolism, such as providing protection against HFD-induced glucose intolerance in male mice. An exacerbated diet-induced obesity in Npvf-/- mice, attributable to reduced energy expenditure observed in females only, and/or higher food intake observed in both genders, suggests a role of NPVF signalling in the regulation of feeding and energy metabolism under positive energy balance. Furthermore, this work reveals important roles of both NPFF and NPVF signalling in the regulation of anxiety-r
- Published
- 2023
17. A GPCR-neuropeptide axis dampens hyperactive neutrophils by promoting an alternative-like polarization during bacterial infection.
- Author
-
Gour, Naina, Yong, Hwan Mee, Magesh, Aishwarya, Atakkatan, Aishwarya, Andrade, Felipe, Lajoie, Stephane, and Dong, Xinzhong
- Subjects
- *
NEUTROPHILS , *BACTERIAL diseases , *CELL physiology , *LUNG infections , *COMMUNICABLE diseases - Abstract
The notion that neutrophils exist as a homogeneous population is being replaced with the knowledge that neutrophils adopt different functional states. Neutrophils can have a pro-inflammatory phenotype or an anti-inflammatory state, but how these states are regulated remains unclear. Here, we demonstrated that the neutrophil-expressed G-protein-coupled receptor (GPCR) Mrgpra1 is a negative regulator of neutrophil bactericidal functions. Mrgpra1-mediated signaling was driven by its ligand, neuropeptide FF (NPFF), which dictated the balance between pro- and anti-inflammatory programming. Specifically, the Mrgpra1-NPFF axis counter-regulated interferon (IFN) γ-mediated neutrophil polarization during acute lung infection by favoring an alternative-like polarization, suggesting that it may act to balance overzealous neutrophilic responses. Distinct, cross-regulated populations of neutrophils were the primary source of NPFF and IFNγ during infection. As a subset of neutrophils at steady state expressed NPFF, these findings could have broad implications in various infectious and inflammatory diseases. Therefore, a neutrophil-intrinsic pathway determines their cellular fate, function, and magnitude of infection. [Display omitted] • Mrgpra1 is a neutrophil-expressed anti-inflammatory Mrgpr • Mrgpra1 drives anti-inflammatory neutrophils and inhibits activated neutrophils • A distinct population of neutrophils makes the Mrgpra1 ligand, neuropeptide FF (NPFF) • The Mrgpra1-NPFF axis inhibits IFNγ-driven anti-bacterial neutrophils Although it is known that pro- and anti-inflammatory neutrophils exist, the mechanisms that dictate their polarization remain unclear. Gour et al. report that neutrophils express a canonical neuronal receptor, Mrgpra1, and a neuropeptide ligand, neuropeptide FF. They find that NPFF-Mrgpra1 signaling inhibits IFNγ-mediated conditioning to promote anti-inflammatory neutrophil polarization, indicating that a neutrophil-intrinsic pathway determines their cellular fate and function. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Ablation of NPFFR2 in Mice Reduces Response to Single Prolonged Stress Model
- Author
-
Ya-Tin Lin, Yi-Ling Huang, Sze-Chi Tsai, and Jin-Chung Chen
- Subjects
neuropeptide FF ,NPFFR2 ,stress ,anxiety ,HPA axis ,SPS ,Cytology ,QH573-671 - Abstract
Mental stress is highly related to many clinical symptoms and disorders, as it activates the hypothalamic-pituitary-adrenocortical (HPA) axis to affect a wide variety of physiological functions. Furthermore, stress leads to the aberrations in HPA axis activity and disruptions of body homeostasis. It was previously shown that neuropeptide FF (NPFF) regulates the HPA axis through the activation of hypothalamus paraventricular nucleus (PVN), and genetic overexpression or pharmacological stimulation of NPFF receptor 2 (NPFFR2) triggers hyperactivity of HPA axis and suppresses behavioral correlates of emotion in mice. In this study, we further examined the role of NPFFR2 in stress response in mice by utilizing a single prolonged stress (SPS). SPS is considered a model of post-traumatic stress disorder (PTSD), and mice undergo physical restraint, forced swimming, and ether anesthesia within a day followed by social isolation for one week. NPFFR2 knockout B6 mice were generated by CRISPR/Cas9 technology and exposed to SPS. The NPFFR2 knockouts showed resistance to stress exposure-induced anxiety-like behaviors and HPA axis hyperactivity. Additionally, the hippocampal mRNA levels of glucocorticoid receptor and mineralocorticoid receptor were reduced in wild-type (WT) mice but not in NPFFR2 knockouts after stress exposure. Our data also suggested that NPFFR2 knockout mice have stronger negative feedback on the HPA axis after exposure to SPS. Mice with intra-PVN Npffr2 shRNA injection displayed trends toward resistance to SPS exposure in both behavioral and molecular assays. Together, our findings suggest that NPFFR2 may be a potential therapeutic target for disorders relating to stress/anxiety and HPA dysregulation.
- Published
- 2020
- Full Text
- View/download PDF
19. Systemic administration of the bifunctional opioid/neuropeptide FF receptors agonist BN-9 produced peripheral antinociception in preclinical mouse models of pain.
- Author
-
Li, Ning, Han, Zheng-Lan, Xu, Biao, Zhang, Meng-Na, Zhang, Ting, Shi, Xue-Rui, Zhao, Wei-Dong, Guo, Yuan-Yuan, Zhang, Qin-Qin, and Fang, Quan
- Subjects
- *
OPIOIDS , *NEUROPEPTIDES , *G protein coupled receptors , *BIFUNCTIONAL catalysis , *INTRAPERITONEAL injections , *ANALGESIA - Abstract
Abstract We recently characterized a novel bifunctional agonist for opioid and neuropeptide FF receptors, named BN-9, which exhibited potent analgesia in the mouse tail-flick test when given centrally. To further evaluate its potential therapeutic efficacy for translational-medical development, the current work was performed to explore the antinociceptive activities of intraperitoneal (i.p.) administration of BN-9 in mouse models of tail-flick assay, formalin pain, visceral pain and post-operative pain. In the tail-flick test, BN-9 induced a dose-related antinociceptive effect, which was fully blocked by systemic pretreatment with the peripheral acting opioid receptor antagonist naloxone methiodide, but not supraspinal naloxone methiodide, implying the involvement of the peripheral opioid receptors. In addition, the systemic antinociception of BN-9 was antagonized by the selective antagonists of the μ- and κ-opioid receptors, independently of the δ-opioid and neuropeptide FF receptors. Similarly, dose-dependent analgesia was also produced by systemic BN-9 in different pain models via the peripheral opioid receptors, independently of the neuropeptide FF receptors. Furthermore, the side-effects of systemic BN-9 on motor performance, tolerance development and gastrointestinal transit inhibition were also evaluated. Repeated systemic injection of BN-9 produced non-tolerance analgesia over 8 days. Compared with morphine, intraperitoneal administration of BN-9 exerted less inhibition of gastrointestinal transit. These data show that BN-9 induced systemic analgesia with reduced side-effects on tolerance and constipation. This article suggests that systemic injection of BN-9 causes effective antinociception in different preclinical pain models via the peripheral opioid receptors, providing an attractive approach to develop peripherally acting opioid analgesics with multiple targeting properties. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
20. A subtype-specific neuropeptide FF receptor antagonist attenuates morphine and nicotine withdrawal syndrome in the rat.
- Author
-
Malin, David H., Henceroth, Mallori M., Elayoubi, Joanne, Campbell, Joseph R., Anderson, Andrea, Goyarzu, Pilar, Izygon, Jonathan, Madison, Caitlin A., Ward, Christopher P., and Burstein, Ethan S.
- Subjects
- *
TREATMENT of drug withdrawal symptoms , *NICOTINE addiction treatment , *MORPHINE abuse , *NEUROPEPTIDES , *LABORATORY rats - Abstract
Highlights • AC-262620 is a small-molecule, systemically active, selective antagonist of the FF1 receptor. • Ten mg/kg AC-262620 significantly reduced naloxone-precipitated somatically expressed morphine withdrawal signs in rats. • The same dose significantly reduced subsequent spontaneous withdrawal signs a day after termination of morphine infusion. • AC-262620 significantly reduced nicotine withdrawal signs precipitated by the nicotinic antagonist mecamylamine. • Results suggest that activation of the FF1 subtype of NPFF receptor contributes to opiate and nicotine physical dependence. Abstract Considerable evidence suggests the Neuropeptide FF (NPFF) and related peptides exert pro-nociceptive and anti-opiate actions, particularly at the supra-spinal level, which may contribute to opiate dependence. The FF1 receptor subtype appears to be primarily responsible for anti-opiate effects. In contrast, stimulation of the FF2 receptor primarily induces pro-opiate effects. AC-262620 is a small molecule, systemically active, selective FF1 receptor antagonist. An initial experiment showed that 10 mg/kg i.p. AC-262620 significantly reduced subsequent naloxone-precipitated somatically expressed withdrawal signs in rats infused s.c. for seven days with 0.3 mg/kg/hr morphine sulfate. A second experiment showed that the same dose of AC-262620 significantly reduced subsequent spontaneous withdrawal signs 23.75 h after termination of seven days s.c. infusion of 0.6 mg/kg/hr morphine sulfate. Chronic nicotine intake may contribute to dependence by overstimulating opiate receptors through release of opiate peptides. By analogy to opiate dependence, it was hypothesized that FF1 receptor activation contributes to nicotine dependence and withdrawal syndrome. AC-262620 significantly reduced somatically expressed withdrawal signs precipitated by the nicotinic antagonist mecamylamine in rats infused for seven days with nicotine bitartrate. Taken together, these findings suggest that NPFF or related neuropeptides contribute to opiate, as well as nicotine, dependence and withdrawal syndrome through the FF1 receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
21. Mechanism of action of anti-opioid peptides at pain syndrome.
- Author
-
Levashova, A. I. and Myagkova, M. A.
- Subjects
- *
PHARMACEUTICAL chemistry , *OPIOIDS , *PEPTIDES , *OPIOID analgesics , *ANALGESIA - Abstract
An important goal of modern medicinal chemistry is to study the mechanisms of pain and analgesia in order to design effective analgesic drugs. Opioid analgesics are the gold standard for the treatment of severe pain; however, the use of opiates is associated with the development of side effects which are, in particular, related to the activation of the anti-opioid system. Mammalians synthesize a number of endogenous peptides, such as orphanin FGGFTGARKSARKLANQ, neuropeptide FF (FLFQPQRF-NH2), tripeptide melanostatin (MIF) PLG-NH2, as well as related compounds. These anti-opioid peptides are to one extent or another involved in homeostatic control of transmission of pain impulses. The present review includes the data published to date in domestic and foreign literature on the involvement of these peptides in such undesirable phenomena as inhibition of opioid analgesia, development of opioid tolerance and dependence, and hyperalgesia. Cell-cell and molecular ligand-receptor and receptor-receptor interactions of the opioid and anti-opioid systems are considered. These data can be useful for the design of new pharmaceuticals for pain relief. The generalization and study of these mechanisms are reflected in various approaches to treatment of pain syndromes and require analysis and further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
22. Molecular cloning and characterization of the SIFamide precursor and receptor in a hymenopteran insect, Bombus terrestris.
- Author
-
Lismont, Els, Mortelmans, Nele, Verlinden, Heleen, and Vanden Broeck, Jozef
- Subjects
- *
BOMBUS terrestris , *INSECT genetics , *NEUROPEPTIDES , *MOLECULAR cloning , *SEXUAL behavior in insects , *INTERNEURONS , *CONSERVED sequences (Genetics) - Abstract
SIFamides (SIFa) are a family of neuropeptides that are highly conserved among arthropods. In insects, this peptide is mainly expressed in four medial interneurons in the pars intercerebralis and affects sexual behavior, sleep regulation and pupal mortality. Furthermore, an influence on the hatching rate has been observed. The first SIFa receptor (SIFR) was pharmacologically characterized in Drosophila melanogaster and is homologous to the vertebrate gonadotropin-inhibitory hormone (GnIH) receptor (NPFFR). In this study, we pharmacologically characterized the SIFR of the buff-tailed bumblebee Bombus terrestris. We demonstrated an intracellular increase in calcium ions and cyclic AMP (cAMP) upon ligand binding with an EC 50 value in the picomolar and nanomolar range, respectively. In addition, we studied the agonistic properties of a range of related and modified peptides. By means of quantitative real time PCR (qPCR), we examined the relative transcript levels of Bomte - SIFa and Bomte - SIFR in a variety of tissues. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
23. Development of Multifunctional and Orally Active Cyclic Peptide Agonists of Opioid/Neuropeptide FF Receptors that Produce Potent, Long-Lasting, and Peripherally Restricted Antinociception with Diminished Side Effects
- Author
-
Run Zhang, Kangtai Xu, Xuerui Shi, Biao Xu, Quan Fang, Dan Chen, Qinqin Zhang, Jiandong Niu, Yonghang Shi, Mengna Zhang, Jian Xiao, and Ning Li
- Subjects
Male ,Receptors, Neuropeptide ,Agonist ,medicine.drug_class ,Chemistry ,Analgesic ,Neuropeptide FF receptor ,Pharmacology ,Ligands ,Peptides, Cyclic ,Analgesics, Opioid ,Mice ,Opioid ,Oral administration ,Drug Discovery ,Neuropathic pain ,medicine ,Animals ,Neuralgia ,Molecular Medicine ,Neuropeptide FF ,Receptor ,medicine.drug - Abstract
We previously reported that a multifunctional opioid/neuropeptide FF receptor agonist, DN-9, achieved peripherally restricted analgesia with reduced side effects. To develop stable and orally bioavailable analogues of DN-9, eight lactam-bridged cyclic analogues of DN-9 between positions 2 and 5 were designed, synthesized, and biologically evaluated. In vitro cAMP assays revealed that these analogues, except 7, were multifunctional ligands that activated opioid and neuropeptide FF receptors. Analogue 1 exhibited improved potency for κ-opioid and NPFF2 receptors. All analogues exhibited potent, long-lasting, and peripherally restricted antinociception in the tail-flick test without tolerance development after subcutaneous administration and produced oral analgesia. Oral administration of the optimized compound analogue 1 exhibited powerful, peripherally restricted antinociceptive effects in mouse models of acute, inflammatory, and neuropathic pain. Remarkably, orally administered analogue 1 had no significant side effects, such as tolerance, dependence, constipation, or respiratory depression, at effective analgesic doses.
- Published
- 2021
- Full Text
- View/download PDF
24. NPFF stimulates human ovarian cancer cell invasion by upregulating MMP-9 via ERK1/2 signaling.
- Author
-
Wu, Ze, Jia, Qiongqiong, Liu, Boqun, Fang, Lanlan, Leung, Peter C.K., and Cheng, Jung-Chien
- Subjects
- *
MATRIX metalloproteinases , *OVARIAN cancer , *G protein coupled receptors , *OVARIAN epithelial cancer , *CANCER cells , *NEUROPEPTIDES - Abstract
Neuropeptide FF (NPFF) belongs to the RFamide peptide family. NPFF regulates a variety of physiological functions by binding to a G protein-coupled receptor (GPCR), NPFFR2. Epithelial ovarian cancer (EOC) is a leading cause of death among gynecological malignancies. The pathogenesis of EOC can be regulated by many local factors, including neuropeptides, through an autocrine/paracrine manner. However, to date, the expression and/or function of NPFF/NPFFR2 in EOC is undetermined. In this study, we show that the upregulation of NPFFR2 mRNA was associated with poor overall survival in EOC. The TaqMan probe-based RT-qPCR showed that NPFF and NPFFR2 were expressed in three human EOC cells, CaOV3, OVCAR3, and SKOV3. In comparison, NPFF and NPFFR2 expression levels were higher in SKOV3 cells than in CaOV3 or OVCAR3 cells. Treatment of SKOV3 cells with NPFF did not affect cell viability and proliferation but stimulated cell invasion. NPFF treatment upregulates matrix metalloproteinase-9 (MMP-9) expression. Using the siRNA-mediated knockdown approach, we showed that the stimulatory effect of NPFF on MMP-9 expression was mediated by the NPFFR2. Our results also showed that ERK1/2 signaling was activated in SKOV3 cells in response to the NPFF treatment. In addition, blocking the activation of ERK1/2 signaling abolished the NPFF-induced MMP-9 expression and cell invasion. This study provides evidence that NPFF stimulates EOC cell invasion by upregulating MMP-9 expression through the NPFFR2-mediated ERK1/2 signaling pathway. [Display omitted] • NPFF and NPFFR2 are expressed in human EOC cells. • Upregulation of NPFFR2 is associated with poor survival. • NPFF stimulates EOC cell invasion. • ERK1/2 signaling mediates NPFF-induced EOC cell invasion and MMP-9 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. NPFF Decreases Activity of Human Arcuate NPY Neurons:A Study in Embryonic-Stem-Cell-Derived Model
- Author
-
Torz, Lola, Niss, Kristoffer, Lundh, Sofia, Rekling, Jens C., Quintana, Carlos Damian, Frazier, Signe Emilie Dannulat, Mercer, Aaron J., Cornea, Anda, Bertelsen, Charlotte Vinther, Gerstenberg, Marina Kjærgaard, Hansen, Ann Maria Kruse, Guldbrandt, Mette, Lykkesfeldt, Jens, John, Linu Mary, Villaescusa, J. Carlos, Petersen, Natalia, Torz, Lola, Niss, Kristoffer, Lundh, Sofia, Rekling, Jens C., Quintana, Carlos Damian, Frazier, Signe Emilie Dannulat, Mercer, Aaron J., Cornea, Anda, Bertelsen, Charlotte Vinther, Gerstenberg, Marina Kjærgaard, Hansen, Ann Maria Kruse, Guldbrandt, Mette, Lykkesfeldt, Jens, John, Linu Mary, Villaescusa, J. Carlos, and Petersen, Natalia
- Abstract
Restoring the control of food intake is the key to obesity management and prevention. The arcuate nucleus (ARC) of the hypothalamus is extensively being studied as a potential anti-obesity target. Animal studies showed that neuropeptide FF (NPFF) reduces food intake by its action in neuropeptide Y (NPY) neurons of the hypothalamic ARC, but the detailed mode of action observed in human neurons is missing, due to the lack of a human-neuron-based model for pharmacology testing. Here, we validated and utilized a human-neural-stem-cell-based (hNSC) model of ARC to test the effects of NPFF on cellular pathways and neuronal activity. We found that in the human neurons, decreased cAMP levels by NPFF resulted in a reduced rate of cytoplasmic calcium oscillations, indicating an inhibition of ARC NPY neurons. This suggests the therapeutic potential of NPFFR2 in obesity. In addition, we demonstrate the use of human-stem-cell-derived neurons in pharmacological applications and the potential of this model to address functional aspects of human hypothalamic neurons.
- Published
- 2022
26. The new kisspeptin derivative - kissorphin (KSO) - attenuates acute hyperlocomotion and sensitization induced by ethanol and morphine in mice.
- Author
-
Gibula-Bruzda, Ewa, Marszalek-Grabska, Marta, Gawel, Kinga, Trzcinska, Roza, Silberring, Jerzy, and Kotlinska, Jolanta H.
- Subjects
- *
KISSPEPTIN neurons , *LOCOMOTION , *SENSITIZATION (Neuropsychology) , *ETHANOL , *ANIMAL models in research , *PHYSIOLOGY , *HUMAN locomotion , *ANIMAL experimentation , *MORPHINE , *TARDIVE akathisia , *DOSE-effect relationship in pharmacology , *PSYCHOMOTOR disorders , *ANIMALS , *MICE - Abstract
Kissorphin (KSO) is a new peptide derived from kisspeptin-10. This peptide possesses neuropeptide FF (NPFF)-like biological activity in vitro; NPFF, in many cases, inhibits opioid and ethanol effects in rodents. Therefore, the current study explored the influence of KSO on acute ethanol- and morphine-induced hyperactivity, and on the development and expression of locomotor sensitization induced by these drugs. In the present study, sensitization to locomotor effects was induced by repeated exposure to ethanol (2.4 g/kg, intraperitoneally [i.p.], 1 × 4 days) or morphine (10 mg/kg, subcutaneously [s.c.], 1 × 7 days). We found that KSO (1-10 nmol/300 μL, intravenously [i.v.]) did not have an impact on locomotor activity of naïve mice. However, it reduced both acute ethanol- (10 nmol/300 μL) and morphine-induced hyperactivity (3 and 10 nmol/300 μL). Pretreatment of animals with KSO (10 nmol/300 μL), before every ethanol or morphine injection during development of sensitization or before the ethanol or morphine challenge, attenuated the development, as well as the expression of locomotor sensitization to both substances. Moreover, prior administration of the NPFF receptor antagonist RF9 (10 nmol/300 μL, i.v.) inhibited the ability of KSO (10 nmol/300 μL) to reduce the expression of ethanol and morphine sensitization. KSO given alone, at all used doses, did not influence the motor coordination measured via the rotarod test. The results from this study show that KSO effectively attenuated acute and repeated effects of ethanol and morphine. Thus, KSO possesses NPFF-like anti-opioid activity in these behavioral studies. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
27. Peripheral and central sites of action for anti-allodynic activity induced by the bifunctional opioid/NPFF receptors agonist BN-9 in inflammatory pain model.
- Author
-
Zhang, Run, Xu, Biao, Zhang, Meng-na, Zhang, Ting, Wang, Zi-long, Zhao, Geng, Zhao, Guang-hai, Li, Ning, Fang, Quan, and Wang, Rui
- Subjects
- *
NEUROPEPTIDES , *OPIOIDS , *CARRAGEENANS , *INFLAMMATION , *PAIN - Abstract
The activation of opioid and neuropeptide FF (NPFF) receptors plays important roles to modulate nociceptive signal in inflammatory pain states. Recently, BN-9 (Tyr- D . Ala-Gly-Phe-Gln-Pro-Gln-Arg-Phe-NH 2 ) was pharmacologically characterized as a novel bifunctional agonist at both opioid and NPFF receptors. In the present study, the anti-allodynic activity and site(s) of action of BN-9 were assessed in a mouse model of carrageenan-induced inflammatory pain. In mice, BN-9 induced a dose-dependent anti-allodinic effect through opioid receptor at supraspinal or spinal level, and this effect was augmented by pretreatment with the NPFF receptor antagonist at the same level. In contrast, peripheral administration of BN-9 produced opioid receptor-mediated anti-allodynia, which was insensitive of the NPFF receptor antagonist. In addition, systemic BN-9 produced anti-allodynic effect via opioid receptors, independent of NPFF system. Therefore, these data indicate that central, peripheral or systemic administrations of BN-9 exert potent analgesic activities in inflammatory pain model via opioid receptor, and central effects of BN-9 are associated with NPFF system. Interestingly, systemic anti-allodynia of BN-9 was blocked by intraperitoneal administration of the opioid receptor antagonists, naloxone and naloxone methiodide, but not by intracerebroventricular injection of the peripherally acting opioid antagonist naloxone methiodide. Furthermore, BN-9-induced systemic anti-allodynia was reversed by intraplantar administration of naloxone, but not by peripheral administration of the NPFF receptor antagonist. Taken together, our data further suggest that systemic BN-9-induced anti-allodynic effect is mainly mediated by peripheral opioid receptors, independent of NPFF receptors. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
28. Adipose tissue macrophages develop from bone marrow–independent progenitors in Xenopus laevis and mouse.
- Author
-
Hassnain Waqas, Syed F., Noble, Anna, Hoang, Anh C., Ampem, Grace, Popp, Manuela, Strauß, Sarah, Guille, Matthew, and Röszer, Tamás
- Subjects
XENOPUS laevis ,ADIPOSE tissues ,YOLK sac ,MACROPHAGES ,MICE - Abstract
ATMs develop from early myeloid progenitors, allowing monocyte‐independent ATM replenishment. ATMs have a metabolic impact in mammals as they contribute to metabolically harmful AT inflammation. The control of the ATM number may have therapeutic potential; however, information on ATM ontogeny is scarce. Whereas it is thought that ATMs develop from circulating monocytes, various tissue‐resident Mϕs are capable of self‐renewal and develop from BM‐independent progenitors without a monocyte intermediate. Here, we show that amphibian AT contains self‐renewing ATMs that populate the AT before the establishment of BM hematopoiesis. Xenopus ATMs develop from progenitors of aVBI. In the mouse, a significant amount of ATM develops from the yolk sac, the mammalian equivalent of aVBI. In summary, this study provides evidence for a prenatal origin of ATMs and shows that the study of amphibian ATMs can enhance the understanding of the role of the prenatal environment in ATM development. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
29. Neuropeptide FF and Its Receptors: Therapeutic Applications and Ligand Development
- Author
-
Thuy Nguyen, Yanan Zhang, Julie A. Marusich, and Jun-Xu Li
- Subjects
Receptors, Neuropeptide ,Drug ,media_common.quotation_subject ,Pain ,Endogeny ,Anxiety ,Ligands ,01 natural sciences ,Article ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,Drug Discovery ,medicine ,Animals ,Humans ,Neuropeptide FF ,Receptor ,030304 developmental biology ,media_common ,0303 health sciences ,Chemistry ,Ligand ,Small molecule ,Rats ,0104 chemical sciences ,Analgesics, Opioid ,010404 medicinal & biomolecular chemistry ,Opioid ,Molecular Medicine ,Peptidomimetics ,Oligopeptides ,Neuroscience ,medicine.drug - Abstract
The endogenous neuropeptide FF (NPFF) and its two cognate G protein-coupled receptors, Neuropeptide FF Receptors 1 and 2 (NPFFR1 and NPFFR2), represent a relatively new target system for many therapeutic applications including pain regulation, modulation of opioid side effects, drug reward, anxiety, cardiovascular conditions and other peripheral effects. Since the cloning of NPFFR1 and NPFFR2 in 2000, significant progress has been made to understand their pharmacological roles and interactions with other receptor systems, notably the opioid receptors. A variety of NPFFR ligands with different mechanisms of action (agonists or antagonists) have been discovered although with limited subtype selectivities. Differential pharmacological effects have been observed for many of these NPFFR ligands, depending on assays/models employed and routes of administration. In this Perspective, we highlight the therapeutic potentials, current knowledge gaps, and the latest updates of the development of peptidic and small molecule NPFFR ligands as tool compounds and therapeutic candidates.
- Published
- 2020
- Full Text
- View/download PDF
30. Neuropeptide FF receptor 2 inhibits capsaicin-induced CGRP Upregulation in mouse trigeminal ganglion
- Author
-
Sze Chi Tsai, Jin-Chung Chen, Po Hung Hsu, Zachary Yu, and Ya Tin Lin
- Subjects
medicine.medical_specialty ,Short Report ,Neuropeptide FF receptor ,lcsh:Medicine ,Stimulation ,Calcitonin gene-related peptide ,Neuropeptide FF (NPFF) ,03 medical and health sciences ,chemistry.chemical_compound ,Trigeminal ganglion ,0302 clinical medicine ,NPFFR2 ,Downregulation and upregulation ,Neuromodulation ,Internal medicine ,Medicine ,Neuropeptide FF ,CGRP ,Migraine ,030304 developmental biology ,0303 health sciences ,Trigeminovascular pathway ,integumentary system ,business.industry ,lcsh:R ,Headache ,General Medicine ,Anesthesiology and Pain Medicine ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Capsaicin ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Background Stimulation of trigeminovascular pathway is widely used to establish the headache animal model. Headache is a common neurological disorder, in which symptomatic attacks are mediated by calcitonin-gene-related peptide (CGRP). CGRP is synthesized and released from the trigeminal ganglion to transmit pain signals under stimulation. On the other hand, Neuropeptide FF (NPFF) is a candidate transmitter/modulator for migraine, and stimulation of its receptor, NPFFR2, increases the expression and release of CGRP in mice sensory neurons. Here, we investigate the impact of NPFFR2 on trigeminal CGRP level in a capsaicin-induced headache mouse model. Methods Mice were intracisternally injected with capsaicin into the cisterna magna to activate the trigeminovascular pathway and induce headache symptoms. Mice pretreated with Npffr2-shRNA or NPFFR2 knockouts were adopted to test the impact of NPFFR2 on capsaicin-induced CGRP upregulation in trigeminal ganglion. The gene silencing effect of Npffr2-shRNA in trigeminal ganglion was confirmed by real-time PCR. Trigeminal CGRP level was determined by immunofluorescence staining, and the percentage of CGRP-positive cell was calculated after setting the signal intensity threshold by Image J software. Amount of trigeminal CGRP in NPFFR2 overexpressed mice was also measured by CGRP ELISA. Findings Infusion of capsaicin into the cisterna magna upregulated the CGRP in trigeminal ganglion and induced spontaneous pain behaviors including the reduction of locomotor activity and the increase of freezing behavior. Intracisternal injection of Npffr2-shRNA reduced the mRNA of Npffr2 in trigeminal ganglion. Mice pretreatment with Npffr2-shRNA prevented capsaicin-induced CGRP upregulation in trigeminal ganglion. Similarly, CGRP upregulation was also reduced in NPFFR2 knockout mice. On the contrary, trigeminal CGRP was increased in NPFFR2 overexpressed mice. Conclusions Reducing the level of NPFFR2 leads to the downregulation of capsaicin-induced CGRP in trigeminal ganglion, which would consequently attenuate the activation of trigeminovascular pathway. Thus, NPFFR2 could serve as a potential target for neuromodulation of cephalic pain.
- Published
- 2020
- Full Text
- View/download PDF
31. RFamide Peptides: Structure, Function, Mechanisms and Pharmaceutical Potential
- Author
-
Maria Findeisen, Annette G. Beck-Sickinger, and Daniel Rathmann
- Subjects
RFamide ,neuropeptide FF ,kisspeptin ,prolactin-releasing peptide ,QRFP ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Different neuropeptides, all containing a common carboxy-terminal RFamide sequence, have been characterized as ligands of the RFamide peptide receptor family. Currently, five subgroups have been characterized with respect to their N-terminal sequence and hence cover a wide pattern of biological functions, like important neuroendocrine, behavioral, sensory and automatic functions. The RFamide peptide receptor family represents a multiligand/multireceptor system, as many ligands are recognized by several GPCR subtypes within one family. Multireceptor systems are often susceptible to cross-reactions, as their numerous ligands are frequently closely related. In this review we focus on recent results in the field of structure-activity studies as well as mutational exploration of crucial positions within this GPCR system. The review summarizes the reported peptide analogs and recently developed small molecule ligands (agonists and antagonists) to highlight the current understanding of the pharmacophoric elements, required for affinity and activity at the receptor family. Furthermore, we address the biological functions of the ligands and give an overview on their involvement in physiological processes. We provide insights in the knowledge for the design of highly selective ligands for single receptor subtypes to minimize cross-talk and to eliminate effects from interactions within the GPCR system. This will support the drug development of members of the RFamide family.
- Published
- 2011
- Full Text
- View/download PDF
32. Pharmacological characterization of EN-9, a novel chimeric peptide of endomorphin-2 and neuropeptide FF that produces potent antinociceptive activity and limited tolerance.
- Author
-
Wang, Zi-long, Li, Ning, Wang, Pei, Tang, Hong-hai, Han, Zheng-lan, Song, Jing-jing, Li, Xu-hui, Yu, Hong-ping, Zhang, Ting, Zhang, Run, Xu, Biao, Zhang, Meng-na, Fang, Quan, and Wang, Rui
- Subjects
- *
CHIMERIC proteins , *OLIGOPEPTIDES , *NEUROPEPTIDES , *ANALGESICS , *DRUG tolerance , *DRUG interactions - Abstract
Mounting evidences indicate the functional interactions between neuropeptide FF (NPFF) and opioids, including the endogenous opioids. In the present work, EN-9, a chimeric peptide containing the functional domains of the endogenous opioid endomorphin-2 (EM-2) and NPFF, was synthesized and pharmacologically characterized. In vitro cAMP assay demonstrated that EN-9 was a multifunctional agonist of κ-opioid, NPFF 1 and NPFF 2 receptors. In the mouse tail-flick test, intracerebroventricularly (i.c.v.) administration of EN-9 produced significant antinociception with an ED 50 value of 13.44 nmol, which lasted longer than that of EM-2. In addition, EN-9 induced potent antinociception after both intravenous (i.v.) and subcutaneous (s.c.) injection. Furthermore, the experiments using the antagonists of opioid and NPFF receptors indicated that the central antinociception of EN-9 was mainly mediated by κ-opioid receptor, independently on NPFF receptors. Notably, the central antinociception of EN-9 was not reduced over a period of 6 days repeated i.c.v. injection. Repeated i.c.v. administration of EN-9 with the NPFF 1 and NPFF 2 receptors antagonist RF9 resulted in a progressive loss of analgesic potency, consistent with the development of tolerance. Moreover, central administration of EN-9 induced the place conditioning aversion only at a high dose of 60 nmol, but not at low doses. At supraspinal level, only high dose of EN-9 (60 nmol, i.c.v.) inhibited gastrointestinal transit via NPFF receptors. Similarly, systemic administration of EN-9 also inhibited gastrointestinal transit at high doses (10 and 30 mg/kg, i.v.). Taken together, the multifunctional agonist of κ-opioid and NPFF receptors EN-9 produced a potent, non-tolerance forming antinociception with limited side effects. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
33. What's in a Name? Roles of RFamide-Related Peptides Beyond Gonadotrophin Inhibition.
- Author
-
Kim, J. S.
- Subjects
- *
GONADOTROPIN-inhibitory hormone , *NEUROPEPTIDES , *NEUROENDOCRINE system , *NEUROENDOCRINOLOGY , *POLYPEPTIDES - Abstract
RFamide-related peptides (RFRPs) have been heavily implicated in the control of reproductive function subsequent to their discovery more than 16 years ago. However, recent studies using genetic and pharmacological tools have challenged their importance in regulating the hypothalamic- pituitary-gonadal axis. It is generally accepted that RFRPs act as part of a wider RFamide system, which involves two receptors, called the neuropeptide FF receptors (NPFFR1 and R2), and includes the closely-related neuropeptide NPFF. NPFF has been studied ever since the 1980s and many of the functions of NPFF are also shared by RFRPs. The current review questions whether these functions of NPFF are indeed specific to just NPFF alone and presents evidence from both neuroendocrine and pharmacological perspectives. Furthermore, recently emerging new functions of RFRPs are discussed with the overall goal of clarifying the functions of RFRPs beyond the hypothalamic-pituitary-gonadal axis. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
34. The effect of neuropeptide FF in the amygdala kindling model.
- Author
-
Buffel, I., Meurs, A., Portelli, J., Raedt, R., De Herdt, V., Poppe, L., De Meulenaere, V., Wadman, W., Bihel, F., Schmitt, M., Vonck, K., Bourguignon, J.‐J., Simonin, F., Smolders, I., and Boon, P.
- Subjects
- *
SEIZURES (Medicine) , *NEUROPEPTIDES , *AMYGDALOID body , *ANTICONVULSANTS , *BRAIN stimulation , *THERAPEUTICS - Abstract
Objective Neuropeptide FF ( NPFF) and its receptors ( NPFF1R and NPFF2R) are differentially distributed throughout the central nervous system. NPFF reduces cortical excitability in rats when administered intracerebroventricularly (i.c.v.), and both NPFF and NPFF1R antagonists attenuate pilocarpine-induced limbic seizures. In this study, our aim was to determine whether NPFF exerts anticonvulsant or anti-epileptogenic effects in the rat amygdala kindling model for temporal lobe seizures. Methods Male Wistar rats were implanted with a recording/stimulation electrode in the right amygdala and a cannula in the left lateral ventricle. In a first group of animals, the afterdischarge threshold ( ADT) was determined after a single i.c.v. infusion of saline ( n = 8) or NPFF (1 nmol/h for 2 h; n = 10). Subsequently, daily infusion of saline ( n = 8) or NPFF (1 nmol/h for 2 h; i.c.v.; n = 9) was performed, followed by a kindling stimulus ( ADT+200 μA). Afterdischarge duration and seizure severity were evaluated after every kindling stimulus. A second group of rats ( n = 7) were fully kindled, and the effect of saline or a high dose of NPFF (10 nmol/h for 2 h, i.c.v.) on ADT and the generalized seizure threshold ( GST) was subsequently determined. Results In naive rats, NPFF significantly increased the ADT compared to control (435 ± 72 μA vs 131 ± 23 μA [ P < 0.05]). When rats underwent daily stimulations above the ADT, NPFF did not delay or prevent kindling acquisition. Furthermore, a high dose of NPFF did not alter ADT or GST in fully kindled rats. Conclusions I.c.v. administration of NPFF reduced excitability in the amygdala in naive, but not in fully kindled rats, and had no effect on kindling acquisition. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
35. NPYFa, A Chimeric Peptide of Met-Enkephalin, and NPFF Induces Tolerance-Free Analgesia.
- Author
-
Mudgal, Annu, Kumar, Krishan, Mollereau, Catherine, and Pasha, Santosh
- Subjects
- *
OPIOID analgesics , *ENKEPHALINS , *NEUROPEPTIDES , *METHIONINE , *CIRCULAR dichroism , *G proteins , *OPIOID receptors - Abstract
Methionine-enkephalin-Arg-Phe is an endogenous amphiactive analgesic peptide. Neuropeptide FF, on the other hand, is reported for its role in opioid modulation and tolerance development. Based on these reports, in the present study we designed a chimeric peptide NPYFa ( YGGFMKKKPQRFamide), having the Met-enkephalin (opioid) and PQRFamide sequence of neuropeptide FF, which can then target both the opioid and neuropeptide FF receptors. We hypothesized that the chimeric peptide so designed would have both analgesic properties and further aid in understanding of the role of neuropeptide FF in the development of opiate tolerance. Our studies indicated that NPYFa induced an early onset, potent, dose-dependent and prolonged antinociception. Additionally, antagonists ( MOR, KOR, and DOR) pretreatment studies determined a KOR-mediated antinociception activity of the ligand. Further, in vitro binding studies using the Eu- GTP- γS binding assay on cell lines expressing opioid and NPFF receptors showed binding to both the opioid and neuropeptide FF receptors suggesting a multiple receptor binding character of NPYFa. Moreover, chronic (6 days) treatment with NPYFa exhibited an absence of tolerance development subsequent to its analgesia. The current study proposes NPYFa as a potent, long-acting antinociceptor lacking tolerance development as well as a probe to study opioid analgesia and the associated complex mechanisms of tolerance development. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
36. Early changes of immunoreactivity to orexin in hypothalamus and to RFamide peptides in brainstem during the development of hypertension
- Author
-
Andries Kalsbeek, Ajda Yilmaz, Ruud M. Buijs, Endocrinology, Laboratory for Endocrinology, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, Amsterdam Gastroenterology Endocrinology Metabolism, and Netherlands Institute for Neuroscience (NIN)
- Subjects
Male ,Nervous system ,medicine.medical_specialty ,animal structures ,Lateral hypothalamus ,Hypothalamus ,Neuropeptide ,Baroreflex ,Autonomic Nervous System ,Rats, Inbred WKY ,Rats, Inbred SHR ,Internal medicine ,medicine ,Animals ,Neuropeptide FF ,Orexins ,business.industry ,General Neuroscience ,Neuropeptides ,Parasympathetic ,Rats ,Orexin ,Endocrinology ,Dorsal motor nucleus ,medicine.anatomical_structure ,nervous system ,Hypertension ,business ,And hypertension ,Brain Stem - Abstract
Baroreflex sensitivity (BRS) is an important function of the nervous system and essential for maintaining blood pressure levels in the physiological range. In hypertension, BRS is decreased both in man and animals. Although increased sympathetic activity is thought to be the main cause of decreased BRS, hence the development of hypertension, the BRS is regulated by both sympathetic (SNS) and parasympathetic (PNS) nervous system. Here, we analyzed neuropeptide changes in the lateral hypothalamus (LH), which favours the SNS activity, as well as in PNS nuclei in the brainstem of spontaneously hypertensive rats (SHR) and their normotensive controls (Wistar Kyoto rats- WKY). The analyses revealed that in the WKY rats the hypothalamic orexin system, known for its role in sympathetic activation, showed a substantial decrease when animals age. At the same time, however, such a decrease was not observed when hypertension developed in the SHR. In contrast, Neuropeptide FF (NPFF) and Prolactin Releasing Peptide (PrRP) expression in the PNS associated Nucleus Tractus Solitarius (NTS) and Dorsal Motor Nucleus of the Vagus (DMV) diminished substantially, not only after the establishment of hypertension but also before its onset. Therefore, the current results indicate early changes in areas of the central nervous system involved in SNS and PNS control of blood pressure and associated with the development of hypertension.
- Published
- 2021
37. Analgesic tolerance and cross-tolerance to the bifunctional opioid/neuropeptide FF receptors agonist EN-9 and μ-opioid receptor ligands at the supraspinal level in mice.
- Author
-
Han, Zhenglan, Jin, Guofei, Tang, Jiancai, Wang, Hanyan, Guo, Dongmei, and Zhang, Jingping
- Abstract
The chimeric peptide EN-9 was reported as a κ-opioid/neuropeptide FF receptors bifunctional agonist that modulated chronic pain with no tolerance. Many lines of evidence have shown that the effect of the κ-opioid receptor is mediated by not only its specific activation but also downstream events participation, especially interaction with the μ-opioid receptor pathway in antinociception and tolerance on most occasions. The present study investigated the acute and chronic cross-tolerance of EN-9 with μ-opioid receptor agonist EM-2, DAMGO, and morphine after intracerebroventricularly (i.c.v) injection in the mouse tail-flick test. In the acute tolerance test, EN-9 showed symmetrical acute cross-tolerance to DAMGO but no cross-tolerance to EM2. In the chronic tolerance test, EN-9 had no tolerance after eight days of repeated administration. However, EN-9 illustrated complete cross-tolerance to morphine and symmetrical cross-tolerance to EM2. In addition, inhibition of NPFF receptor could induce the tolerance development of EN-9. These findings indicated that supraspinal EN-9-induced antinociception contains additional components, which are mediated by the downstream μ-opioid receptor pathway both in acute and chronic treatment, whereas the subtypes of μ-opioid receptor or NPFF system pathway involved in antinociceptive effects induced by EN-9 are complex. Identifying the receptor mechanism could help design preferable bifunctional opioid compounds. • EN-9 is a new chimeric peptide exerted significant analgesic effect with non-tolerance. • EN-9 activates different pathways in acute and chronic treatment. • Identify the underlying mechanism of EN-9 could help design preferable bi-functional or/and multi-functional opioid compounds [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
38. Knockout of NPFFR2 Prevents LPS-Induced Depressive-Like Responses in Mice
- Author
-
Ya Tin Lin, Jin-Chung Chen, and Zachary Yu
- Subjects
Lipopolysaccharides ,Receptors, Neuropeptide ,0301 basic medicine ,Hippocampus ,Neuropeptide FF receptor ,Mice ,0302 clinical medicine ,neuropeptide FF ,Neuropeptide FF ,Biology (General) ,Receptor ,Spectroscopy ,Mice, Knockout ,Behavior, Animal ,Chemistry ,General Medicine ,Computer Science Applications ,Receptor, Serotonin, 5-HT1A ,depression ,Female ,Tumor necrosis factor alpha ,medicine.medical_specialty ,LPS ,QH301-705.5 ,Motor Activity ,Serotonergic ,Article ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,NPFFR2 ,Downregulation and upregulation ,Internal medicine ,medicine ,Animals ,Physical and Theoretical Chemistry ,Molecular Biology ,QD1-999 ,Tumor Necrosis Factor-alpha ,Organic Chemistry ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,serotonin 1A receptor (5-HT1AR) ,030217 neurology & neurosurgery ,Behavioural despair test - Abstract
The precise neural mechanisms underlying the pathogenesis of depression are largely unknown, though stress-induced brain inflammation and serotonergic plasticity are thought to be centrally involved. Moreover, we previously demonstrated that neuropeptide FF receptor 2 (NPFFR2) overexpression provokes depressive-like behaviors in mice. Here, we assess whether NPFFR2 is involved in priming of depressive-like behaviors and downregulation of serotonergic 1A receptor (5HT1AR) after lipopolysaccharide (LPS) treatment. The forced swimming test (FST) and sucrose preference test (SPT) were used to quantify depressive-like phenotypes in wild-type (WT) and NPFFR2-knockout (KO) mice. A single dose of LPS (i.p. 1 mg/kg) readily caused increases in toll-like receptor 4 and tumor necrosis factor-α along with decreases in 5-HT1AR mRNA in the ventral hippocampus of WT mice. Furthermore, LPS treatment of WT mice increased immobility time in FST and decreased sucrose preference in SPT. In contrast, none of these effects were observed in NPFFR2-KO mice. While WT mice injected with lentiviral 5-HT1AR shRNA in the ventral hippocampus displayed an unaltered response after LPS challenge, LPS-challenged NPFFR2-KO mice displayed a profound decrease in sucrose preference when pretreated with 5-HT1AR shRNA. Taken together, these results suggest that NPFFR2 modulates LPS-induced depressive-like behavioral phenotypes by downregulating 5HT1AR in the ventral hippocampus.
- Published
- 2021
- Full Text
- View/download PDF
39. Balanced release of neuropeptide FF and gonadotropin-releasing hormone 3 modulates male sexual behavior
- Author
-
Yasuhisa Akazome, Yoshitaka Oka, Yamamoto E, Yasunori Mori, Kenji Okubo, Shinji Kanda, Nobuaki Yoshida, and Chie Umatani
- Subjects
Preoptic area ,medicine.anatomical_structure ,Neuromodulation ,medicine ,Neuropeptide ,Terminal nerve ,Sensory system ,Gonadotropin-releasing hormone ,Neuropeptide FF ,Biology ,Indirect pathway of movement ,Neuroscience - Abstract
Animals properly perform sexual behaviors by using multiple sensory cues. However, neural mechanisms integrating multiple sensory cues and regulating motivation for sexual behaviors remain unclear. Here, we focused on peptidergic neurons, terminal nerve gonadotropin-releasing hormone (TN-GnRH) neurons, which receive inputs from various sensory systems and co-express neuropeptide FF (NPFF) in addition to GnRH. Our behavioral analyses using knockout medaka of GnRH (gnrh3) and/or NPFF (npff) demonstrated that some sexual behavioral repertories were ‘delayed’, not disrupted, in gnrh3-/- and npff-/- males, while the double knockout showed normal behaviors. We also found anatomical evidence to show that both neuropeptides modulate the sexual behavior-controlling brain areas. Furthermore, we demonstrated that NPFF activates neurons in the preoptic area via indirect pathway, which is considered to induce the increase in the motivation for male sexual behaviors. Considering these results, we propose a novel mechanism by which balanced release of co-existing peptides is important for the neuromodulatory function of TN-GnRH neurons in the control of behavioral motivation. Our results may go a long way toward understanding the functional significance of peptidergic neuromodulation in response to external environments.
- Published
- 2021
- Full Text
- View/download PDF
40. Palmitoylation of Prolactin-Releasing Peptide Increased Affinity for and Activation of the GPR10, NPFF-R2 and NPFF-R1 Receptors: In Vitro Study
- Author
-
Blanka Železná, Veronika Strnadová, Jaroslav Kuneš, Alena Karnošová, Lucie Holá, and Lenka Maletínská
- Subjects
Receptors, Neuropeptide ,medicine.medical_specialty ,GPR10 ,QH301-705.5 ,Prolactin-releasing peptide ,Lipoylation ,Neuropeptide ,CHO Cells ,In Vitro Techniques ,Catalysis ,Article ,prolactin-releasing peptide ,Receptors, G-Protein-Coupled ,Inorganic Chemistry ,Cricetulus ,neuropeptide FF ,Palmitoylation ,NPFF-R2 ,Internal medicine ,Cricetinae ,medicine ,NPFF-R1 ,Animals ,Humans ,Neuropeptide FF ,Physical and Theoretical Chemistry ,Biology (General) ,Receptor ,Molecular Biology ,Protein kinase B ,QD1-999 ,Spectroscopy ,Prolactin-Releasing Hormone ,Chemistry ,Organic Chemistry ,General Medicine ,Neuropeptide Y receptor ,signaling pathways ,Computer Science Applications ,Endocrinology ,binding properties ,Calcium ,Signal transduction - Abstract
The anorexigenic neuropeptide prolactin-releasing peptide (PrRP) is involved in the regulation of food intake and energy expenditure. Lipidization of PrRP stabilizes the peptide, facilitates central effect after peripheral administration and increases its affinity for its receptor, GPR10, and for the neuropeptide FF (NPFF) receptor NPFF-R2. The two most potent palmitoylated analogs with anorectic effects in mice, palm11-PrRP31 and palm-PrRP31, were studied in vitro to determine their agonist/antagonist properties and mechanism of action on GPR10, NPFF-R2 and other potential off-target receptors related to energy homeostasis. Palmitoylation of both PrRP31 analogs increased the binding properties of PrRP31 to anorexigenic receptors GPR10 and NPFF-R2 and resulted in a high affinity for another NPFF receptor, NPFF-R1. Moreover, in CHO-K1 cells expressing GPR10, NPFF-R2 or NPFF-R1, palm11-PrRP and palm-PrRP significantly increased the phosphorylation of extracellular signal-regulated kinase (ERK), protein kinase B (Akt) and cAMP-responsive element-binding protein (CREB). Palm11-PrRP31, unlike palm-PrRP31, did not activate either c-Jun N-terminal kinase (JNK), p38, c-Jun, c-Fos or CREB pathways in cells expressing NPFF-1R. Palm-PrRP31 also has higher binding affinities for off-target receptors, namely, the ghrelin, opioid (KOR, MOR, DOR and OPR-L1) and neuropeptide Y (Y1, Y2 and Y5) receptors. Palm11-PrRP31 exhibited fewer off-target activities, therefore, it has a higher potential to be used as an anti-obesity drug with anorectic effects.
- Published
- 2021
41. Identification of an N -acylated- D Arg-Leu-NH 2 Dipeptide as a Highly Selective Neuropeptide FF1 Receptor Antagonist That Potently Prevents Opioid-Induced Hyperalgesia
- Author
-
Patrick Gizzi, Brigitte Ilien, Raphaëlle Quillet, Valérie Kugler, Frédéric Simonin, Valérie Simonneaux, Martine Schmitt, Frédéric Bihel, Khadija Elhabazi, Valérie Utard, Jo B. Henningsen, Armand Drieu la Rochelle, Séverine Schneider, Biotechnologie et signalisation cellulaire (BSC), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Institut de recherche de l'Ecole de biotechnologie de Strasbourg (IREBS), Laboratoire d'Innovation Thérapeutique (LIT), Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Institut de Chimie du CNRS (INC), Institut des Neurosciences Cellulaires et Intégratives (INCI), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS), Plate-forme de chimie biologique intégrative de Strasbourg (PCBiS), Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA), ANR-17-EURE-0022,EURIDOL,Ecole Universitaire de Recherche Interdisciplinaire sur la Douleur(2017), and ANR-10-LABX-0034,Medalis,Medalis Drug Discovery Center(2010)
- Subjects
0303 health sciences ,Chemistry ,medicine.drug_class ,Antagonist ,Chronic pain ,Neuropeptide ,Pharmacology ,Receptor antagonist ,medicine.disease ,01 natural sciences ,3. Good health ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,03 medical and health sciences ,Drug Discovery ,Hyperalgesia ,medicine ,Molecular Medicine ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Neuropeptide FF ,medicine.symptom ,Receptor ,Opioid-induced hyperalgesia ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology - Abstract
RFamide-related peptide-3 (RFRP-3) and neuropeptide FF (NPFF) target two different receptor subtypes called neuropeptide FF1 (NPFF1R) and neuropeptide FF2 (NPFF2R) that modulate several functions. However, the study of their respective role is severely limited by the absence of selective blockers. We describe here the design of a highly selective NPFF1R antagonist called RF3286, which potently blocks RFRP-3-induced hyperalgesia in mice and luteinizing hormone release in hamsters. We then showed that the pharmacological blockade of NPFF1R in mice prevents the development of fentanyl-induced hyperalgesia while preserving its analgesic effect. Altogether, our data indicate that RF3286 represents a useful pharmacological tool to study the involvement of the NPFF1R/RFRP-3 system in different functions and different species. Thanks to this compound, we showed that this system is critically involved in the development of opioid-induced hyperalgesia, suggesting that NPFF1R antagonists might represent promising therapeutic tools to improve the use of opioids in the treatment of chronic pain.
- Published
- 2021
- Full Text
- View/download PDF
42. NPFFR2-deficient mice fed a high-fat diet develop strong intolerance to glucose.
- Author
-
Karnošová A, Strnadová V, Železná B, Kuneš J, Kašpárek P, and Maletínská L
- Subjects
- Animals, Female, Male, Mice, Adipose Tissue metabolism, Adipose Tissue, White metabolism, Diet, High-Fat, Glucose metabolism, Liver metabolism, Mice, Inbred C57BL, Mice, Knockout, Obesity metabolism, Glucose Intolerance metabolism, Insulin Resistance
- Abstract
A previous study on neuropeptide FF receptor 2 (NPFFR2)-deficient mice has demonstrated that NPFFR2 is involved in the control of energy balance and thermogenesis. Here, we report on the metabolic impact of NPFFR2 deficiency in male and female mice that were fed either a standard diet (STD) or a high-fat diet (HFD) and each experimental group consisted of ten individuals. Both male and female NPFFR2 knockout (KO) mice exhibited severe glucose intolerance that was exacerbated by a HFD diet. In addition, reduced insulin pathway signaling proteins in NPFFR2 KO mice fed a HFD resulted in the development of hypothalamic insulin resistance. HFD feeding did not cause liver steatosis in NPFFR2 KO mice of either sex, but NPFFR2 KO male mice fed a HFD had lower body weights, white adipose tissues, and liver and lower plasma leptin levels compared with their wild-type (WT) controls. Lower liver weight in NPFFR2 KO male mice compensated for HFD-induced metabolic stress by increased liver PPARα and plasma FGF21 hepatokine, which supported fatty acid β-oxidation in the liver and white adipose tissue. Conversely, NPFFR2 deletion in female mice attenuated the expression of Adra3β and Pparγ, which inhibited lipolysis in adipose tissue., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
43. Cholesterol-rich lipid rafts are involved in neuropeptide FF anti-nociceptin/orphanin FQ effect.
- Author
-
Ding, Zhong and Zajac, Jean‐Marie
- Subjects
- *
LIPID rafts , *NOCICEPTIN , *NEUROPEPTIDES , *G protein coupled receptors , *PEPTIDE receptors - Abstract
The participation of a signaling platform to the anti-nociceptin/orphanin FQ (N/ OFQ) effect of neuropeptide FF ( NPFF) receptors was investigated in both acutely dissociated neurons and SH- SY5Y human neuroblastoma cells. The NPFF anti-N/ OFQ, not anti-μ opioid effect, on the Ca2+ transient triggered by depolarization was reversed by methyl-β-cyclodextrin which depletes cholesterol from cell membranes. While the inactive α-cyclodextrin had no effect. By using [35S] GTPγS binding assay, a significant 20% decrease in the activity of nociceptin/orphanin FQ peptide receptors induced by the NPFF analog 1 DMe was observed in detergent-resistant membranes, but not in total membranes of SH- SY5Y cells. Moreover, si RNA knock-down of G-protein-coupled receptor kinase 2 indicated that G-protein-coupled receptor kinase 2, but not protein kinase C, acted as the mediator in the NPFF anti-N/ OFQ process. These data indicate that cholesterol-rich lipid rafts play an important role in the anti-N/ OFQ effect of NPFF receptors. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
44. Effects of neuropeptide FF and related peptides on the antinociceptive activities of VD-hemopressin(α) in naive and cannabinoid-tolerant mice.
- Author
-
Pan, Jia-Xin, Wang, Zi-Long, Li, Ning, Zhang, Nan, Wang, Pei, Tang, Hong-Hai, Zhang, Ting, Yu, Hong-Ping, Zhang, Run, Zheng, Ting, Fang, Quan, and Wang, Rui
- Subjects
- *
NEUROPEPTIDES , *ANALGESICS , *CANNABINOID receptors , *LABORATORY mice , *ANALGESIA - Abstract
Neuropeptide FF (NPFF) system has recently been reported to modulate cannabinoid-induced antinociception. The aim of the present study was to further investigate the roles of NPFF system in the antinociceptive effects induced by intracerebroventricular (i.c.v.) administration of mouse VD-hemopressin(α), a novel endogenous agonist of cannabinoid CB 1 receptor, in naive and VD-hemopressin(α)-tolerant mice. The effects of NPFF system on the antinociception induced by VD-hemopressin(α) were investigated in the radiant heat tail-flick test in naive mice and VD-hemopressin(α)-tolerant mice. The cannabinoid-tolerant mice were produced by given daily injections of VD-hemopressin(α) (20 nmol, i.c.v.) for 5 days and the antinociception was measured on day 6. In naive mice, intracerebroventricular injection of NPFF dose-dependently attenuated central analgesia of VD-hemopressin(α). In contrast, neuropeptide VF (NPVF) and D .NP( N -Me)AFLFQPQRF-NH 2 (dNPA), two highly selective agonists for Neuropeptide FF 1 and Neuropeptide FF 2 receptors, enhanced VD-hemopressin(α)-induced antinociception in a dose-dependent manner. In addition, the VD-hemopressin(α)-modulating activities of NPFF and related peptides were antagonized by the Neuropeptide FF receptors selective antagonist 1-adamantanecarbonyl-RF-NH 2 (RF9). In VD-hemopressin(α)-tolerant mice, NPFF failed to modify VD-hemopressin(α)-induced antinociception. However, both neuropeptide VF and dNPA dose-dependently potentiated the antinociception of VD-hemopressin(α) and these cannabinoid-potentiating effects were reduced by RF9. The present works support the cannabinoid-modulating character of NPFF system in naive and cannabinoid-tolerant mice. In addition, the data suggest that a chronic cannabinoid treatment modifies the pharmacological profiles of NPFF, but not the cannabinoid-potentiating effects of neuropeptide VF and dNPA. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
45. Neuropeptide FF receptors as novel targets for limbic seizure attenuation.
- Author
-
Portelli, Jeanelle, Meurs, Alfred, Bihel, Frederic, Hammoud, Hassan, Schmitt, Martine, De Kock, Joery, Utard, Valerie, Humbert, Jean-Paul, Bertin, Isabelle, Buffel, Ine, Coppens, Jessica, Tourwe, Dirk, Maes, Veronique, De Prins, An, Vanhaecke, Tamara, Massie, Ann, Balasubramaniam, Ambikaipakan, Boon, Paul, Bourguignon, Jean-Jacques, and Simonin, Frederic
- Subjects
- *
NEUROPEPTIDE Y receptors , *LIMBIC system injuries , *ANTICONVULSANTS , *TREATMENT of epilepsy , *LABORATORY rats , *DRUG administration - Abstract
Neuropeptide Y (NPY) is a well established anticonvulsant and first-in-class antiepileptic neuropeptide. In this study, the controversial role of NPY 1 receptors in epilepsy was reassessed by testing two highly selective NPY 1 receptor ligands and a mixed NPY 1 /NPFF receptor antagonist BIBP3226 in a rat model for limbic seizures. While BIBP3226 significantly attenuated the pilocarpine-induced seizures, neither of the highly selective NPY 1 receptor ligands altered the seizure severity. Administration of the NPFF1/NPFF2 receptor antagonist RF9 also significantly attenuated limbic seizure activity. To further prove the involvement of NPFF receptors in these seizure-modulating effects, low and high affinity antagonists for the NPFF receptors were tested. We observed that the low affinity ligand failed to exhibit anticonvulsant properties while the two high affinity ligands significantly attenuated the seizures. Continuous NPFF1 receptor agonist administration also inhibited limbic seizures whereas bolus administration of the NPFF1 receptor agonist was without effect. This suggests that continuous agonist perfusion could result in NPFF1 receptor desensitization and mimic NPFF1 receptor antagonist administration. Our data unveil for the first time the involvement of the NPFF system in the management of limbic seizures. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
46. Neuropeptide FF and prolactin-releasing peptide decrease cortical excitability through activation of NPFF receptors.
- Author
-
Buffel, Ine, Meurs, Alfred, Portelli, Jeanelle, Raedt, Robrecht, De Herdt, Veerle, Sioncke, Lynn, Wadman, Wytse, Bihel, Frederic, Schmitt, Martine, Vonck, Kristl, Bourguignon, Jean‐Jacques, Simonin, Frederic, Smolders, Ilse, and Boon, Paul
- Subjects
- *
NEUROPEPTIDES , *PROLACTIN , *MOTOR cortex physiology , *TREATMENT of epilepsy , *NEURAL stimulation , *THERAPEUTICS - Abstract
Objective Drugs with a novel mechanism of action are needed to reduce the number of people with epilepsy that are refractory to treatment. Increasing attention is paid to neuropeptide systems and several anticonvulsant neuropeptides have already been described, such as galanin, ghrelin, and neuropeptide Y ( NPY). Many others, however, have not been investigated for their ability to affect epileptic seizures. In this study, the potential anticonvulsant activities of three members of the RF-amide neuropeptide family, neuropeptide FF ( NPFF), prolactin-releasing peptide (Pr RP), and kisspeptin (Kp) and other receptor ligands ( NPFF1/2R, GPR10, and GRP54, respectively) were tested in the motor cortex stimulation model. Methods A train of pulses with increasing intensity (0-10 mA over 150 s, 50 Hz, pulse width 2 msec) was delivered to the motor cortex of rats. The threshold intensity for eliciting a motor response (i.e., motor threshold) was determined through behavioral observation and used as a measure for cortical excitability. The threshold was determined before, during, and after the intracerebroventricular (i.c.v.) administration of various NPFF1/2R, GPR10, and GPR54 receptor ligands. Results NPFF and Pr RP significantly increased the motor threshold by a maximum of 143 ± 27 and 83 ± 13 μA, respectively, for the doses of 1 nmol/h (p < 0.05). The increase of motor threshold by NPFF and Pr RP was prevented by pretreatment and co-treatment with the NPFF1/2R antagonist RF9. Pretreatment with a selective NPFF1R antagonist also prevented the threshold increase induced by NPFF. Kp did not increase motor threshold. Significance Intracerebroventricular infusion of NPFF or Pr RP decreases cortical excitability in rats through activation of NPFFRs. Furthermore, the NPFF1R is required for the NPFF-induced decrease in cortical excitability. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
47. Transcriptomic Changes in Mouse Bone Marrow-Derived Macrophages Exposed to Neuropeptide FF
- Author
-
Zhuo Zuo, Yulong Sun, and Yuanyuan Kuang
- Subjects
Receptors, Neuropeptide ,0301 basic medicine ,Bone marrow-derived macrophage ,QH426-470 ,Article ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Genetics ,Animals ,Neuropeptide FF ,KEGG ,Gene ,neuropeptide ,Cells, Cultured ,Genetics (clinical) ,Binding Sites ,Chemistry ,Macrophages ,RNA ,MODELLER ,RNA sequencing ,bioinformatics ,bone marrow-derived macrophage ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Oligopeptides ,030217 neurology & neurosurgery ,transcriptomic profiles ,Protein Binding - Abstract
Neuropeptide FF (NPFF) is a neuropeptide that regulates various biological activities. Currently, the regulation of NPFF on the immune system is an emerging field. However, the influence of NPFF on the transcriptome of primary macrophages has not been fully elucidated. In this study, the effect of NPFF on the transcriptome of mouse bone marrow-derived macrophages (BMDMs) was explored by RNA sequencing, bioinformatics, and molecular simulation. BMDMs were treated with 1 nM NPFF for 18 h, followed by RNA sequencing. Differentially expressed genes (DEGs) were obtained, followed by GO, KEGG, and PPI analysis. A total of eight qPCR-validated DEGs were selected as hub genes. Subsequently, the three-dimensional (3-D) structures of the eight hub proteins were constructed by Modeller and Rosetta. Next, the molecular dynamics (MD)-optimized 3-D structure of hub protein was acquired with Gromacs. Finally, the binding modes between NPFF and hub proteins were studied by Rosetta. A total of 2655 DEGs were obtained (up-regulated 1442 vs. down-regulated 1213), and enrichment analysis showed that NPFF extensively regulates multiple functional pathways mediated by BMDMs. Moreover, the 3-D structure of the hub protein was obtained after MD-optimization. Finally, the docking modes of NPFF-hub proteins were predicted. Besides, NPFFR2 was expressed on the cell membrane of BMDMs, and NPFF 1 nM significantly activated NPFFR2 protein expression. In summary, instead of significantly inhibiting the expression of the immune-related gene transcriptome of RAW 264.7 cells, NPFF simultaneously up-regulated and down-regulated the gene expression profile of a large number of BMDMs, hinting that NPFF may profoundly affect a variety of cellular processes dominated by BMDMs. Our work provides transcriptomics clues for exploring the influence of NPFF on the physiological functions of BMDMs.
- Published
- 2021
48. Activation of MrgprA3 and MrgprC11 on Bladder-Innervating Afferents Induces Peripheral and Central Hypersensitivity to Bladder Distension
- Author
-
Joel Castro, Ashlee Caldwell, Sonia Garcia-Caraballo, Luke Grundy, Xinzhong Dong, David Grundy, Nick J. Spencer, Stuart M. Brierley, and Andrea M. Harrington
- Subjects
0301 basic medicine ,Agonist ,Male ,Pathology ,medicine.medical_specialty ,Patch-Clamp Techniques ,medicine.drug_class ,Lumbosacral Plexus ,Urinary Bladder ,TRPV1 ,TRPV Cation Channels ,Distension ,Pharmacology ,urologic and male genital diseases ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,Mice ,Calcium imaging ,0302 clinical medicine ,Dorsal root ganglion ,Ganglia, Spinal ,Physical Stimulation ,medicine ,Animals ,Patch clamp ,Neuropeptide FF ,Neurons, Afferent ,Receptor ,Research Articles ,Mice, Knockout ,business.industry ,General Neuroscience ,Interstitial cystitis ,medicine.disease ,Spinal cord ,Mice, Inbred C57BL ,Posterior Horn Cells ,030104 developmental biology ,medicine.anatomical_structure ,Overactive bladder ,Female ,business ,030217 neurology & neurosurgery - Abstract
Understanding the sensory mechanisms innervating the bladder is paramount to developing efficacious treatments for chronic bladder hypersensitivity conditions. The contribution of Mas-gene-related G protein-coupled receptors (Mrgpr) to bladder signalling is currently unknown. Here we show in mice with single-cell RT-PCR that sub-populations of dorsal root ganglion (DRG) neurons innervating the mouse bladder express MrgprA3 (14%) and MrgprC11 (38%), either individually or in combination, with high levels of co-expression with Trpv1 (81-89%). Calcium imaging studies demonstrated MrgprA3 and MrgprC11 agonists (chloroquine, BAM8-22 and neuropeptide FF) activated sub-populations of bladder-innervating DRG neurons, showing functional evidence of co-expression between MrgprA3, MrgprC11 and TRPV1. In ex vivo bladder-nerve preparations chloroquine, BAM8-22 and neuropeptide FF all evoked mechanical hypersensitivity in sub-populations (20-41%) of bladder afferents. These effects were absent in recordings from Mrgpr-clusterΔ−/− mice. In vitro whole-cell patch clamp recordings showed that application of an MrgprA3/C11 agonist cocktail induced neuronal hyper-excitability in 44% of bladder-innervating DRG neurons. Finally, in vivo instillation of an MrgprA3/C11 agonist cocktail into the bladder of wild-type mice induced a significant activation of dorsal horn neurons within the lumbosacral spinal cord, as quantified by pERK-immunoreactivity. This MrgprA3/C11 agonist-induced activation was particularly apparent within the superficial dorsal horn and the sacral parasympathetic nuclei of wild-type, but not Mrgpr-clusterΔ−/− mice. This study demonstrates, for the first time, functional expression of MrgprA3 and MrgprC11 in bladder afferents. Activation of these receptors is not required for normal bladder function but does trigger hypersensitivity to distension, a critically valuable factor for therapeutic target development.Significance statementDetermining how bladder afferents become sensitized is the first step in finding effective treatments for common urological disorders such as overactive bladder and interstitial cystitis/bladder pain syndrome. Here we show that two of the key receptors, MrgprA3 and MrgprC11, that mediate itch from the skin are also expressed on afferents innervating the bladder. Activation of these receptors results in sensitization of bladder afferents, resulting in sensory signals being sent into the spinal cord that prematurely indicate bladder fullness. Targeting bladder afferents expressing MrgprA3 or MrgprC11 and preventing their sensitisation may provide a novel approach for treating overactive bladder and interstitial cystitis/bladder pain syndrome.
- Published
- 2021
49. MCRT, a multifunctional ligand of opioid and neuropeptide FF receptors, attenuates neuropathic pain in spared nerve injury model
- Author
-
Jing Zhang, Xiaoli Wang, Hao Wang, Yaofeng Zhao, Jagat Narayan Shah, Chan Ma, Lanxia Zhou, Hailan Li, Shouliang Dong, Chunbo He, Wenting Su, Shasha Chen, and Zhe Zhang
- Subjects
Receptors, Neuropeptide ,Narcotic Antagonists ,Analgesic ,Pregabalin ,Receptors, Opioid, mu ,Pharmacology ,Toxicology ,Ligands ,Mice ,medicine ,Animals ,Neuropeptide FF ,Morphine ,business.industry ,General Medicine ,Nerve injury ,Analgesics, Opioid ,Opioid ,Hyperalgesia ,Neuropathic pain ,Receptors, Opioid ,Neuralgia ,Endorphins ,medicine.symptom ,business ,medicine.drug - Abstract
Chimeric peptide MCRT (YPFPFRTic-NH2 ) was a multifunctional ligand of opioid and neuropeptide FF (NPFF) receptors and reported to be potentially antalgic in acute tail-flick test. Here, we developed spared nerve injury (SNI) model to explore its efficacy in chronic neuropathic pain. Analgesic tolerance, opioid-induced hyperalgesia and gastrointestinal transit were measured for safety evaluation. Intracerebroventricular (i.c.v.) and intraplantar (i.pl.) injections were conducted as central and peripheral routes, respectively. Results demonstrated that MCRT alleviated neuropathic pain effectively and efficiently, with the ED50 values of 4.93 nmol/kg at the central level and 3.11 nmol/kg at the peripheral level. The antagonist blocking study verified the involvement of mu-, delta-opioid and NPFF receptors in MCRT produced anti-allodynia. Moreover, the separation of analgesia from unwanted effects was preliminarily achieved and that MCRT caused neither analgesic tolerance nor hyperalgesia after chronic i.c.v. administration, nor constipation after i.pl. administration. Notably, the local efficacy of MCRT in SNI mice was about one thousandfold higher than morphine and ten thousandfold higher than pregabalin, indicating a great promise in the future treatment of neuropathic pain.
- Published
- 2021
50. Elevated Serum Neuropeptide FF Levels Are Associated with Cognitive Decline in Patients with Spinal Cord Injury
- Author
-
Meng Yan, Yuanzhen Chen, Shifei Sun, Bin Shi, and Shilong Sun
- Subjects
Male ,medicine.medical_specialty ,Medicine (General) ,Article Subject ,Clinical Biochemistry ,Neuropeptide ,Gastroenterology ,R5-920 ,Internal medicine ,Genetics ,Medicine ,Humans ,Cognitive Dysfunction ,Neuropeptide FF ,Prospective Studies ,Cognitive decline ,Prospective cohort study ,Molecular Biology ,Spinal cord injury ,Spinal Cord Injuries ,Triiodothyronine ,business.industry ,Biochemistry (medical) ,Montreal Cognitive Assessment ,General Medicine ,Middle Aged ,medicine.disease ,Prognosis ,Case-Control Studies ,Female ,Analysis of variance ,business ,Oligopeptides ,Biomarkers ,Research Article ,Follow-Up Studies - Abstract
Background. Spinal cord injury (SCI) has high incidence globally and is frequently accompanied by subsequent cognitive decline. Accurate early risk-categorization of SCI patients for cognitive decline using biomarkers can enable the timely application of appropriate neuroprotective measures and the development of new agents for the management of SCI-associated cognitive decline. Neuropeptide FF is an endogenous neuropeptide with a multitude of functions and is associated with neuroinflammatory processes. This prospective study investigated the predictive value of serum neuropeptide FF levels measured after acute SCI for subsequent cognitive decline. Methods. 88 patients presenting with acute SCI without preexisting neurological injury, brain trauma, or severe systemic illness and 60 healthy controls were recruited. Serum neuropeptide FF levels, clinical, and routine laboratory variables including low-density lipoprotein, high-density lipoprotein, fasting blood glucose, total triiodothyronine (TT3), total thyroxine (TT4), and thyroid-stimulating hormone (TSH) levels collected from all subjects were assessed. Montreal cognitive assessment (MoCA) was performed 3 months after enrollment. SCI patients were grouped according to quartile of serum neuropeptide FF level and MoCA scores were compared using ANOVA. Additionally, multivariate linear regression with clinical and laboratory variables was performed to predict MoCA scores. Results. SCI patients displayed significantly higher baseline serum neuropeptide FF levels than healthy controls ( 38.5 ± 4.1 versus 23.4 ± 2.0 pg / ml , p < 0.001 ∗ ∗ ). SCI patients in higher quartiles of baseline serum neuropeptide FF displayed significantly lower MoCA scores at 3 months. Linear regression analysis indicated serum neuropeptide FF levels as a significant independent predictor of worse MoCA scores after SCI ( r = 0.331 , p = 0.034 ∗ ). Conclusion. Early serum neuropeptide FF levels significantly and independently predicted cognitive decline after acute SCI among patients without preexisting neurological disorders.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.