echemi logo
Product
  • Product
  • Supplier
  • Inquiry
    Home > Active Ingredient News > Study of Nervous System > Current Biology︱Chen Zhong's team made new achievements in the mechanism of histamine regulation of feeding: H2 receptor-dependent medial septal nucleus histaminergic circuit

    Current Biology︱Chen Zhong's team made new achievements in the mechanism of histamine regulation of feeding: H2 receptor-dependent medial septal nucleus histaminergic circuit

    • Last Update: 2022-04-28
    • Source: Internet
    • Author: User
    Search more information of high quality chemicals, good prices and reliable suppliers, visit www.echemi.com

    Written by ︱Xu Lingyu Edited ︱Wang Sizhen Obesity is a disease caused by excessive accumulation of fat in the body, which can cause damage to liver and kidney function and increase the probability of type II diabetes, cardiovascular and cerebrovascular diseases, and cancer.
    It poses a threat to the health of more than 600 million people worldwide and causes approximately four million deaths worldwide each year [1, 2]
    .

    Unfortunately, there is currently a lack of safe and effective control of food intake in clinical practice.
    Drugs used to treat obesity in clinical practice can easily cause side effects such as vomiting and arrhythmia, and even affect mood and cognitive function, leading to anxiety, depression and drug dependence.
    problems and poor patient compliance [3–5]
    .

    The neural circuits and molecular mechanisms of feeding behavior regulation have not been fully elucidated, which is an important reason for restricting the development of anti-obesity drugs
    .

     On March 25, 2022, Professor Zhong Chen's team from the School of Pharmacy, Zhejiang University published a paper titled "An H2R-dependent medial septum histaminergic circuit mediates feeding behavior" in Current Biology, a sub-journal of Cell, reporting that histamine regulates feeding behavior.
    The latest research results in the mechanism
    .

    This study used optogenetics, chemical genetics, pharmacology, combined with food-borne obesity model and genetic intervention and other technologies, and found for the first time that the direct projection of histaminergic nerves from the papillary nucleus of the hypothalamus to the medial septal nucleus can be bidirectional.
    In regulating feeding behavior, histamine plays a role in inhibiting feeding and regulating body weight by activating H2 receptors on glutamatergic neurons in the medial septal nucleus
    .

     Histamine is an important neurotransmitter and neuromodulator, mainly synthesized and released by histaminergic neurons
    .

    Histaminergic neurons originate from the tuberomammillary nucleus (TMN) of the hypothalamus, project widely to the whole brain, and play a regulatory role in various brain functions, such as sleep arousal, learning and memory, feeding behavior, etc.
    [6]
    .

    Early studies suggest that histaminergic nerves inhibit feeding behavior mainly by acting on the ventromedial hypothalamus or the paraventricular nucleus of the hypothalamus [7–9]
    .

    In recent years, more and more attention has been paid to the role of brain regions other than the hypothalamus in the regulation of feeding behavior
    .

    Some studies have found that the food-seeking behavior of starving rats is reduced after TMN damage, and at the same time, neuronal activation in multiple regions of the cortex, ventral basal forebrain, and dorsolateral tegmental area is inhibited [10, 11]
    .

    However, the specific functional characteristics and neural circuit mechanisms of histaminergic neurons in regulating feeding behavior have not been fully elucidated for a long time
    .

    Therefore, it is very necessary to further analyze the neural circuit mechanism of histaminergic nerves in the regulation of feeding behavior and find safe and effective drug intervention targets
    .

     To this end, the research group first constructed HDC-CreERT2 mice, injected AAV-CAG-Flex-ArchT-EGFP virus into the TMN of the mice, and used whole-brain microscopic optical section tomography (fMOST) to detect histaminergic Whole-brain three-dimensional reconstruction of neural projections revealed that the density of histaminergic nerve fibers was high in the medial septal nucleus
    .

    Recent studies have found that the medial septum (MS) plays an important role in the regulation of feeding behavior, and three types of neurons (cholinergic, glutamatergic, GABAergic, and GABAergic) in MS are regulated by optogenetics or pharmacogenetics.
    Neurons) can cause significant changes in food intake [12–14], but the upstream regulatory mechanism remains unclear, and the molecular targets regulating MS function have not been reported
    .

    So, is there a direct functional connection between histaminergic nerves and MS and regulate feeding behavior through MS? To this end, we used in vivo calcium signal optical fiber recording combined with histamine fluorescent probe to monitor the calcium signal of TMN histaminergic neuron cell bodies and the changes of MS histamine content in real time during the feeding process of mice
    .

    The results showed that when the mice ate, the calcium signal of histaminergic neurons and the content of MS histamine decreased concomitantly (Figure 1)
    .

    Further use of optogenetics to specifically inhibit histaminergic nerve endings in MS significantly increased food intake in mice, but specific activation of histaminergic nerve endings in MS significantly inhibited food intake (Fig.
    2)
    .

    Fig.
    1 Calcium signal of histaminergic neurons and histamine content in MS decreased concomitantly when mice ingested food (Source: Xu L, et al.
    , Current Biology, 2022)Fig.
    2 Activation or inhibition of TMNHA-MS circuit Feeding can be regulated bidirectionally (Credit: Xu L, et al.
    , Current Biology, 2022) To clarify the direct projection of histaminergic neurons to MS, this study used the immune panel after optogenetic activation of MS histaminergic nerve endings.
    The expression of neuronal activity marker cfos in MS was detected by chemical analysis, and the results showed that the activated neurons in MS were mainly glutamatergic neurons after activating MS histaminergic nerve endings
    .

    And the results of rabies virus (RV) cross-synaptic reversal and calcium signal test results show that MS glutamatergic neurons and TMN histaminergic neurons have dual functional and structural connections
    .

    Furthermore, pharmacogenetic inhibition of MS glutamatergic neuron activity could reverse the inhibitory effect of activation of MS histaminergic nerve endings on feeding behavior (Fig.
    3)
    .

    The above results suggest that there is a direct functional connection between TMN histaminergic neurons and MS glutamatergic neurons, and direct regulation of this projection can regulate feeding behavior in both directions
    .

    Figure 3 The main downstream projections of TMNHA-MS are MS glutamatergic neurons (Source: Xu L, et al.
    , Current Biology, 2022) Then, the histaminergic neuronal projections from TMN to MS regulate feeding sub-receptors What is the type? Early studies using lateral ventricle injection or local injection of histamine H1 or H2 receptor antagonists in the hypothalamus found that only H1 receptor antagonists can promote feeding behavior in rats, so the role of histamine in regulating feeding behavior has long been considered to be a Mediated by H1 receptors, H2 receptors are not involved in the regulation of feeding behavior
    .

    Unexpectedly, this study is the first to show that the feeding inhibition induced by optogenetic activation of MS histaminergic nerve endings can be abrogated by H2 receptors, but not by H1 receptor antagonists (Fig.
    4)
    .

    At the same time, antagonizing HCN channel antagonism downstream of H2 with the HCN channel (ie, hyperpolarization-activated cyclic nucleotide-gated channel) blocker ZD7288 can also reverse the inhibitory effect of activating this pathway, suggesting that histamine in MS activates H2 by activating H2.
    , rather than H1 receptors that play a role in regulating feeding behavior
    .

     In order to verify whether H2 receptors in MS glutamatergic neurons specifically mediate feeding behavior, in situ hybridization and immunofluorescence techniques were used to detect H2 receptors in different types of MS neurons in food-induced obese mice.
    The results showed that compared with normal-weight mice, the level of H2 receptors in MS glutamatergic neurons of food-induced obese mice was significantly decreased, but the levels of MS cholinergic neurons and GABAergic neurons were significantly reduced.
    There were no significant changes in H2 receptor levels (Figure 4), suggesting that H2 receptors in MS may have a cell-specific role in the regulation of feeding behavior
    .

    Figure 4 TMNHA-MS loop mainly regulates feeding behavior through downstream H2R receptors (Source: Xu L, et al.
    , Current Biology, 2022) Further, in order to explore whether H2 receptors in MS glutamatergic neurons associated with obesity
    .

    In this study, Cre-Loxp technology was used to inject conditional H2 receptor knockdown virus into MS vglut2-Cre mice to specifically knock down H2 receptors in MS glutamatergic neurons.
    The results also showed that: specific knockdown H2 receptors in MS glutamatergic neurons can promote weight gain in mice in a food-borne obesity model; and long-term administration of the clinical H2R agonist amthamine through the micro-pump drug delivery system can also effectively delay body weight in obese mice growth (Figure 5)
    .

    The above results suggest that H2 receptors in MS glutamatergic neurons can further regulate the activity of glutamatergic neurons and participate in the occurrence of obesity
    .

    Figure 5 Specific knockdown or activation of H2 receptors on MS glutamatergic neurons can bidirectionally regulate body weight in obese mice (Source: Xu L, et al.
    , Current Biology, 2022) Conclusions and discussions of the article, inspired by In conclusion, this study is the first to find that direct projections of histaminergic nerves from the tubercular papillary nucleus (TMN) to the medial septal nucleus (MS) can bidirectionally regulate feeding behavior, and histamine activates glutamatergic neurons in MS glutamatergic neurons.
    H2 receptors act to inhibit food intake and ultimately regulate body weight
    .

    This is the first time that H2 receptors have been reported to play an important role in the regulation of feeding and body weight in specific neural circuits, while the role of histamine receptors specific to other neuronal circuits remains to be further explored
    .

    This study will help to elucidate the neural circuits and molecular mechanisms of histaminergic neurons in regulating feeding behavior, provide potential new targets for drug intervention for the safe and effective regulation of feeding behavior in obese patients, and provide intervention for clinical treatment of obesity new strategy
    .

    Link to the original text: https://doi.
    org/10.
    1016/j.
    cub.
    2022.
    03.
    010 The first author of this research paper is Xu Lingyu, a doctoral student at Zhejiang University School of Pharmacy, and Lin Wenkai, a doctoral student at Zhejiang University School of Pharmacy, is the co-first author.
    Prof.
    Zhong is the only corresponding author of this article
    .

    This study was also supported and helped by Professor Li Yulong of Peking University.

    .

    The first author Xu Lingyu (the sixth from the left in the lower row), the co-first author Lin Wenkai (the fifth from the left in the upper row), and the corresponding author Professor Chen Zhong (the eighth from the right in the upper row) (Photo courtesy of Zhejiang University) Professor Chen Zhong's laboratory) Professor Chen Zhong's research group has long been engaged in the analysis of the pathogenesis of chronic encephalopathy and the research on new drug targets[15–19]
    .

    Among them, the neuropharmacological function of histaminergic neurons in the brain is one of the important directions of the research group.
    A series of previous studies have found that histamine is involved in schizophrenia, ischemic brain injury and other central nervous system diseases.
    Mechanism of action, published papers in well-known journals such as Nature Communications, Stem cell reports, Journal of Experimental Medicine [20–23], and was invited to write a review in the internationally renowned journal Pharmacology & Therapeutics [24], summarizing the central histamine and its effects.
    The latest research progress of the body
    .

    Selected Previous Articles【1】Front Cell Neurosci︱Shi Peng/Liu Zhen’s research group collaborated to reveal the common molecular mechanism of sensorineural hearing loss caused by multiple factors【21】Cell Death Dis︱Li Xian’s research group revealed the pre-oligodendrocyte The role of somatic ferroptosis in white matter damage after intracerebral hemorrhage [3] Front Mol Neurosci︱ Gao Shangbang's research group analyzes the composition and molecular mechanism of motor neuron oscillators [4] Nat Neurosci Review︱ Two-photon holographic optogenetics detection Neural coding【5】Mol Psychiatry︱The role of biological clock gene Bmal1 in a mouse model of autism and cerebellar ataxia 【6】Science︱Rapid eye movement sleep in mice is regulated by dopamine signaling in the basolateral amygdala【7】Nat Neurosci︱ Down-regulation of neuroimmunity and synapse-related pathways in the amygdala and anterior cingulate gyrus in patients with bipolar disorder [8] Nat Commun︱ Zhou Xiaoming/Sun Ziyi's team revealed the molecules of its ligand entry pathway based on the open conformation of Sigma-1 receptor Mechanism【9】Glia︱Yuan Jianqiang’s group reveals a new mechanism for regulating the proliferation of oligodendrocyte precursor cells: c-Abl phosphorylates Olig2【10】HBM︱Yu Lianchun’s group reveals the relationship between brain avalanche criticality and fluid intelligence and working memory Relevant high-quality scientific research training courses recommended [1] Scientific research skills︱The 4th near-infrared brain function data analysis class (Online: 2022.
    4.
    18~4.
    30) [2] Scientific research skills︱Introduction to Magnetic Resonance Brain Network Analysis (Online: 2022.
    4 .
    6~4.
    16)【3】Training Course︱Scientific Research Mapping·Academic Image Special Training【4】Seminar on Single-Cell Sequencing and Spatial Transcriptomics Data Analysis (2022.
    4.
    2-3 Tencent Online) References (swipe up and down to view) [ 1]NCD Risk Factor Collaboration (NCD-RisC).
    Worldwide Trends in Body-Mass Index, Underweight, Overweight, and Obesity from 1975 to 2016: A Pooled Analysis of 2416 Population-Based Measurement Studies in 128·9 Million Children, Adolescents,and Adults.
    Lancet (London, England) 2017, 390 (10113), 2627–2642.
    https://doi.
    org/10.
    1016/S0140-6736(17)32129-3.
    [2]Aune, D.
    ; Sen, A.
    ; Prasad, M.
    ; Norat, T.
    ; Janszky, I.
    ; Tonstad, S.
    ; Romundstad, P.
    ; Vatten, LJ BMI and All Cause Mortality: Systematic Review and Non-Linear Dose-Response Meta-Analysis of 230 Cohort Studies with 3.
    74 Million Deaths among 30.
    3 Million Participants.
    BMJ (Online).
    2016.
    https://doi.
    org/10.
    1136/bmj.
    i2156.
    [3] Son, JW; Kim, S.
    Comprehensive Review of Current and Upcoming Anti-Obesity Drugs.
    Diabetes and Metabolism Journal.
    2020.
    https://doi.
    org/10.
    4093/DMJ.
    2020.
    0258.
    [4]Lei, XG; Ruan, JQ; Lai, C.
    ; Sun, Z.
    ; Yang, X .
    Efficacy and Safety of Phentermine/Topiramate in Adults with Overweight or Obesity: A Systematic Review and Meta-Analysis.
    Obesity 2021.
    https://doi.
    org/10.
    1002/oby.
    23152.
    [5]Tak, YJ; Lee, SYLong-Term Efficacy and Safety of Anti-Obesity Treatment: Where Do We Stand? Current Obesity Reports.
    2021.
    https://doi.
    org/10.
    1007/s13679-020-00422-w.
    [6]Haas, HL; Sergeeva, OA; Selbach, O.
    Histamine in the Nervous System.
    Physiol.
    Rev.
    2008, 88 (3), 1183–1241.
    https://doi.
    org/10.
    1152/physrev.
    00043.
    2007.
    [7]Clineschmidt, BV; Lotti, VJ Histamine: Intraventricular Injection Suppresses Ingestive Behavior of the Cat.
    Arch.
    Int.
    Pharmacodyn.
    Ther.
    1973, 206 (2), 288–298.
    [8]Ookuma, K.
    ; Sakata, T.
    ; Fukagawa, K.
    ; Yoshimatsu , H.
    ; Kurokawa, M.
    ; Machidori, H.
    ; Fujimoto, K.
    Neuronal Histamine in the Hypothalamus Suppresses Food Intake in Rats.
    Brain Res.
    1993, 628 (1–2), 235–242.
    https://doi .
    org/10.
    1016/0006-8993(93)90960-U.
    [9]Sakata, T.
    ; Ookuma, K.
    ; Fukagawa, K.
    ; Fujimoto, K.
    ; Yoshimatsu, H.
    ; Shiraishi, T.
    ; Wada, H.
    Blockade of the Histamine H1-Receptor in the Rat Ventromedial Hypothalamus and Feeding Elicitation.
    Brain Res.
    1988, 441 (1–2), 403–407.
    https://doi.
    org/10.
    1016/0006-8993(88)91423- 0.
    [10]Valdés, JL; Sánchez, C.
    ; Riveros, ME; Blandina, P.
    ; Contreras, M.
    ; Farías, P.
    ; Torrealba, F.
    The Histaminergic Tuberomammillary Nucleus Is Critical for Motivated Arousal.
    Eur.
    J .
    Neurosci.
    2010, 31 (11), 2073–2085.
    https://doi.
    org/10.
    1111/j.
    1460-9568.
    2010.
    07241.
    x.
    [11]Valdés, JL; Farías, P.
    ; Ocampo-Garcés, A.
    ; Cortés, N.
    ; Serón-Ferré, M.
    ; Torrealba, F.
    Arousal and Differential Fos Expression in Histaminergic Neurons of the Ascending Arousal System during a Feeding-Related Motivated Behaviour.
    Eur.
    J.
    Neurosci.
    2005.
    https: //doi.
    org/10.
    1111/j.
    1460-9568.
    2005.
    04013.
    x.
    [12]Herman, AM; Ortiz-Guzman, J.
    ; Kochukov, M.
    ; Herman, I.
    ; Quast, KB; Patel, JM;Tepe, B.
    ; Carlson, JC; Ung, K.
    ; Selever, J.
    ; Tong, Q.
    ; Arenkiel, BR A Cholinergic Basal Forebrain Feeding Circuit Modulates Appetite Suppression.
    Nature 2016, 538 (7624), 253–256.
    https ://doi.
    org/10.
    1038/nature19789.
    [13]Sweeney, P.
    ; Li, C.
    ; Yang, Y.
    Appetite Suppressive Role of Medial Septal Glutamatergic Neurons.
    Proc.
    Natl.
    Acad.
    Sci.
    USA 2017, 114 ( 52), 13816–13821.
    https://doi.
    org/10.
    1073/pnas.
    1707228114.
    [14] Sweeney, P.
    ; Yang, Y.
    An Inhibitory Septum to Lateral Hypothalamus Circuit That Suppresses Feeding.
    J.
    Neurosci.
    2016, 36 (44), 11185–11195.
    https://doi.
    org/10.
    1523/JNEUROSCI.
    2042-16.
    2016.
    [15]Wang, Y.
    ; Xu, C.
    ; Xu, Z.
    ; Ji, C.
    ; Liang, J.
    ; Wang, Y.
    ; Chen, B.
    ; Wu, X.
    ; Gao, F.
    ; Wang, S.
    ; Guo, Y.
    ; Li, X.
    ; Luo, J.
    ; Duan, S.
    ; Chen, Z.
    Depolarized GABAergic Signaling in Subicular Microcircuits Mediates Generalized Seizure in Temporal Lobe Epilepsy.
    Neuron 2017.
    https://doi.
    org/10.
    1016/j.
    neuron.
    2017.
    06.
    004.
    [16]Wu, D.
    ; Fei, F.
    ; Zhang, Q.
    ; Wang, X.
    ; Gong, Y.
    ; Chen, X.
    ; Zheng, Y.
    ; Tan, B.
    ; Xu, C.
    ; Xie, H.
    ; Fang, W.
    ; Chen, Z.
    ; Wang, Y.
    Nanoengineered On-Demand Drug Delivery System Improves Efficacy of Pharmacotherapy for Epilepsy.
    Sci.
    Adv.
    2022, 8 (2), 1–15.
    https://doi.
    org/10.
    1126/sciadv.
    abm3381.
    [17]Liu, J.
    ; Li, F.
    ; Wang, Y.
    ; Pan, L.
    ; Lin, P.
    ; Zhang, B.
    ; Zheng, Y.
    ; Xu, Y.
    ; Liao, H.
    ; Ko, G.
    ; Fei, F.
    ; Xu, C.
    ; Du, Y.
    ; Shin, K.
    ; Kim, D.
    ; Jang, SS; Chung, HJ; Tian, ​​H.
    ; Wang, Q.
    ; Guo, W.
    ; Nam, JM; Chen, Z.
    ; Hyeon, T.
    ; Ling, D.
    A Sensitive and Specific Nanosensor for Monitoring Extracellular Potassium Levels in the Brain.
    Nat.
    Nanotechnol.
    2020.
    https://doi.
    org/10.
    1038/s41565-020-0634-4.
    [18]Xu, C.
    ; Wang, Y.
    ; Zhang, S.
    ; Nao, J.
    ; Liu, Y.
    ; Wang, Y.
    ; Ding, F.
    ; Zhong, K.
    ; Chen, L.
    ; Ying, X.
    ; Wang, S.
    ; Zhou, Y.
    ; Duan, S.
    ; Chen, Z.
    Subicular Pyramidal Neurons Gate Drug Resistance in Temporal Lobe Epilepsy.
    Ann.
    Neurol.
    2019.
    https ://doi.
    org/10.
    1002/ana.
    25554.
    [19]Zheng, Y.
    ; Zhang, X.
    ; Wu, X.
    ; Jiang, L.
    ; Ahsan, A.
    ; Ma, S.
    ; Xiao, Z.
    ; Han, F.
    ; Qin, ZH; Hu, W.
    ; Chen, Z.
    Somatic Autophagy of Axonal Mitochondria in Ischemic Neurons.
    J.
    Cell Biol.
    2019.
    https://doi.
    org/10.
    1083/JCB.
    201804101.
    [ 20]Liao, R.
    ; Chen, Y.
    ; Cheng, L.
    ; Fan, L.
    ; Chen, H.
    ; Wan, Y.
    ; You, Y.
    ; Zheng, Y.
    ; Jiang, L.
    ; Chen, Z .
    ; Zhang, X.
    ; Hu, W.
    Histamine H1 Receptors in Neural Stem Cells Are Required for the Promotion of Neurogenesis Conferred by H3 Receptor Antagonism Following Traumatic Brain Injury.
    Stem Cell Reports 2019, 12 (3), 532–544.
    https ://doi.
    org/10.
    1016/j.
    stemcr.
    2019.
    01.
    004.
    [21]Yan, H.
    ; Zhang, X.
    ; Hu, W.
    ; Ma, J.
    ; Hou, W.
    ; Zhang, X.
    ; Wang , X.
    ; Gao, J.
    ; Shen, Y.
    ; Lv, J.
    ; Ohtsu, H.
    ; Han, F.
    ; Wang, G.
    ; Chen, Z.
    Histamine H3 Receptors Aggravate Cerebral Ischaemic Injury by Histamine-Independent Mechanisms .
    Nat.
    Commun.
    2014.
    https://doi.
    org/10.
    1038/ncomms4334.
    [22] Cheng, L.
    ; Xu, C.
    ; Wang, L.
    ; An, D.
    ; Jiang, L.
    ; Zheng, Y .
    ; Xu, Y.
    ; Wang, Y.
    ; Wang, Y.
    ; Zhang, K.
    ; Wang, X.
    ; Zhang, X.
    ; Bao, A.
    ; Zhou, Y.
    ; Yang, J.
    ; Duan, S .
    ; Swaab, DF; Hu, W.
    ; Chen, Z.
    Histamine H1 Receptor Deletion in Cholinergic Neurons Induces Sensorimotor Gating Ability Deficit and Social Impairments in Mice.
    Nat.
    Commun.
    2021.
    https://doi.
    org/10.
    1038/s41467 -021-21476-x.
    [23]Jiang, L.
    ; Cheng, L.
    ; Chen, H.
    ; Dai, H.
    ; An, D.
    ; Ma, Q.
    ; Zheng, Y.
    ; Zhang, X.
    ; Hu, W.
    ; Chen, Z.
    Histamine H2 Receptor Negatively Regulates Oligodendrocyte Differentiation in Neonatal Hypoxic-Ischemic White Matter Injury.
    J.
    Exp.
    Med.
    2021, 218 (1).
    https://doi.
    org/10.
    1084/JEM.
    20191365.
    [24]Hu, W.
    ; Chen, Z.
    The Roles of Histamine and Its Receptor Ligands in Central Nervous System Disorders: An Update.
    Pharmacol.
    Ther.
    2017, 175, 116–132.
    https://doi.
    org/10.
    1016/j.
    pharmthera.
    2017.
    02.
    039.
    Plate making︱Wang Sizhen End of this article
    This article is an English version of an article which is originally in the Chinese language on echemi.com and is provided for information purposes only. This website makes no representation or warranty of any kind, either expressed or implied, as to the accuracy, completeness ownership or reliability of the article or any translations thereof. If you have any concerns or complaints relating to the article, please send an email, providing a detailed description of the concern or complaint, to service@echemi.com. A staff member will contact you within 5 working days. Once verified, infringing content will be removed immediately.

    Contact Us

    The source of this page with content of products and services is from Internet, which doesn't represent ECHEMI's opinion. If you have any queries, please write to service@echemi.com. It will be replied within 5 days.

    Moreover, if you find any instances of plagiarism from the page, please send email to service@echemi.com with relevant evidence.