Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 821
Filter
1.
J Comp Neurol ; 532(10): e25671, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39387358

ABSTRACT

Odor information is modulated by centrifugal inputs from other brain regions to the olfactory bulb (OB). Neurons containing monoamines, such as serotonin, acetylcholine, and noradrenaline, are well known as centrifugal inputs; however, the role of histamine, which is also present in the OB, is not well understood. In this study, we examined the histaminergic neurons projecting from the hypothalamus to the OB. We used an antibody against histidine decarboxylase (HDC), a synthesizing enzyme of histamine, to identify histaminergic neurons and assess their localization within the OB and the ultrastructure of their fibers and synapses using multiple immunostaining laser microscopy, ultra-high voltage electron microscopy (EM), and EM to confirm their relationships with other neurons. To further identify the origin nucleus of the histaminergic neurons projecting to the OB, we injected the retrograde tracer FluoroGold and analyzed the pathway to the OB anterogradely. HDC-immunoreactive (-ir) fibers were abundant in the olfactory nerve (ON) layer compared to other monoamines. HDC-ir neurons received asymmetrical synapses from ONs and formed synapses containing pleomorphic vesicles with variable postsynaptic densities to non-ON elements, thus forming serial synapses. We also confirmed that histaminergic neurons project from the rostral ventral tuberomammillary nucleus to the granule cell layer of the OB and, for the first time, successfully visualized their axons from the hypothalamus to the OB. These findings indicate that histamine may regulate odor discrimination in the OB, suggesting a regulatory relationship between hypothalamic function and olfaction. We thus elucidate morphological mechanisms with tuberomammillary nucleus-derived histaminergic neurons involved in olfactory information.


Subject(s)
Histamine , Neurons , Olfactory Bulb , Animals , Olfactory Bulb/metabolism , Olfactory Bulb/ultrastructure , Histamine/metabolism , Mice , Male , Neurons/metabolism , Neurons/ultrastructure , Mice, Inbred C57BL , Histidine Decarboxylase/metabolism , Synapses/metabolism , Synapses/ultrastructure , Nerve Net/metabolism , Nerve Net/chemistry , Olfactory Pathways/metabolism
2.
EMBO J ; 43(20): 4472-4491, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39242788

ABSTRACT

Monoamine neurotransmitters generated by de novo synthesis are rapidly transported and stored into synaptic vesicles at axon terminals. This transport is essential both for sustaining synaptic transmission and for limiting the toxic effects of monoamines. Here, synthesis of the monoamine histamine by histidine decarboxylase (HDC) and subsequent loading of histamine into synaptic vesicles are shown to be physically and functionally coupled within Drosophila photoreceptor terminals. This process requires HDC anchoring to synaptic vesicles via interactions with N-ethylmaleimide-sensitive fusion protein 1 (NSF1). Disassociating HDC from synaptic vesicles disrupts visual synaptic transmission and causes somatic accumulation of histamine, which leads to retinal degeneration. We further identified a proteasome degradation system mediated by the E3 ubiquitin ligase, purity of essence (POE), which clears mislocalized HDC from the soma, thus eliminating the cytotoxic effects of histamine. Taken together, our results reveal a dual mechanism for translocation and degradation of HDC that ensures restriction of histamine synthesis to axonal terminals and at the same time rapid loading into synaptic vesicles. This is crucial for sustaining neurotransmission and protecting against cytotoxic monoamines.


Subject(s)
Drosophila Proteins , Histamine , Presynaptic Terminals , Synaptic Vesicles , Animals , Histamine/metabolism , Synaptic Vesicles/metabolism , Presynaptic Terminals/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Histidine Decarboxylase/metabolism , Histidine Decarboxylase/genetics , Synaptic Transmission , Drosophila melanogaster/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Proteolysis , Proteasome Endopeptidase Complex/metabolism , Drosophila/metabolism , Biological Transport
3.
FEBS J ; 291(17): 3924-3937, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38973142

ABSTRACT

Accumulating evidence shows that inflammation is essential for embryo implantation and decidualization. Histamine, a proinflammatory factor that is present in almost all mammalian tissues, is synthesized through decarboxylating histidine by histidine decarboxylase (HDC). Although histamine is known to be essential for decidualization, the underlying mechanism remains undefined. In the present study, histamine had no obvious direct effects on in vitro decidualization in mice. However, the obvious differences in HDC protein levels between day 4 of pregnancy and day 4 of pseudopregnancy, as well as between delayed and activated implantation, suggested that the blastocyst may be involved in regulating HDC expression. Furthermore, blastocyst-derived tumor necrosis factor α (TNFα) significantly increased HDC levels in the luminal epithelium. Histamine increased the levels of amphiregulin (AREG) and disintegrin and metalloproteinase domain-containing protein 17 (ADAM17) proteins, which was abrogated by treatment with famotidine, a specific histamine type 2 receptor (H2R) inhibitor, or by TPAI-1 (a specific inhibitor of ADAM17). Intraluminal injection of urocanic acid (HDC inhibitor) on day 4 of pregnancy significantly reduced the number of implantation sites on day 5 of pregnancy. TNFα-stimulated increases in HDC, AREG and ADAM17 protein levels was abrogated by urocanic acid, a specific inhibitor of HDC. Additionally, AREG treatment significantly promoted in vitro decidualization. Collectively, our data suggests that blastocyst-derived TNFα induces luminal epithelial histamine secretion, and histamine increases mouse decidualization through ADAM17-mediated AREG release.


Subject(s)
ADAM17 Protein , Amphiregulin , Embryo Implantation , Histamine , Animals , Amphiregulin/metabolism , Amphiregulin/genetics , Female , Mice , Pregnancy , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Histamine/metabolism , Embryo Implantation/drug effects , Decidua/metabolism , Decidua/drug effects , Tumor Necrosis Factor-alpha/metabolism , Histidine Decarboxylase/metabolism , Histidine Decarboxylase/genetics , Blastocyst/metabolism , Blastocyst/drug effects
4.
Syst Appl Microbiol ; 47(5): 126539, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39029335

ABSTRACT

Histamine is an important biogenic amine known to impact a variety of patho-physiological processes ranging from allergic reactions, gut-mediated anti-inflammatory responses, and neurotransmitter activity. Histamine is found both endogenously within specialized host cells and exogenously in microbes. Exogenous histamine is produced through the decarboxylation of the amino acid L-histidine by bacterial-derived histidine decarboxylase enzymes. To investigate how widespread histamine production is across bacterial species, we examined 102,018 annotated genomes in the Integrated Microbial Genomes Database and identified 3,679 bacterial genomes (3.6 %) which possess the enzymatic machinery to generate histamine. These bacteria belonged to 10 phyla: Bacillota, Bacteroidota, Actinomycetota, Pseudomonadota, Lentisphaerota, Fusobacteriota, Armatimonadota, Cyanobacteriota, Thermodesulfobacteriota, and Verrucomicrobiota. The majority of the identified bacteria were terrestrial or aquatic in origin, although several bacteria originated in the human gut microbiota. We used liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based targeted metabolomics to confirm our genome discoveries correlated with L-histidine-to-histamine conversion in a chemically defined bacterial growth medium by a cohort of select environmental and human gut bacteria. We found that environmental microbes Vibrio harveyi, Pseudomonas fluorescens and Streptomyces griseus generated considerable levels of histamine (788 - 8,730 ng/mL). Interestingly, we found higher concentrations of histamine produced by gut-associated Fusobacterium varium, Clostridium perfringens, Limosilactobacillus reuteri and Morganella morganii (8,510--82,400 ng/mL). This work expands our knowledge of histamine production by diverse microbes.


Subject(s)
Bacteria , Genome, Bacterial , Histamine , Phylogeny , Histamine/metabolism , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Genome, Bacterial/genetics , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Humans , Tandem Mass Spectrometry , Gastrointestinal Microbiome , Histidine/metabolism , Chromatography, Liquid , Metabolomics
5.
Int Immunopharmacol ; 138: 112639, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38972209

ABSTRACT

Cisplatin (CDDP) is extensively utilized in the management of diverse types of cancers, but its ototoxicity cannot be ignored, and clinical interventions are not ideal. Histidine decarboxylase (HDC) is the exclusive enzyme for histamine synthesis. Anti-histamine receptor drugs are ubiquitously employed in the therapeutics of allergies and gastrointestinal diseases. Yet, the specific role of histamine and its signaling in the inner ear is not fully understood. This study utilized cisplatin treated mice and HEI-OC1 auditory hair cell line to establish a cisplatin-induced ototoxicity (CIO) model. Histidine decarboxylase knockout (HDC-/-) mice and histamine receptor 1 (H1R) antagonist were utilized to investigate the influence of HDC/histamine/H1R signaling on ototoxicity. The results identified HDC and H1R expression in mouse hair cells. Transcriptomics indicated that the expression levels of oxidative stress-related genes in the cochlea of HDC-/- mice increased. Furthermore, histamine deficiency or suppression of H1R signaling accelerated HC ferroptosis, a pivotal factor underlying the aggravation of CIO in vivo and in vitro, conversely, the supplementation of exogenous histamine reversed these deleterious effects. Mechanistically, this study revealed that the malfunction of HDC/histamine/H1R signaling induced upregulation of NRF2 expression, accompanied by the upregulation of ACSL4 and downregulation of GPX4 expression, which are major regulatory factors of ferroptosis. In summary, histamine deficiency may induce hair cell death by regulating the H1R pathway and exacerbate CIO. Our findings have indicated a potential therapeutic target for CIO.


Subject(s)
Cisplatin , Ferroptosis , Hair Cells, Auditory , Histamine , Histidine Decarboxylase , Mice, Knockout , Signal Transduction , Animals , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Histamine/metabolism , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Hair Cells, Auditory/metabolism , Mice , Ferroptosis/drug effects , Signal Transduction/drug effects , Ototoxicity , Receptors, Histamine H1/metabolism , Receptors, Histamine H1/genetics , Antineoplastic Agents/adverse effects , Mice, Inbred C57BL , Cell Line , Male , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics
6.
J Appl Microbiol ; 135(1)2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38140945

ABSTRACT

AIM: To investigate the inhibitory impact of chlorogenic acid (CGA) on the growth of Morganella psychrotolerans and its ability to form histamine. METHODS AND RESULTS: The antimicrobial effect of CGA on M. psychrotolerans was evaluated using the minimum inhibitory concentration (MIC) method, revealing an MIC value of 10 mg ml-1. The alkaline phosphatase (AKP) activity, cell membrane potential, and scanning electron microscopy images revealed that CGA treatment disrupted cell structure and cell membrane. Moreover, CGA treatment led to a dose-dependent decrease in crude histidine decarboxylase (HDC) activity and gene expression of histidine decarboxylase (hdc). Molecular docking analysis demonstrated that CGA interacted with HDC through hydrogen bonds. Furthermore, in situ investigation confirmed the efficacy of CGA in controlling the growth of M. psychrotolerans and significantly reducing histamine formation in raw tuna. CONCLUSION: CGA had good activity in controlling the growth of M. psychrotolerans and histamine formation.


Subject(s)
Chlorogenic Acid , Histamine , Histamine/analysis , Chlorogenic Acid/pharmacology , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Molecular Docking Simulation , Seafood
7.
Proc Natl Acad Sci U S A ; 120(14): e2216231120, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36976764

ABSTRACT

Histamine is a conserved neuromodulator in mammalian brains and critically involved in many physiological functions. Understanding the precise structure of the histaminergic network is the cornerstone in elucidating its function. Herein, using histidine decarboxylase (HDC)-CreERT2 mice and genetic labeling strategies, we reconstructed a whole-brain three dimensional (3D) structure of histaminergic neurons and their outputs at 0.32 × 0.32 × 2 µm3 pixel resolution with a cutting-edge fluorescence microoptical sectioning tomography system. We quantified the fluorescence density of all brain areas and found that histaminergic fiber density varied significantly among brain regions. The density of histaminergic fiber was positively correlated with the amount of histamine release induced by optogenetic stimulation or physiological aversive stimulation. Lastly, we reconstructed a fine morphological structure of 60 histaminergic neurons via sparse labeling and uncovered the largely heterogeneous projection pattern of individual histaminergic neurons. Collectively, this study reveals an unprecedented whole-brain quantitative analysis of histaminergic projections at the mesoscopic level, providing a foundation for future functional histaminergic study.


Subject(s)
Brain , Histamine , Mice , Animals , Brain/metabolism , Neurons/metabolism , Brain Mapping , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Mammals/metabolism
8.
Free Radic Biol Med ; 192: 98-114, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36165929

ABSTRACT

Doxorubicin (DOX) is widely used in the treatment of various cancers, increasing the great risk of adverse cardiovascular events, while the clinical intervention effect is not ideal. Histamine has been documented to participate in pathophysiological processes of cardiovascular diseases and inflammation-associated carcinogenesis. However, the potential roles of histamine in antitumor-related cardiotoxicity have not been fully elucidated. In this study, cardiomyocytes (hiPSC-CMs, HL-1 cells) and mice were treated with DOX to establish DOX-induced cardiotoxicity (DIC) models. Histidine decarboxylase knockout mice (HDC-/-) mice and histamine 1 receptor (H1R) antagonist were used to explore the effect of histamine/H1R signaling on DIC. Our results demonstrated that histamine deficiency or pharmaceutical inhibition of H1R accelerated myocardial ferroptosis, which is responsible for the aggravated DIC both in vivo and in vitro, while the supplementation of exogenous histamine reversed these changes. Our data revealed that the dysfunction of histamine/H1R signaling repressed the activation of transducer and activator of transcription 3 (STAT3), accompanying with decreased expression of solute carrier family7member11 (SLC7A11), a major modulator of ferroptosis. Conclusively, the disruption of histamine/H1R axis triggered ferroptosis and exacerbated DIC possibly by modulating STAT3-SLC7A11 pathway. Our findings point to a potential therapeutic target for DIC and provide more consideration on the usage of antihistamine drugs.


Subject(s)
Ferroptosis , Histamine , Animals , Cardiotoxicity/genetics , Doxorubicin/metabolism , Doxorubicin/toxicity , Ferroptosis/genetics , Histamine/metabolism , Histamine/pharmacology , Histamine Antagonists/metabolism , Histamine Antagonists/pharmacology , Histamine H1 Antagonists/pharmacology , Histidine Decarboxylase/metabolism , Histidine Decarboxylase/pharmacology , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Pharmaceutical Preparations/metabolism
9.
Plant Cell Rep ; 41(8): 1721-1731, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35739429

ABSTRACT

KEY MESSAGE: In SlHDC-A promoter, SlHDC-A core-ES is an essential region for fruit-specific expression and interacts with GATA, HSF and AP1. Triplication of essential region was proposed as a minimal fruit-specific promoter. In plant biotechnology, fruit-specific promoter is an important tool for the improvement and utilization of tomato fruit. To expand our understanding on fruit-specific expression, it is necessary to determine the promoter region involved in fruit-specific transcriptional activity and transcriptional regulations of the promoter. In previous study, we isolated a fruit-specific SlHDC-A core promoter specifically expressed during tomato ripening stages. In this study, we identified SlHDC-A promoter region (SlHDC-A core-ES) that is essential for fruit-specific expression of the SlHDC-A. To understand the molecular mechanisms of fruit-specific expression of the SlHDC-A promoter, we first identified the putative transcription factor binding elements in the SlHDC-A core promoter region and corresponding putative transcription factors which are highly expressed during fruit maturation. Yeast one hybrid analysis confirmed that GATA, HSF, and AP1 interact with the SlHDC-A core-ES promoter region. Further transactivation analysis revealed that expression of the three transcription factors significantly activated expression of a reporter gene driven by SlHDC-A core-ES promoter. These results suggest that GATA, HSF, and AP1 are involved in the fruit-specific expression of SlHDC-A promoter. Furthermore, the synthetic promoter composed of three tandem repeats of SlHDC-A core-ES showed relatively higher activity than the constitutive 35S promoter in the transgenic tomato fruits at the orange stage. Taken together, we propose a new synthetic promoter that is specifically expressed during fruit ripening stage.


Subject(s)
Solanum lycopersicum , Fruit/metabolism , Gene Expression Regulation, Plant/genetics , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Solanum lycopersicum/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Acta Pharmacol Sin ; 43(11): 2807-2816, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35581293

ABSTRACT

Our previous study showed that H3 receptor antagonists reduced neuronal apoptosis and cerebral infarction in the acute stage after cerebral ischemia, but through an action independent of activation of histaminergic neurons. Because enhanced angiogenesis facilitates neurogenesis and neurological recovery after ischemic stroke, we herein investigated whether antagonism of H3R promoted angiogenesis after brain ischemia. Photothrombotic stroke was induced in mice. We showed that administration of H3R antagonist thioperamide (THIO, 10 mg·kg-1·d-1, i.p., from D1 after cerebral ischemia) significantly improved angiogenesis assessed on D14, and attenuated neurological defects on D28 after cerebral ischemia. Compared with wild-type mice, Hrh3-/- mice displayed more blood vessels in the ischemic boundary zone on D14, and THIO administration did not promote angiogenesis in these knockout mice. THIO-promoted angiogenesis in mice was reversed by i.c.v. injection of H3R agonist immepip, but not by H1 and H2 receptor antagonists, histidine decarboxylase inhibitor α-fluoromethylhistidine, or histidine decarboxylase gene knockout (HDC-/-), suggesting that THIO-promoted angiogenesis was independent of activation of histaminergic neurons. In vascular endothelial cells (bEnd.3), THIO (10-9-10-7 M) dose-dependently facilitated cell migration and tube formation after oxygen glucose deprivation (OGD), and H3R knockdown caused similar effects. We further revealed that H3R antagonism reduced the interaction between H3R and Annexin A2, while knockdown of Annexin A2 abrogated THIO-promoted angiogenesis in bEnd.3 cells after OGD. Annexin A2-overexpressing mice displayed more blood vessels in the ischemic boundary zone, which was reversed by i.c.v. injection of immepip. In conclusion, this study demonstrates that H3R antagonism promotes angiogenesis after cerebral ischemia, which is independent of activation of histaminergic neurons, but related to the H3R on vascular endothelial cells and its interaction with Annexin A2. Thus, H3R antagonists might be promising drug candidates to improve angiogenesis and neurological recovery after ischemic stroke.


Subject(s)
Annexin A2 , Brain Ischemia , Ischemic Stroke , Receptors, Histamine H3 , Animals , Mice , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Receptors, Histamine H3/metabolism , Histamine , Endothelial Cells/metabolism , Brain Ischemia/drug therapy , Mice, Knockout , Cerebral Infarction
11.
Head Neck ; 44(7): 1554-1562, 2022 07.
Article in English | MEDLINE | ID: mdl-35411649

ABSTRACT

BACKGROUND: Tumor-associated macrophages in the tumor microenvironment (TME), as a factor affecting lymphocytes, have received much attention. Both lymphocytes and macrophages can switch the expression of histamine receptors. In this study, we investigated the role of histamine in the TME of tongue squamous cell carcinoma (SCC). METHODS: Sixty-seven patients with stage I tongue SCC were studied. Histamine was evaluated by the expression of L-histidine decarboxylase (HDC). Macrophages, T lymphocytes, and lymph vessel density, as well as the Ki-67 labeling index (LI) and depth of invasion (DOI), were compared with HDC expression. RESULTS: HDC expression was significantly affected by the TME. The DOI, worst pattern of invasion, and Ki-67 LI were associated with histamine expression. C-C motif chemokine ligand (CCL) 2 and CCL22 were co-expressed with histamine H1 and H2 receptors. Histamine expression was most affected by the DOI. CONCLUSIONS: Tongue SCC expressing histamine affected the TME via histamine receptors and chemokines.


Subject(s)
Carcinoma, Squamous Cell , Tongue Neoplasms , Carcinoma, Squamous Cell/pathology , Chemokines , Histamine/metabolism , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Humans , Ki-67 Antigen , Receptors, Histamine , Tongue/pathology , Tumor Microenvironment
12.
Elife ; 112022 03 01.
Article in English | MEDLINE | ID: mdl-35229720

ABSTRACT

Neurotransmitters are generated by de novo synthesis and are essential for sustained, high-frequency synaptic transmission. Histamine, a monoamine neurotransmitter, is synthesized through decarboxylation of histidine by histidine decarboxylase (Hdc). However, little is known about how histidine is presented to Hdc as a precursor. Here, we identified a specific histidine transporter, TADR (torn and diminished rhabdomeres), which is required for visual transmission in Drosophila. Both TADR and Hdc localized to neuronal terminals, and mutations in tadr reduced levels of histamine, thus disrupting visual synaptic transmission and phototaxis behavior. These results demonstrate that a specific amino acid transporter provides precursors for monoamine neurotransmitters, providing the first genetic evidence that a histidine amino acid transporter plays a critical role in synaptic transmission. These results suggest that TADR-dependent local de novo synthesis of histamine is required for synaptic transmission.


Subject(s)
Drosophila , Histidine , Animals , Drosophila/genetics , Histamine/metabolism , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Membrane Transport Proteins/metabolism , Neurotransmitter Agents , Synaptic Transmission
13.
J Cardiovasc Transl Res ; 15(3): 621-634, 2022 06.
Article in English | MEDLINE | ID: mdl-34734351

ABSTRACT

Myocardial infarction (MI) is a significant contributor to the development of heart failure. Histidine decarboxylase (HDC), the unique enzyme that converts L-histidine to histamine, is highly expressed in CD11b+ immature myeloid cells. However, the relationship between HDC-expressing macrophages and cardiac myofibroblasts remains to be explained. Here, we demonstrate that the GFP (green fluorescent protein)-labeled HDC+CD11b+ myeloid precursors and their descendants could differentiate into fibroblast-like cells in myocardial interstitium. Furthermore, we prove that CD11b+Ly6C+ monocytes/macrophages, but not CD11b+Ly6G+ granulocytes, are identified as the main cellular source for bone marrow-derived myofibroblast transformation, which could be regulated via histamine H1 and H2 receptor-dependent signaling pathways. Using HDC knockout mice, we find that histamine deficiency promotes myofibroblast transformation from Ly6C+ macrophages and cardiac fibrosis partly through upregulating the expression of Krüppel-like factor 5 (KLF5). Taken together, our data uncover a central role of HDC in regulating bone marrow-derived macrophage-to-myofibroblast transformation but also identify a histamine receptor (HR)-KLF5 related signaling pathway that mediates myocardial fibrosis post-MI. CD11b+Ly6C+ monocytes/macrophages are the main cellular source for bone marrow-derived myofibroblast transformation. Histamine inhibits myofibroblasts transformation via H1R and H2R-dependent signaling pathways, and ameliorates cardiac fibrosis partly through upregulating KLF5 expression.


Subject(s)
Histamine , Histidine Decarboxylase , Myeloid Cells , Myocardial Infarction , Myofibroblasts , Animals , Fibrosis , Histamine/deficiency , Histidine Decarboxylase/metabolism , Mice , Myeloid Cells/cytology , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myofibroblasts/cytology
14.
Curr Top Behav Neurosci ; 59: 291-302, 2022.
Article in English | MEDLINE | ID: mdl-34761361

ABSTRACT

Three of the four histamine receptors have been identified in zebrafish. Whereas only one histamine receptor 1 gene (hrh1) is known, two copies of histamine receptor 2 (hrh2a and hrh2b) have been identified. Although initially only one gene encoding for histamine receptor 3 (hrh3) was recognized in zebrafish, the genome database contains information for two more hrh3-like genes, whereas no genes corresponding for histamine receptor 4 with expression mainly in the immune system have been identified. Hrh1 and hrh3 show prominent uneven expression in the zebrafish brain, with the strongest expression in the dorsal telencephalon. Quantitatively significant expression of hrh1, hrh2, and hrh3 can also be found in several peripheral organs. Whereas antagonists of hrh1, hrh2, and hrh3 all affect the locomotor activity of zebrafish larvae, interpretation of the data is hampered by a lack of information on receptor binding and signaling characteristics. Zebrafish mutants lacking any of the three histamine receptors have shown modest behavioral phenotypes, possibly due to genetic compensation. None of the receptor mutant fish have shown significant sleep phenotypes. Adult zebrafish lacking hrh3 display decreased locomotor activity. The zebrafish histamine system shows significant life-long plasticity: presenilin 1 mutant zebrafish develop an abnormally large number of histamine neurons and increased thigmotaxis and anxiety-related phenotype. Overexpression of histidine decarboxylase (hdc) in larval zebrafish is associated with an increased number of hypocretin neurons, whereas translation inhibition of hdc or exposure to α-fluoromethylhistidine leads to decreased numbers of hypocretin neurons. Current pharmacological evidence suggests that this may be mediated by hrh1. Further studies using acute, e.g., pharmacogenetic or optogenetic manipulation of selected components of brain circuits, are required to understand the full range of physiological functions of zebrafish histamine receptors.


Subject(s)
Histamine , Zebrafish , Animals , Brain/metabolism , Histamine/metabolism , Histamine/pharmacology , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Orexins/metabolism , Presenilin-1/metabolism , Receptors, Histamine/genetics , Receptors, Histamine/metabolism , Zebrafish/metabolism
15.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34299019

ABSTRACT

Myocardial infarction (MI) is one of the most common cardiovascular diseases. Although previous studies have shown that histidine decarboxylase (HDC), a histamine-synthesizing enzyme, is involved in the stress response and heart remodeling after MI, the mechanism underlying it remains unclear. In this study, using Hdc-deficient mice (Hdc-/- mice), we established an acute myocardial infarction mouse model to explore the potential roles of Hdc/histamine in cardiac immune responses. Comprehensive analysis was performed on the transcriptomes of infarcted hearts. Differentially expressed gene (DEG) analysis identified 2126 DEGs in Hdc-deficient groups and 1013 in histamine-treated groups. Immune related pathways were enriched in Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Then we used the ssGSEA algorithm to evaluate 22 kinds of infiltrated immunocytes, which indicated that myeloid cells and T memory/follicular helper cells were tightly regulated by Hdc/histamine post MI. The relationships of lncRNAs and the Gene Ontology (GO) functions of protein-coding RNAs and immunocytes were dissected in networks to unveil immune-associated lncRNAs and their roles in immune modulation after MI. Finally, we screened out and verified four lncRNAs, which were closely implicated in tuning the immune responses after MI, including ENSMUST00000191157, ENSMUST00000180693 (PTPRE-AS1), and ENSMUST-00000182785. Our study highlighted the HDC-regulated myeloid cells as a driving force contributing to the government of transmission from innate immunocytes to adaptive immunocytes in the progression of the injury response after MI. We identified the potential role of the Hdc/histamine-lncRNAs network in regulating cardiac immune responses, which may provide novel promising therapeutic targets for further promoting the treatment of ischemic heart disease.


Subject(s)
Histidine Decarboxylase/metabolism , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , RNA, Long Noncoding/metabolism , Transcriptome/genetics , Algorithms , Animals , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Gene Ontology , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Histidine Decarboxylase/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Cells/immunology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , RAW 264.7 Cells , RNA, Long Noncoding/genetics , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Helper-Inducer/immunology
17.
J Immunol ; 206(9): 2029-2037, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33846226

ABSTRACT

Histamine is best known for its role in allergies, but it could also be involved in autoimmune diseases such as multiple sclerosis. However, studies using experimental autoimmune encephalomyelitis (EAE), the most widely used animal model for multiple sclerosis, have reported conflicting observations and suggest the implication of a nonclassical source of histamine. In this study, we demonstrate that neutrophils are the main producers of histamine in the spinal cord of EAE mice. To assess the role of histamine by taking into account its different cellular sources, we used CRISPR-Cas9 to generate conditional knockout mice for the histamine-synthesizing enzyme histidine decarboxylase. We found that ubiquitous and cell-specific deletions do not affect the course of EAE. However, neutrophil-specific deletion attenuates hypothermia caused by IgE-mediated anaphylaxis, whereas neuron-specific deletion reduces circadian activity. In summary, this study refutes the role of histamine in EAE, unveils a role for neutrophil-derived histamine in IgE-mediated anaphylaxis, and establishes a new mouse model to re-explore the inflammatory and neurologic roles of histamine.


Subject(s)
Anaphylaxis/immunology , Circadian Rhythm/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Histamine/immunology , Histidine Decarboxylase/immunology , Anaphylaxis/genetics , Anaphylaxis/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Histamine/metabolism , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Humans , Kaplan-Meier Estimate , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Neutrophils/cytology , Neutrophils/immunology , Neutrophils/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism
18.
Anat Rec (Hoboken) ; 304(5): 1136-1150, 2021 05.
Article in English | MEDLINE | ID: mdl-33034098

ABSTRACT

Histidine decarboxylase (HDC), histamine synthase, is expressed in hematopoietic stem cells and in lineage-committed progenitors in the bone marrow (BM). However, the role of histamine in hematopoiesis is not well described. To evaluate the role of histamine in hematopoiesis, we analyzed the changes in HDC expression at hematopoietic sites, the BM, spleen, and liver of 2-, 3-, and 6-week-old wild-type mice. We also performed morphological analyses of the hematopoietic sites using HDC-deficient (HDC-KO) mice. In wild-type adults, HDC expression in the BM was higher than that in the spleen and liver and showed an age-dependent increase. Histological analysis showed no significant change in the adult BM and spleen of HDC-KO mice compared to wild-type mice. In the liver, HDC expression was temporarily increased at 3 weeks and decreased at 6 weeks of age. Morphological analysis of the liver revealed more numerous hematopoietic colonies and megakaryocytes in HDC-KO mice compared to wild-type mice at 2 and 3 weeks of age, whereas no changes were observed in adults. Most of these hematopoietic colonies consisted of B220-positive B-lymphocytes and TER119-positive erythroblasts and were positive for the cell proliferation marker PCNA. Notably, these hematopoietic colonies declined in HDC-KO mice upon N-acetyl histamine treatment. A significant increase in the expression of hematopoiesis-related cytokines, Il3, Il7, Epo, Gcsf, and Cxcl12 mRNA was observed in the liver of 3-week-old HDC-KO mice compared to wild-type mice. These results suggest that histamine-deficiency may maintain an microenvironment suitable for hematopoiesis by regulating hematopoiesis-related cytokine expression in the liver of postnatal mice.


Subject(s)
Hematopoiesis, Extramedullary/physiology , Histidine Decarboxylase/metabolism , Liver/metabolism , Spleen/metabolism , Animals , Histidine Decarboxylase/genetics , Mice , Mice, Knockout
19.
J Pineal Res ; 70(2): e12699, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33020940

ABSTRACT

Our daily rhythmicity is controlled by a circadian clock with a specific set of genes located in the suprachiasmatic nucleus in the hypothalamus. Mast cells (MCs) are major effector cells that play a protective role against pathogens and inflammation. MC distribution and activation are associated with the circadian rhythm via two major pathways, IgE/FcεRI- and IL-33/ST2-mediated signaling. Furthermore, there is a robust oscillation between clock genes and MC-specific genes. Melatonin is a hormone derived from the amino acid tryptophan and is produced primarily in the pineal gland near the center of the brain, and histamine is a biologically active amine synthesized from the decarboxylation of the amino acid histidine by the L-histidine decarboxylase enzyme. Melatonin and histamine are previously reported to modulate circadian rhythms by pathways incorporating various modulators in which the nuclear factor-binding near the κ light-chain gene in B cells, NF-κB, is the common key factor. NF-κB interacts with the core clock genes and disrupts the production of pro-inflammatory cytokine mediators such as IL-6, IL-13, and TNF-α. Currently, there has been no study evaluating the interdependence between melatonin and histamine with respect to circadian oscillations in MCs. Accumulating evidence suggests that restoring circadian rhythms in MCs by targeting melatonin and histamine via NF-κB may be promising therapeutic strategy for MC-mediated inflammatory diseases. This review summarizes recent findings for circadian-mediated MC functional roles and activation paradigms, as well as the therapeutic potentials of targeting circadian-mediated melatonin and histamine signaling in MC-dependent inflammatory diseases.


Subject(s)
Histamine/metabolism , Mast Cells/metabolism , Melatonin/metabolism , Pineal Gland/metabolism , Animals , Circadian Rhythm/physiology , Histidine Decarboxylase/metabolism , Humans , Interleukin-13/metabolism , Interleukin-6/metabolism , Mast Cells/immunology , Pineal Gland/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Biochem Biophys Res Commun ; 535: 19-24, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33340761

ABSTRACT

Histamine-producing cells include storage-type cells (e.g., mast cells and basophils), which store histamine intracellularly, and inducible-type cells (e.g., keratinocytes and macrophages), which induce histidine decarboxylase (HDC, a key enzyme for histamine biosynthesis) activity but do not have a storage pool of histamine. Most of the studies focused on identifying HDC-expressing cells by using cultured cells, and few on investigating the localization of HDC by using skin tissues. Hence, this study conducted immunohistochemical studies using human healthy skin samples. HDC-positive and cytokeratin 14 (a marker of basal keratinocytes)-negative cells were present around the basal layer of the epidermis. These cells did not immunohistochemically react with mast cell tryptase but expressed tyrosinase (a key enzyme for melanin biosynthesis) and microphthalmia-associated transcription factor (MITF, a transcription factor controlling the expression of tyrosinase genes). Melanin granules were clearly observed around HDC-positive and MITF-positive cells. Moreover, HDC mRNA and protein were both detected in cultured normal human epidermal melanocytes. In conclusion, HDC-positive and cytokeratin 14-negative cells around the basal layer of the epidermis are melanocytes.


Subject(s)
Histidine Decarboxylase/metabolism , Melanocytes/enzymology , Skin/cytology , Adolescent , Adult , Cells, Cultured , Collagen Type IV/metabolism , Epidermis/metabolism , Female , Histidine Decarboxylase/genetics , Humans , Male , Mast Cells/enzymology , Melanins/metabolism , Melanocytes/cytology , Microphthalmia-Associated Transcription Factor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tryptases , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL