Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.137
Filter
1.
J Pharmacol Sci ; 151(4): 177-186, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36925216

ABSTRACT

Histamine is a well-known inflammatory mediator, but how histamine induces angiogenesis remains poorly understood. In the present study, we demonstrated a dose-dependent dynamic tube formation in the human endothelial cell line EA.hy926 in the presence of histamine that was completely blocked by histamine H1 receptor (H1R) and protein kinase C (PKC) inhibitors. However, histamine H2, H3, and H4 receptor inhibitors did not inhibit tube formation, suggesting that H1R-PKC signaling is involved in histamine-induced tube formation. Moreover, we found an H1-specific induction of vascular endothelial growth factor (VEGF) expression. Inhibition of VEGF receptor 2 (VEGFR2) suppressed the histamine-induced tube formation, indicating that VEGF is downstream of histamine signaling. Additionally, we demonstrated that histamine stimulation induces the expression of critical regulators of angiogenesis such as matrix metalloproteinase (MMP)-9 and MMP-14 metalloproteases, as histamine-induced tube formation is blocked by MMP inhibitors. In summary, our study indicates that histamine can activate the H1R in human endothelial cells and thereby promote tube formation through the PKC, MMP, and VEGF signaling pathways.


Subject(s)
Histamine , Vascular Endothelial Growth Factor A , Humans , Histamine/pharmacology , Histamine/physiology , Vascular Endothelial Growth Factor A/metabolism , Endothelial Cells/metabolism , Receptors, Histamine H1/genetics , Receptors, Histamine H1/metabolism , Vascular Endothelial Growth Factors
2.
J Neurosci ; 40(34): 6557-6571, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32709692

ABSTRACT

A reduction in the synthesis of the neuromodulator histamine has been associated with Tourette's syndrome and obsessive-compulsive disorder. Symptoms of these disorders are thought to arise from a dysfunction or aberrant development ofcorticostriatal circuits. Here, we investigated how histamine affects developing corticostriatal circuits, both acutely and longer-term, during the first postnatal weeks, using patch-clamp and field recordings in mouse brain slices (C57Bl/6, male and female). Immunohistochemistry for histamine-containing axons reveals striatal histaminergic innervation by the second postnatal week, and qRT-PCR shows transcripts for H1, H2, and H3 histamine receptors in striatum from the first postnatal week onwards, with pronounced developmental increases in H3 receptor expression. Whole-cell patch-clamp recordings of striatal spiny projection neurons and histamine superfusion demonstrates expression of functional histamine receptors from the first postnatal week onwards, with histamine having diverse effects on their electrical properties, including depolarization of the membrane potential while simultaneously decreasing action potential output. Striatal field recordings and electrical stimulation of corticostriatal afferents revealed that histamine, acting at H3 receptors, negatively modulates corticostriatal synaptic transmission from the first postnatal week onwards. Last, we investigated effects of histamine on longer-term changes at developing corticostriatal synapses and show that histamine facilitates NMDA receptor-dependent LTP via H3 receptors during the second postnatal week, but inhibits synaptic plasticity at later developmental stages. Together, these results show that histamine acutely modulates developing striatal neurons and synapses and controls longer-term changes in developing corticostriatal circuits, thus providing insight into the possible etiology underlying neurodevelopmental disorders resulting from histamine dysregulation.SIGNIFICANCE STATEMENT Monogenic causes of neurologic disorders, although rare, can provide opportunities to both study and understand the brain. For example, a nonsense mutation in the coding gene for the histamine-synthesizing enzyme has been associated with Tourette's syndrome and obsessive-compulsive disorder, and dysfunction of corticostriatal circuits. Nevertheless, the etiology of these neurodevelopmental disorders and histamine's role in the development of corticostriatal circuits have remained understudied. Here we show that histamine is an active neuromodulator during the earliest periods of postnatal life and acts at developing striatal neurons and synapses. Crucially, we show that histamine permits NMDA receptor-dependent corticostriatal synaptic plasticity during an early critical period of postnatal development, which suggests that genetic or environmental perturbations of histamine levels can impact striatal development.


Subject(s)
Cerebral Cortex/growth & development , Cerebral Cortex/physiology , Corpus Striatum/growth & development , Corpus Striatum/physiology , Histamine/physiology , Neuronal Plasticity , Receptors, Histamine/physiology , Animals , Corpus Striatum/drug effects , Female , Histamine/administration & dosage , Male , Membrane Potentials , Mice, Inbred C57BL , Neural Pathways/growth & development , Neural Pathways/physiology , Receptors, Histamine H1/physiology , Receptors, Histamine H2/physiology , Receptors, Histamine H3/physiology , Synaptic Transmission
3.
Genes Cells ; 25(7): 443-449, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32394600

ABSTRACT

Histamine is a bioactive monoamine that is synthesized by the enzymatic activity of histidine decarboxylase (HDC) in basophils, mast cells, gastric enterochromaffin-like (ECL) cells and histaminergic neuronal cells. Upon a series of cellular stimuli, these cells release stored histamine, which elicits allergies, inflammation, and gastric acid secretion and regulates neuronal activity. Recent studies have shown that certain other types of myeloid lineage cells also produce histamine with HDC induction under various pathogenic stimuli. Histamine has been shown to play a series of pathophysiological roles by modulating immune and inflammatory responses in a number of disease conditions, whereas the mechanistic aspects underlying induced HDC expression remain elusive. In the present review, we summarize the current understanding of the regulatory mechanism of Hdc gene expression and the roles played by histamine in physiological contexts as well as pathogenic processes. We also introduce a newly developed histaminergic cell-monitoring transgenic mouse line (Hdc-BAC-GFP) that serves as a valuable experimental tool to identify the source of histamine and dissect upstream regulatory signals.


Subject(s)
Histamine/metabolism , Histidine Decarboxylase/metabolism , Receptors, Histamine/metabolism , Sepsis/immunology , Animals , Chromosomes, Artificial, Bacterial , Gene Expression Regulation, Enzymologic/immunology , Histamine/physiology , Histidine Decarboxylase/genetics , Histones/metabolism , Methylation , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Sepsis/metabolism
4.
Int J Mol Sci ; 22(13)2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34281281

ABSTRACT

Atopic dermatitis (AD) is a prototypic inflammatory disease that presents with intense itching. The pathophysiology of AD is multifactorial, involving environmental factors, genetic susceptibility, skin barrier function, and immune responses. A recent understanding of pruritus transmission provides more information about the role of pruritogens in the pathogenesis of AD. There is evidence that pruritogens are not only responsible for eliciting pruritus, but also interact with immune cells and act as inflammatory mediators, which exacerbate the severity of AD. In this review, we discuss the interaction between pruritogens and inflammatory molecules and summarize the targeted therapies for AD.


Subject(s)
Dermatitis, Atopic/etiology , Pruritus/etiology , Cholestasis/complications , Cholestasis/physiopathology , Cholestasis/therapy , Cytokines/physiology , Dermatitis, Atopic/physiopathology , Dermatitis, Atopic/therapy , Histamine/physiology , Humans , Inflammation Mediators/physiology , Models, Biological , Neuroimmunomodulation/physiology , Pruritus/physiopathology , Pruritus/therapy , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy
5.
Am J Physiol Gastrointest Liver Physiol ; 319(6): G655-G668, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32996781

ABSTRACT

Early-life adversity contributes to the development of functional bowel disorders later in life through unresolved mechanisms. Here, we tested the hypothesis that early-life adversity alters anatomical and functional interactions between mast cells and enteric glia. The effects of early-life stress were studied using the neonatal maternal separation (NMS) stress mouse model. Anatomical relationships between mast cells and enteric glia were assessed using immunohistochemistry and mast cell reporter mice (Mcpt5Cre;GCaMP5g-tdT). Immunohistochemistry was used to assess the expression of histamine, histamine 1 (H1) receptors, and glial fibrillary acidic protein. Functional responses of glia to mast cell mediators were assessed in calcium imaging experiments using Sox10CreERT2;GCaMP5g-tdT mice and cultured human enteric glial cells. NMS increases mast cell numbers at the level of the myenteric plexus and their proximity to myenteric ganglia. Myenteric glia respond to mediators released by activated mast cells that are blocked by H1 receptor antagonists in mice and humans and by blocking neuronal activity with tetrodotoxin in mouse tissue. Histamine replicates the effects of mast cell supernatants on enteric glia, and NMS increases histamine production by mast cells. NMS reduces glial responses to mast cell mediators in mouse tissue, while potentiating responses in cultured human enteric glia. NMS increases myenteric glial fibrillary acidic protein expression and reduces glial process length but does not cause neurodegeneration. Histamine receptor expression is not altered by NMS and is localized to neurons in mice, but glia in humans. Early-life stress increases the potential for interactions between enteric glia and mast cells, and histamine is a potential mediator of mast cell-glial interactions through H1 receptors. We propose that glial-mast cell signaling is a mechanism that contributes to enteric neuroplasticity driven by early-life adversity.NEW & NOTEWORTHY Early-life adversity places an individual at risk for developing functional gastrointestinal disorders later in life through unknown mechanisms. Here, we show that interactions between mast cells and glia are disrupted by early-life stress in mice and that histamine is a potential mediator of mast cell-glial interactions.


Subject(s)
Histamine/physiology , Life Change Events , Mast Cells/physiology , Neuroglia/physiology , Neurons/physiology , Animals , Animals, Newborn , Cell Count , Cells, Cultured , Chymases/genetics , Female , Glial Fibrillary Acidic Protein/metabolism , Histamine H1 Antagonists/pharmacology , Humans , Maternal Deprivation , Mice , Mice, Inbred C57BL , Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Pregnancy , Receptors, Histamine H1/metabolism , Stress, Psychological/physiopathology
6.
J Pharmacol Sci ; 137(2): 122-128, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29858014

ABSTRACT

Astrocytes play key roles in regulating brain homeostasis and neuronal activity. This is, in part, accomplished by the ability of neurotransmitters in the synaptic cleft to bind astrocyte membrane receptors, activating signalling cascades that regulate concentration of intracellular Ca2+ ([Ca2+]i) and gliotransmitter release, including ATP and glutamate. Gliotransmitters contribute to dendrite formation and synaptic plasticity, and in some cases, exacerbate neurodegeneration. The neurotransmitter histamine participates in several physiological processes, such as the sleep-wake cycle and learning and memory. Previous studies have demonstrated the expression of histamine receptors on astrocytes, but until now, only a few studies have examined the effects of histamine on astrocyte intracellular signalling and gliotransmitter release. Here, we used the human astrocytoma cell line 1321N1 to study the role of histamine in astrocyte intracellular signalling and gliotransmitter release. We found that histamine activated astrocyte signalling through histamine H1 and H2 receptors, leading to distinct cellular responses. Activation of histamine H1 receptors caused concentration-dependent release of [Ca2+]i from internal stores and concentration-dependent increase in glutamate release. Histamine H2 receptor activation increased cyclic adenosine monophosphate (cAMP) levels and phosphorylation of transcription factor cAMP response-element binding protein. Taken together, these data emphasize a role for histamine in neuron-glia communication.


Subject(s)
Astrocytes/metabolism , Glutamates/metabolism , Histamine/pharmacology , Histamine/physiology , Adenosine Triphosphate/metabolism , Animals , Astrocytes/physiology , Calcium/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Humans , Neurotransmitter Agents/metabolism , Phosphorylation/drug effects , Rats , Receptors, Histamine H1/metabolism , Receptors, Histamine H2/metabolism , Signal Transduction/drug effects
7.
Neurobiol Learn Mem ; 145: 1-6, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28838882

ABSTRACT

Recent findings have reasserted the role of histamine in the regulation of memory consolidation first proposed in 1986 in an inhibitory avoidance task in rats. They indicate that histamine is indeed a major regulator of memory consolidation in various tasks, through H2 receptors in the dorsal hippocampus and through H3 receptors in the basolateral amygdala, depending on the task. In the object recognition task, the memory enhancing effect is mediated by the three receptors (H1, H2, H3) in the dorsal hippocampus. In social recognition, the consolidation effect is mediated by H2 receptors in both amygdala and dorsal hippocampus. Data have suggested, in addition, influences on retrieval; this has been best studied in the dorsal hippocampus in step-down inhibitory avoidance task. Depending on the recent history of the conditioned stimulus (i.e., whether it has been recently reinforced or not), histamine acts on hippocampal H1 receptors, facilitating retrieval, or on H2 receptors, inhibiting it.


Subject(s)
Brain/physiology , Histamine/physiology , Memory Consolidation/physiology , Amygdala/physiology , Animals , Avoidance Learning/physiology , Hippocampus/physiology , Receptors, Histamine/physiology
8.
Curr Opin Gastroenterol ; 33(6): 430-438, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28787289

ABSTRACT

PURPOSE OF REVIEW: The present review summarizes the past year's literature, both clinical and basic science, regarding physiologic and pharmacologic regulation of gastric acid secretion in health and disease. RECENT FINDINGS: Gastric acid kills microorganisms, assists digestion, and facilitates absorption of iron, calcium, and vitamin B12. The main stimulants of acid secretion are the hormone gastrin, released from antral G cells; paracrine agent histamine, released from oxyntic enterochromaffin-like cells; and neuropeptide acetylcholine, released from antral and oxyntic intramural neurons. Gastrin is also a trophic hormone that participates in carcinogenesis. Helicobacter pylori may increase or decrease acid secretion depending upon the acuity and predominant anatomic focus of infection; most patients manifest hypochlorhydria. Despite the fact that proton pump inhibitors (PPIs) are amongst the most widely prescribed drugs, they are underutilized in patients at high risk for UGI bleeding. Although generally considered well tolerated, concerns have been raised regarding associations between PPI use and dementia, kidney disease, myocardial infarction, pneumonia, osteoporosis, dysbiosis, small bowel injury, micronutrient deficiency, and fundic gland polyps. SUMMARY: Our understanding of the physiologic, pathophysiologic, and pharmacologic regulation of gastric secretion continues to advance. Such knowledge is crucial for improved and safe management of acid-peptic disorders.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Acetylcholine/physiology , Gastric Acid/physiology , Gastrins/physiology , Helicobacter Infections/metabolism , Helicobacter pylori , Histamine/physiology , Humans , Proton Pump Inhibitors/pharmacology
9.
Exerc Sport Sci Rev ; 45(1): 16-23, 2017 01.
Article in English | MEDLINE | ID: mdl-27741023

ABSTRACT

In humans, histamine is a molecular transducer of physical activity responses, and antihistamines modify more than 25% of the genes responding to exercise. Although the upstream signal that results in release of histamine within exercising skeletal muscle remains to be identified, it is likely a fundamental exercise response and not an allergic reaction.


Subject(s)
Exercise/physiology , Histamine/physiology , Athletic Performance/physiology , Glucose/metabolism , Glycogen/metabolism , Heart Rate/physiology , Histamine/metabolism , Histamine Antagonists/pharmacology , Humans , Inflammation/physiopathology , Muscle, Skeletal/metabolism , Pain Perception/physiology , Receptors, Histamine/metabolism , Vasodilation/physiology
10.
Cereb Cortex ; 26(4): 1590-1608, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25595181

ABSTRACT

The medial entorhinal cortex (MEC) plays a crucial role in spatial learning and memory. Whereas the MEC receives a dense histaminergic innervation from the tuberomamillary nucleus of the hypothalamus, the functions of histamine in this brain region remain unclear. Here, we show that histamine acts via H1Rs to directly depolarize the principal neurons in the superficial, but not deep, layers of the MEC when recording at somata. Moreover, histamine decreases the spontaneous GABA, but not glutamate, release onto principal neurons in the superficial layers by acting at presynaptic H3Rs without effect on synaptic release in the deep layers. Histamine-induced depolarization is mediated via inhibition of Kir channels and requires the activation of protein kinase C, whereas the inhibition of spontaneous GABA release by histamine depends on voltage-gated Ca(2+) channels and extracellular Ca(2+). Furthermore, microinjection of the H1R or H3R, but not H2R, antagonist respectively into the superficial, but not deep, layers of MEC impairs rat spatial learning as assessed by water maze tasks but does not affect the motor function and exploratory activity in an open field. Together, our study indicates that histamine plays an essential role in spatial learning by selectively regulating neuronal excitability and synaptic transmission in the superficial layers of the MEC.


Subject(s)
Entorhinal Cortex/physiology , Histamine/physiology , Neurons/physiology , Spatial Learning/physiology , Animals , Calcium Channels/physiology , Entorhinal Cortex/drug effects , Glutamic Acid/physiology , Histamine/administration & dosage , Male , Neurons/drug effects , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/physiology , Rats , Rats, Sprague-Dawley , Receptors, Histamine H1/physiology , Spatial Learning/drug effects , Synaptic Potentials/drug effects , gamma-Aminobutyric Acid/physiology
11.
J Physiol ; 594(17): 5009-23, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27061420

ABSTRACT

KEY POINTS: Histamine is a primordial signalling molecule, capable of activating cells in an autocrine or paracrine fashion via specific cell surface receptors, in a variety of pathways that probably predate its more recent role in innate and adaptive immunity. Although histamine is normally associated with pathological conditions or allergic and anaphylactic reactions, it may contribute beneficially to the normal changes that occur within skeletal muscle during the recovery from exercise. We show that the human response to exercise includes an altered expression of thousands of protein-coding genes, and much of this response appears to be driven by histamine. Histamine may be an important molecular transducer contributing to many of the adaptations that accompany chronic exercise training. ABSTRACT: Histamine is a primordial signalling molecule, capable of activating cells in an autocrine or paracrine fashion via specific cell surface receptors. In humans, aerobic exercise is followed by a post-exercise activation of histamine H1 and H2 receptors localized to the previously exercised muscle. This could trigger a broad range of cellular adaptations in response to exercise. Thus, we exploited RNA sequencing to explore the effects of H1 and H2 receptor blockade on the exercise transcriptome in human skeletal muscle tissue harvested from the vastus lateralis. We found that exercise exerts a profound influence on the human transcriptome, causing the differential expression of more than 3000 protein-coding genes. The influence of histamine blockade post-exercise was notable for 795 genes that were differentially expressed between the control and blockade condition, which represents >25% of the number responding to exercise. The broad histamine footprint on the human exercise transcriptome crosses many cellular functions, including inflammation, vascular function, metabolism, and cellular maintenance.


Subject(s)
Exercise/physiology , Histamine/physiology , Transcriptome , Adult , Female , Hemodynamics , Histamine Antagonists/pharmacology , Histamine H1 Antagonists, Non-Sedating/pharmacology , Histamine H2 Antagonists/pharmacology , Humans , Knee/physiology , Male , Muscle, Skeletal/physiology , Ranitidine/pharmacology , Receptors, Histamine H1/physiology , Receptors, Histamine H2/physiology , Terfenadine/analogs & derivatives , Terfenadine/pharmacology , Young Adult
12.
J Pharmacol Exp Ther ; 356(1): 64-73, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26491061

ABSTRACT

Paeoniflorin (PF, C23H28O11), one of the principal active ingredients of Paeonia Radix, exerts depressant effects on the central nervous system. We determined whether PF could modulate sleep behaviors and the mechanisms involved. Electroencephalogram and electromyogram recordings in mice showed that intraperitoneal PF administered at a dose of 25 or 50 mg/kg significantly shortened the sleep latency and increased the amount of non-rapid eye movement (NREM). Immunohistochemical study revealed that PF decreased c-fos expression in the histaminergic tuberomammillary nucleus (TMN). The sleep-promoting effects and changes in c-fos induced by PF were reversed by 8-cyclopentyl-1,3-dimethylxanthine (CPT), an adenosine A1 receptor antagonist, and PF-induced sleep was not observed in adenosine A1 receptor knockout mice. Whole-cell patch clamping in mouse brain slices showed that PF significantly decreased the firing frequency of histaminergic neurons in TMN, which could be completely blocked by CPT. These results indicate that PF increased NREM sleep by inhibiting the histaminergic system via A1 receptors.


Subject(s)
Central Nervous System Depressants/pharmacology , Glucosides/pharmacology , Monoterpenes/pharmacology , Receptor, Adenosine A1/drug effects , Sleep/drug effects , Animals , Electroencephalography/drug effects , Electromyography/drug effects , Gene Expression/drug effects , Genes, fos/drug effects , Glucosides/antagonists & inhibitors , Histamine/physiology , Injections, Intraperitoneal , Male , Mammillary Bodies/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoterpenes/antagonists & inhibitors , Neurons/drug effects , Paeonia/chemistry , Patch-Clamp Techniques , Receptor, Adenosine A1/genetics , Theophylline/analogs & derivatives , Theophylline/pharmacology
13.
Curr Opin Gastroenterol ; 32(6): 452-460, 2016 11.
Article in English | MEDLINE | ID: mdl-27607343

ABSTRACT

PURPOSE OF REVIEW: The present review summarizes the past year's literature, both clinical and basic science, regarding neuroendocrine and intracellular regulation of gastric acid secretion and proper use of antisecretory medications. RECENT FINDINGS: Gastric acid kills microorganisms, modulates the gut microbiome, assists in digestion of protein, and facilitates absorption of iron, calcium, and vitamin B12. The main stimulants of acid secretion are gastrin, released from antral G cells; histamine, released from oxyntic enterochromaffin-like cells; and acetylcholine, released from antral and oxyntic intramural neurons. Other stimulants include ghrelin, motilin, and hydrogen sulfide. The main inhibitor of acid secretion is somatostatin, released from oxyntic and antral D cells. Glucagon-like peptide-1 also inhibits acid secretion. Proton pump inhibitors (PPIs) reduce acid secretion and, as a result, decrease somatostatin and thus stimulate gastrin secretion. Although considered well tolerated drugs, concerns have been raised this past year regarding associations between PPI use and kidney disease, dementia, and myocardial infarction; the quality of evidence, however, is very low. SUMMARY: Our understanding of the physiology of gastric secretion and proper use of PPIs continues to advance. Such knowledge is crucial for improved management of acid-peptic disorders.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Chloride Channels/physiology , Gastrins/physiology , Gastrointestinal Microbiome/drug effects , Histamine/physiology , Humans , Neurosecretory Systems/physiology , Potassium Channels/physiology , Proton Pump Inhibitors/adverse effects , Somatostatin/physiology
14.
J Pharmacol Sci ; 130(4): 212-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26980430

ABSTRACT

Antihistamines inhibit histamine signaling by blocking histamine H1 receptor (H1R) or suppressing H1R signaling as inverse agonists. The H1R gene is upregulated in patients with pollinosis, and its expression level is correlated with the severity of nasal symptoms. Here, we show that antihistamine suppressed upregulation of histidine decarboxylase (HDC) mRNA expression in patients with pollinosis, and its expression level was correlated with that of H1R mRNA. Certain antihistamines, including mepyramine and diphenhydramine, suppress toluene-2,4-diisocyanate (TDI)-induced upregulation of HDC gene expression and increase HDC activity in TDI-sensitized rats. However, d-chlorpheniramine did not demonstrate any effect. The potencies of antihistamine suppressive effects on HDC mRNA elevation were different from their H1R receptor binding affinities. In TDI-sensitized rats, the potencies of antihistamine inhibitory effects on sneezing in the early phase were related to H1R binding. In contrast, the potencies of their inhibitory effects on sneezing in the late phase were correlated with those of suppressive effects on HDC mRNA elevation. Data suggest that in addition to the antihistaminic and inverse agonistic activities, certain antihistamines possess additional properties unrelated to receptor binding and alleviate nasal symptoms in the late phase by inhibiting synthesis and release of histamine by suppressing HDC gene transcription.


Subject(s)
Gene Expression/drug effects , Histamine Antagonists/metabolism , Histamine Antagonists/pharmacology , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Receptors, Histamine H1/metabolism , Toluene 2,4-Diisocyanate/pharmacology , Up-Regulation/drug effects , Animals , Depression, Chemical , Dose-Response Relationship, Drug , Histamine/physiology , Histamine Agonists , Histamine H1 Antagonists , Humans , Hypersensitivity/drug therapy , Hypersensitivity/genetics , Male , Molecular Targeted Therapy , Protein Binding , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects
15.
Pathol Int ; 66(7): 376-85, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27321390

ABSTRACT

We have reported that the function of histamine and its receptors (HRs) has a close relationship with the development of nonalcoholic fatty liver disease (NAFLD). However, much less is known regarding its pathogenic and molecular mechanism(s), including the early stage of hepatic and intestinal function for lipid and bile acid (BA) metabolism. We used H1R and H2R knockout mice (H1/2R-KO) to clarify those pivotal roles in cholesterol/BA metabolism, in which H1/2R-KO mice were separately fed a short-term 1% cholesterol or cholic acid (CA) diet. [(3) H]Cholesterol absorption study revealed that significantly enhanced accumulation occurred in the jejunum, blood and liver, but not in the feces, of H2R-KO mice, compared to wild-type and H1R-KO mice. Furthermore, four weeks after the high-cholesterol diet, the H2R-KO jejunum but not liver exhibited increased expressions of cholesterol transporters, consistent with higher plasma lipoprotein levels. Five days after CA diet, the H2R-KO mice showed significantly higher expressions of ileal BA-reabsorption and hepatic BA-efflux factors, corresponding to higher serum but lower fecal BA levels. The following long-term CA diets resulted in severe injury to the H2R-KO liver. Histamine/H2R signaling might have a protective role in the initial phase during NAFLD progression, correlated with cholesterol and BA metabolism in the liver/intestine.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/metabolism , Histamine/physiology , Non-alcoholic Fatty Liver Disease/physiopathology , Receptors, Histamine H2/metabolism , Signal Transduction , Animals , Liver/physiopathology , Mice
16.
Am J Respir Cell Mol Biol ; 53(6): 802-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25909280

ABSTRACT

Histamine is an important mediator in the pathogenesis of asthma. Variation in genes along the histamine production, response, and degradation pathway may be important in predicting response to antihistamines. We hypothesize that differences exist among single-nucleotide polymorphisms (SNPs) in genes of the histamine pathway between children with allergic versus nonallergic asthma. Children (7-18 yr of age; n = 202) with asthma were classified as allergic or nonallergic based on allergy skin testing. Genotyping was performed to detect known SNPs (n = 10) among genes (HDC, HNMT, ABP1, HRH1, and HRH4) within the histamine pathway. Chi square tests and Cochran-Armitage Trend were used to identify associations between genetic variants and allergic or nonallergic asthma. Significance was determined by P < 0.05 and false-positive report probability. After correction for race differences in genotype were observed, HRH1-17 TT (6% allergic versus 0% nonallergic; P = 0.04), HNMT-464 TT (41% allergic versus 29% nonallergic; P = 0.04), and HNMT-1639 TT (30% allergic versus 20% nonallergic; P = 0.04) were overrepresented among children with allergic asthma. Genotype differences specifically among the African-American children were also observed: HRH1-17 TT (13% allergic versus 0% nonallergic; P = 0.04) and HNMT-1639 TT (23% allergic versus 3% nonallergic; P = 0.03) genotypes were overrepresented among African-American children with allergic asthma. Our study suggests that genetic variation within the histamine pathway may be associated with an allergic versus nonallergic asthma phenotype. Further studies are needed to determine the functional significance of identified SNPs and their impact on antihistamine response in patients with asthma and allergic disease.


Subject(s)
Amine Oxidase (Copper-Containing)/genetics , Asthma/genetics , Histamine N-Methyltransferase/genetics , Histamine/physiology , Receptors, Histamine/genetics , Adolescent , Black or African American , Asthma/ethnology , Asthma/immunology , Child , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide , Signal Transduction , White People
17.
Eur J Neurosci ; 42(7): 2467-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26215659

ABSTRACT

Histamine, a neurotransmitter/neuromodulator implicated in the control of arousal state, exerts a potent phase-shifting effect on the circadian clock in the rodent suprachiasmatic nucleus (SCN). In this study, the mechanisms by which histamine resets the circadian clock in the mouse SCN were investigated. As a first step, Ca(2+) -imaging techniques were used to demonstrate that histamine increases intracellular Ca(2+) concentration ([Ca(2+) ]i ) in acutely dissociated SCN neurons and that this increase is blocked by the H1 histamine receptor (H1R) antagonist pyrilamine, the removal of extracellular Ca(2+) and the L-type Ca(2+) channel blocker nimodipine. The histamine-induced Ca(2+) transient is reduced, but not blocked, by application of the ryanodine receptor (RyR) blocker dantrolene. Immunohistochemical techniques indicated that CaV 1.3 L-type Ca(2+) channels are expressed mainly in the somata of SCN cells along with the H1R, whereas CaV 1.2 channels are located primarily in the processes. Finally, extracellular single-unit recordings demonstrated that the histamine-elicited phase delay of the circadian neural activity rhythm recorded from SCN slices is blocked by pyrilamine, nimodipine and the knockout of CaV 1.3 channel. Again, application of dantrolene reduced but did not block the histamine-induced phase delays. Collectively, these results indicate that, to reset the circadian clock, histamine increases [Ca(2+) ]i in SCN neurons by activating CaV 1.3 channels through H1R, and secondarily by causing Ca(2+) -induced Ca(2+) release from RyR-mediated internal stores.


Subject(s)
Calcium Channels, L-Type/metabolism , Circadian Clocks/physiology , Histamine/physiology , Receptors, Histamine H1/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Calcium Channel Blockers/pharmacology , Dantrolene/pharmacology , Histamine H1 Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Nimodipine/pharmacology , Pyrilamine/pharmacology , Signal Transduction
18.
Curr Opin Gastroenterol ; 31(6): 479-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26376477

ABSTRACT

PURPOSE OF REVIEW: This review summarizes the past year's literature regarding the neuroendocrine and intracellular regulation of gastric acid secretion, discussing both basic and clinical aspects. RECENT FINDINGS: Gastric acid facilitates the digestion of protein as well as the absorption of iron, calcium, vitamin B12, and certain medications. High acidity kills ingested microorganisms and limits bacterial overgrowth, enteric infection, and possibly spontaneous bacterial peritonitis. The main stimulants of acid secretion are gastrin, released from antral gastrin cells; histamine, released from oxyntic enterochromaffin-like cells; and acetylcholine, released from antral and oxyntic intramural neurons. Ghrelin and coffee also stimulate acid secretion whereas somatostatin, cholecystokinin, glucagon-like peptide-1, and atrial natriuretic peptide inhibit acid secretion. Although 95% of parietal cells are contained within the oxyntic mucosa (fundus and body), 50% of human antral glands contain parietal cells. Proton pump inhibitors are considered well tolerated drugs, but concerns have been raised regarding dysbiosis, atrophic gastritis, hypergastrinemia, hypomagnesemia, and enteritis/colitis. SUMMARY: Our understanding of the functional anatomy and physiology of gastric secretion continues to advance. Such knowledge is crucial for improved management of acid-peptic disorders, prevention and management of neoplasia, and the development of novel medications.


Subject(s)
Gastric Acid/metabolism , Gastrins/physiology , H(+)-K(+)-Exchanging ATPase/physiology , Histamine/physiology , Humans , Neurosecretory Systems/anatomy & histology , Neurosecretory Systems/pathology , Neurosecretory Systems/physiology , Proton Pump Inhibitors/adverse effects , Signal Transduction/physiology
19.
Brain ; 137(Pt 2): 313-22, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23794605

ABSTRACT

Research over the past 15 years has helped to clarify the anatomy and physiology of itch, the clinical features of neuropathic itch syndromes and the scientific underpinning of effective treatments. Two itch-sensitive pathways exist: a histamine-stimulated pathway that uses mechanically insensitive C-fibres, and a cowhage-stimulated pathway primarily involving polymodal C-fibres. Interactions with pain continue to be central to explaining various aspects of itch. Certain spinal interneurons (Bhlhb5) inhibit itch pathways within the dorsal horn; they may represent mediators between noxious and pruritic pathways, and allow scratch to inhibit itch. In the brain, functional imaging studies reveal diffuse activation maps for itch that overlap, but not identically, with pain maps. Neuropathic itch syndromes are chronic itch states due to dysfunction of peripheral or central nervous system structures. The most recognized are postherpetic itch, brachioradial pruritus, trigeminal trophic syndrome, and ischaemic stroke-related itch. These disorders affect a patient's quality of life to a similar extent as neuropathic pain. Treatment of neuropathic itch focuses on behavioural interventions (e.g., skin protection) followed by stepwise trials of topical agents (e.g., capsaicin), antiepileptic drugs (e.g., gabapentin), injection of other agents (e.g., botulinum A toxin), and neurostimulation techniques (e.g., cutaneous field stimulation). The involved mechanisms of action include desensitization of nerve fibres (in the case of capsaicin) and postsynaptic blockade of calcium channels (for gabapentin). In the future, particular histamine receptors, protease pathway molecules, and vanilloids may serve as targets for novel antipruritic agents.


Subject(s)
Nervous System Diseases/physiopathology , Pruritus/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzamidines , Guanidines/pharmacology , Guanidines/therapeutic use , Histamine/physiology , Histamine Antagonists/pharmacology , Histamine Antagonists/therapeutic use , Humans , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Nervous System Diseases/diagnosis , Nervous System Diseases/therapy , Pruritus/diagnosis , Pruritus/therapy , Spinothalamic Tracts/drug effects , Spinothalamic Tracts/physiology
20.
Alcohol Alcohol ; 50(1): 51-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25371045

ABSTRACT

AIMS: To clarify the effect of chronic alcohol consumption on the brain histaminergic neurons in rats. METHODS: Male Wistar rats were given 20% ethanol as the only source of drinking during 6 months, control rats had a free access to water. The samples of hypothalamus were prepared for light and electron microscopy accompanied by morphometry to examine the brain histaminergic neurons of E2 group. RESULTS: Chronic ethanol consumption increased the amount of histologically abnormal forms of histaminergic neurons and decreased the whole amount of E2 histaminergic neurons (for 5%). The neuron bodies and nuclei increased in size and sphericity, the nuclear/cytoplasmic ratio decreased by 15%. The ultrastructural changes in histaminergic neurons demonstrate the activation of their nuclear apparatus, both destruction and hypertrophy and hyperplasia of organelles, especially lysosomes. Chronic ethanol consumption induces the disturbances in cytoplasmic enzymes of neurons: increases the activity of type B monoamine oxidase, dehydrogenases of lactate and NADH and, especially, marker enzyme of lysosomes acid phosphatase as well as inhibits the activity of dehydrogenases of succinate and glucose-6-phosphate. CONCLUSION: Chronic alcohol consumption affects significantly the structure and metabolism of the brain histaminergic neurons, demonstrating both the neurotoxic effect of ethanol and processes of adaptation in those neurons, necessary for their survival.


Subject(s)
Alcohol Drinking/adverse effects , Brain/drug effects , Histamine , Hypothalamus/drug effects , Neurons/drug effects , Animals , Brain/cytology , Brain/pathology , Brain/ultrastructure , Histamine/physiology , Hypothalamus/cytology , Hypothalamus/pathology , Hypothalamus/ultrastructure , Male , Microscopy, Electron , Monoamine Oxidase/metabolism , NADH Dehydrogenase/metabolism , Neurons/cytology , Neurons/ultrastructure , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL