Dear reader of ADxS.org, please excuse the disruption.

ADxS.org needs about $63500 in 2024. In 2023 we received donations of about $ 32200. Unfortunately, 99.8% of our readers do not donate. If everyone who reads this request makes a small contribution, our fundraising campaign for 2024 would be over after a few days. This donation request is displayed 23,000 times a week, but only 75 people donate. If you find ADxS.org useful, please take a minute and support ADxS.org with your donation. Thank you!

Since 01.06.2021 ADxS.org is supported by the non-profit ADxS e.V..

$8975 of $63500 - as of 2024-02-29
14%
Header Image
Histamine

Sitemap

Histamine

Histamine is a neurotransmitter and hormone and could play a relevant role in ADHD.
Histamine regulates the release of dopamine. Almost all ADHD medications increase histamine. Histamine appears to be reduced in ADHD.
H3 antagonists, which increase histamine as a result, have a beneficial effect on various ADHD symptoms as well as on social symptoms in ASD. ADHD and ASD very often occur comorbidly. Histamine could also play a role in the day-night rhythm disorders that are common in ADHD.

Histamine is toxic in high doses. Spoiled fish triggers histamine poisoning within 20 minutes.
Histamine is a potent inflammatory mediator with pleiotropic effects that is involved in the regulation of innate and adaptive immunity.1

1. Development of histamine

1.1. Pathway of histamine formation

Conversion of the amino acid L-histidine to histamine by

  • Pyridoxal phosphate-dependent oxidative decarboxylation using the enzyme histidine decarboxylase (EC 4.1.1.22) or
  • Non-specific aromatic L-amino acid decarboxylase

α-fluoromethyl-histidine suppresses histamine synthesis.2

1.2. Site of histamine formation in the brain

Only a few nerve cells in the brain produce histamine.3 Synthesis of histamine and storage in vesicles of:4

  • Tuberomammillary nucleus (TMN) of the hypothalamus

    • Main site of the histamine neurons
      • Human: 64,000 histaminergic neurons
      • Rats: 4,600 in the brain in total
    • TMH cells produce GABA as well as histamine.
    • Project to the cerebral cortex
    • Significantly regulate arousal and alertness.
    • There appear to be 5 groups of TMN cells that differ in the expression of the H3 receptor and in the co-expression of GABA, among other things
    • Deactivation of TMN by the GABA agonist muscimol leads to prolonged REM-free sleep; optogenetic activation of a subpopulation of TMN neurons induces wakefulness5
    • The activity of TMN neurons varies depending on the waking state: it is low in the calm waking state, moderate in the active waking state and highest in the alert waking state.6
    • Histamine maintains alertness through direct projections of TMN cells to the thalamus and cortex and indirectly through activation of cholinergic (via H1 and H2 receptors)7, GABAergic89 and noradrenergic cells (in the locus coeruleus).10
  • Mastocytes (mast cells)

    • Contain considerable amounts of histamine from the brain
    • Are only found in
      • Thalamus
      • Hypothalamus
      • Dura mater
      • Leptomeninx
      • Choroid plexus
  • Microglia

  • Microvascular endothelial cells

  • Projections are widely used, e.g. in4

    • Cerebrum
    • Cerebellum
    • Posterior pituitary gland
    • Spinal cord

1.3. Histamine in the body (peripheral)

Synthesis of histamine and storage in vesicles peripherally in4

  • Mast cells
  • Basophils (basophilic granulocytes, a small subgroup of white blood cells (leukocytes))
  • Epidermal cells
  • Gastric mucosa
    • Enterochromaffin-like cells, control the release of gastric acid

Peripheral histamine is mainly involved in1

  • local immune reactions
  • Digestive system

2. Storage of histamine

Storage bound to heparin in vesicles, primarily in

  • Mast cells
  • Basophilic granulocytes
  • Mucous membranes
  • Bronchi
  • Gastrointestinal tract

3. Release of histamine

Release from vesicles by

  • IgE-mediated allergic reactions of the “immediate type” (type I)
  • Complement factors (e.g. in the case of endotoxin-induced shock)

4. Breakdown / reuptake of histamine

Degradation by diamine oxidases (DOA, extracellular) and aldehyde oxidases (intracellular) to imidazolylacetic acid.
After ribosylation excretion by kidney.
In the case of HNMT deficiency, degradation by DAO can increase.
Only slight degradation due to histamine methylation.

4.1. Dismantling

4.1.1. Degradation in the brain (CNS) primarily due to HNMT

Histamine is inactivated in the brain by the enzyme histamine N-methyltransferase (intracellular) to inactive Nτ-methylhistamine.
HNMT (EC 2.1.1.8) catalyzes the transfer of a methyl group from S-adenosyl-l-methionine (SAM) to histamine, resulting in Nτ-methylhistamine and S-adenosyl-l-homocysteine.11 Nτ-methylhistamine is oxidatively degraded via monoamine oxidases, diamine oxidases (extracellular) and aldehyde oxidases (intracellular) to Nτ-methylimidazolylacetic acid.12

HNMT is found in the human brain in neurons and glial cells in:11

  • Cerebellum (high quantity)
  • frontal cortices (medium quantity)
  • parietal cortices
    • Hippocampus (average amount)
    • Caudate nucleus (medium quantity)
  • temporal cortices
  • occipital cortices

In humans, HNMT shows binding affinity to:11

  • S-Adenosyl-l-methionine (SAM) (Km: 2.0-6.2 µM)
  • Histamine (Km: 13-20 µM)

HNMT knockout mice showed:13

  • drastically increased histamine levels in the brain, intracellular and extracellular
  • other monoamines in the brain unchanged
  • high aggressiveness due to excessive H2R activation
  • reduced movement activity in home cages
    • probably due to disturbed sleep-wake cycle caused by excessive H1R activation
  • no anxiety-like behavior
  • no depression-like behaviors
  • no memory impairment
  • no motor impairments

Inhibitors of HNMT:11

  • Metoprine
    • Blood-brain-barrier-crossing
    • Antinociception
    • Suppression of energy absorption
    • hyperglycemic effect
    • Improvement of cognitive functions
    • antiepileptic effect
    • Attenuation of methamphetamine-induced behavioral problems
    • Inhibition of dihydrofolate reductase
    • Reduction in cellular folate metabolism, which inhibits cell growth
  • SKF91488
    • has difficulty crossing the blood-brain barrier
  • Amodiaquine
  • Chloroquine
  • Dimaprite
  • Etoprine
  • Quinacrine
  • Tacrine

4.1.2. Degradation in the body primarily by diamine oxidase (DAO, histaminase)

Diamine oxidase (DAO, also known as histaminase) is a homodimeric protein and is encoded by the AOC1 gene. DAO is mainly found in the microvilli of enterocytes.
DAO oxidatively deaminates various amines, among others:11

  • Histamine (Km: 19 µM in the intestine)14
  • Cadaverine (1,5-diaminopentane)
  • Putrescine (Km: 83 µM)
  • N Tau-methylhistamine (N-methylhistamine, 1-methylhistamine) (Km: 97 µM)
  • Spermidine
  • Benzylamine (low)
  • Methylamine (low).
    (Km: Michaelis constant; substrate concentration at which the reaction rate becomes half-maximum)

DAO is mainly expressed in the digestive tract, where it serves to detoxify histamine from food in order to reduce histamine uptake by enterocytes. If DAO is impaired, this results in a higher histamine uptake and thus an increased histamine blood level.11
DAO is also expressed to a lesser extent in the kidneys, placenta15 and lungs. DAO plays little to no role in the brain.16
The optimum pH value for the breakdown of histamine by DAO is 6.4 to 6.6.14
DAO dysfunctions appear to play a role in various diseases.

In the intestine:

  • Histamine intolerance17
  • ischemic bowel syndrome14
  • Mesenteric infarction14
  • Ulcerative colitis14

In the placenta15

  • Gestational diabetes
  • threatened and missed miscarriages
  • trophoblastic disorders.

4.2. Resumption

Astrocytes and endothelial cells play an important role in histamine excretion.1819
The plasma membrane monoamine transporter (PMAT) and the organic cation transporter 3 (OCT3) transport histamine in humans mainly into astrocytes, where it is metabolized by HNMT.20
PMAT and OCT 1 to 3 are polyspecific transporters. They transport various monoamines, including serotonin, dopamine, noradrenaline and histamine.
In contrast to the catecholamines (e.g. DAT, NET) and serotonin (SERT), no high-affinity “uptake 1” transporter has been identified for histamine to date. While uptake 1 transporters generally show strong affinity with Km values below 5 μM, the affinity of uptake 2 transporters is considerably weaker with Km values Km (here: for histamine) of at least 100 μM.20 PMAT and OCT 1 to 3 are therefore low-affinity “uptake 2” transporters.21 Nevertheless, “uptake 2” transporters such as OCR3 contribute to reuptake not only at high concentrations, but at any concentration.

4.2.1. Plasma membrane monoamine transporter (PMAT)

The main description of the PMAT can be found at Dopamine reuptake by the plasma membrane monoamine transporter (PMAT) in the article Dopamine reuptake, dopamine degradation

Discovered in 2004,22 PMAT (encoded by the SLC29A4 gene) is widespread in the human brain and appears to be involved in dopamine and serotonin clearance in addition to histamine reuptake.11
PMAT is significantly more involved in histamine reuptake than OCT319 in human astrocytes.20
Whether PMAT reuptakes histamine not only in astrocytes but also presynaptically in histamine neurons remains to be seen.
Km for histamine is at least 100 μM.20
PMAT gene polymorphisms with reduced transport activity for the monoamines serotonin and dopamine as well as the neurotoxin 1-methyl-4-phenylpyridinium (MPP(+)) correlate with autism spectrum disorders (ASD).23
PMAT-KO mice (which therefore have a PMAT deficiency) show neither a strong change in brain histamine levels nor behavioral abnormalities outside of stressful situations.2411

4.2.2. Cation transporter 3, Organic cation transporter 3 (OCT3, SLC22A3)

The main presentation of OCT can be found at Dopamine reuptake by organic cation transporters (OCT) In the article Dopamine reuptake, dopamine degradation

Km of OCT3 for histamine is at least 100 μM.20
Whether OCT3 reuptakes histamine not only in astrocytes but also presynaptically in histamine neurons remains to be seen.20

4.4.3. Cation transporter 2, Organic cation transporter 2 (OCT2, SLC22A2)

In addition to OCT3, the organic cation transporter 2 (OCT2, SLC22A2) also reuptakes histamine, but was not found in human astrocytes,19 but in the human brain, as well as OCT1 (SLC22A1), OCTN1 (SLC22AN1) and OCTN2 (SLC22AN2).25
The human OCT2 transports:20

  • Cations
    • Tetraethylammonium
    • 1-Methyl-4-phenylpyridinium (MPP)
  • Medication
    • Cimetidine (H2R antagonist)
  • Neurotransmitters
    • Histamine
    • Acetylcholine
    • Dopamine
    • Noradrenaline
    • Serotonin
      OCT2 is found in the brain at presynaptic nerve endings in20
  • Cortex
  • Hippocampus
  • Thalamus
  • Hypothalamus
  • dorsal raphe nucleus
  • Locus coeruleus

Km for histamine:20

  • 111 μM (mouse)
  • 0.89 mM (rat)
  • 0.94 mM to 1.3 mM (human).

5. Histamine receptors

Histamine can further influence

  • Learning and memory processes26
  • Thermoregulation26
  • Satiety (due to histamine in the brain)27
  • Energy consumption is increased by histamine in the hypothalamus26
  • Glucose uptake and insulin function in the body26
  • Feeding behavior is reduced by histamine in the hypothalamus26 while histamine generally increases arousal for feeding27
  • Improvement of motor balance and motor coordination via H2 receptors in the cerebellum28
  • Increases motor activity/exploratory behavior via H2 receptors, not via H1 receptors2
  • Increases anxiety primarily via H2 receptors and concomitantly via H1 receptors2

5.1. H1 histamine receptor

  • About as often as H2R4
    • Nevertheless functionally significant as H2R
  • Postsynaptic20
  • Low histamine affinity1
  • Is expressed in various cells, including mast cells1
  • Involved in type 1 hypersensitivity reactions1
  • Coupled to4
    • Gq/11 protein
    • Phospholipase C
      • Promotes inositol trisphosphate (IP3)-dependent Ca2+ release from intracellular Ca2+ stores
      • Is directly involved in the formation of diacylglycerol
        • Activates protein kinase C, which phosphorylates intracellular proteins
    • Activated4
      • AMP kinase
      • Nuclear factor Kappa B
      • Nitric oxide synthases
      • Phospholipase A2 (PLA2)
        • Induces the formation of arachidonic acid
  • Regulatory area
    • Systemic vasodilation (vasodilatation)
    • Reddening of the skin
    • Day-night rhythm
      • Mice without H1 receptors have a disturbed day-night rhythm and are not awakened by H3 antagonists.3
      • Sleep29
    • Vomiting
    • Bronchoconstriction
    • Neurotransmission
    • Possibly antidepressant
    • Possibly anticonvulsant
    • Possibly appetite suppressant
  • Agonists
    • Histamine
    • Histaprodifen
  • Antagonists
    • Loratadine
    • Cetirizine
    • Fexofenadine
    • Doxylamine
    • Diphenhydramine
    • Chlorpheniramine (antagonist / inverse agonist)30

5.2. H2 histamine receptor

  • About as often as H1R4
    • Nevertheless functionally less significant than H1R
  • Postsynaptic20
  • Low histamine affinity1
  • Coupled to4
    Gs
  • Stimulates adenylyl cyclase4
    • Which increases intracellular cyclic adenosine monophosphate (cAMP)
      • Which activates protein kinase A (PKA) and the transcription factor cAMP response element-binding protein (CREB)
  • Blocks the Ca2+-activated potassium conductivity4
  • Inhibits PLA24
  • Inhibits the release of arachidonic acid4
  • Regulatory area
    • Is mainly involved in the cytokine production of Th1 lymphocytes1
    • Gastric acid secretion
    • Reflex tachycardia
  • Agonists
    • Histamine
    • Betazol
  • Antagonists
    • Cimetidine
    • Famotidine
    • Ranitidine
    • Roxatidine

5.3. H3 histamine receptor

  • High histamine affinity1
  • Short
    • Is found on histaminergic neurons (autoreceptor, less common)420
    • On
      • Soma
      • Dendrites
      • Axons
    • Negative feedback to inhibit histamine synthesis and the release of histamine
  • Long
    • Is found on other neurons, including glutamate, acetylcholine, GABA (heteroreceptor), large majority4
      • Other sources report H3R only as an autoreceptor20
    • On axons of the basal ganglia
      • Especially in the dorsal and ventral striatum
      • It is unclear to us: why postsynaptically on axons (the sending neuron parts) and not on dendrites (the receiving ones)?
  • H3R as well as HNMT, H1R and H2R appear to be frequently expressed in ASA.31
  • Signal paths:4
    • Coupled to Gi/o
      • Inhibits adenylyl cyclase and activated Ca2+ channels
      • Controls histamine synthesis and neurotransmitter release
    • Inhibition of the Na+/H+ pump
    • Amplification of G-protein-driven inwardly rectifying K+ channels
    • Activation of phospholipase C
    • Controls mitogen-activated protein kinase (MAPK) pathway
    • Controls phosphatidylinositol 3-kinase (PI3K) pathway
  • Regulatory area
    • Neurotransmission
      Regulation of the release of
      • Histamine (autoregulation)
        • H3 antagonists increase the release of histamine2
      • Acetylcholine
      • Noradrenaline
      • Serotonin
      • Dopamine
      • Glutamate
    • Regulation of the circadian rhythm
    • Involved in the function of the blood-brain barrier1
  • Agonists
    • Histamine
    • Α-Methylhistamine
    • Immepip
    • Imetit
  • Antagonists / inverse agonists
    increase the activity of histaminergic neurons in the brain and thus promote arousal and cognition32
    • Ciproxifan
    • Thioperamide
      • Parkinson’s disease33
      • Increases motor activity / exploratory behavior2
      • Increases anxiety2
    • Pitolisant (antagonist/inverse agonist); Wakix® (pitolisanth hydrochloride)33, selective H3R antagonist/inverse agonist34
      • Narcolepsy, sleep disorders
        • In narcolepsy, a significantly reduced histamine level was found in the brain35
      • No effects on:34
        • Striatal dopamine
          • Other than AMP, Modafinil, Solriamfetol
        • Locomotion
          • Even attenuated hyperlocomotion triggered by modafinil or solriamfetol
        • Food intake
    • H3RA 2-1833
      • Epilepsy
    • Clobenpropit (antagonist, inverse agonist)
      • Apparently directly inhibited dopamine reuptake in vitro, as well as somewhat weaker norepinephrine reuptake in striatal and cerebro-cortical synaptosomes36
    • Iodophenpropit
      • Apparently directly inhibited dopamine reuptake in vitro, as well as somewhat weaker norepinephrine reuptake36
    • ABT-288 (selective H3R antagonist)37
    • ABT-239 [4-(2-{2-[(2R)-2-methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile] (selective H3R antagonist)3839
      • Showed full efficacy in the five-time inhibitory avoidance acquisition model in rat pups at 0.1 mg/kg
      • Full efficacy in the social recognition memory model in adult rats at 0.01 mg/kg.
      • Did not stimulate the locomotor system
      • Minor side effects
    • A-349821 (((4’-(3-((R,R)2,5-dimethyl-pyrrolidin-1-yl)-propoxy)-biphenyl-4-yl)-morpholin-4-yl-methanone))40
    • A-304121 [4-(3-((2R)-2-aminopropanoyl-1-piperazinyl)propoxy)phenyl)cyclopropylmethanone]41
      • improved cognitive performance in a five-trial avoidance test for rat pups (10 mg/kg); similar to thioperamide (10 mg/kg), ciproxifan (3 mg/kg) and GT-2331 (1 mg/kg)42
      • social memory in the adult rat significantly improved by 3 and 10 mg/kg42
    • A-317920 [N-((1R)-2-(4-(3-(4-(cyclopropylcarbonyl)phenoxy)propyl)-1-piperazinyl)-1-methyl-2-oxo-ethyl-)-2-furamide]41
      • improved cognitive performance in a five-trial avoidance test for rat pups (3 mg/kg); similar to thioperamide (10 mg/kg), ciproxifan (3 mg/kg) and GT-2331 (1 mg/kg)42
      • significantly improved social memory in the adult rat at 1 and 3 mg/kg42
    • GT-2331 [(1R,2R)-4-(2-(5,5-dimethylhex-1-ynyl)cyclopropyl)imidazole] (1 mg/kg)42
    • Enerisant (competitive antagonist, inverse agonist)32
      • [1-(4-{3-[(2R)-2-methylpyrrolidin-1-yl]propoxy}phenyl)-1H-pyrazol-4-yl](morpholin-4-yl)Methanone monohydrochloride (enerisant hydrochloride)
      • Binds dose-dependently and selectively to the histamine H3 receptor in the frontal cortex
      • Increased extracellular histamine levels in the posterior hypothalamus
      • Increased dopamine and acetylcholine levels in the medial prefrontal cortex
      • Improved cognition
      • Reversed scopolamine-induced cognitive impairment in a social recognition test and a novel object recognition test at 0.03 to 0.3 mg/kg, p.o.
      • Wakefulness-inducing effects at 3 to 10 mg/kg, p.o.
    • ST-713
      • H3 antagonist (Ki = 1.21 nM)
      • D2 antagonist (Ki = 41 nM)
      • D3 antagonist (Ki = 50 nM)
      • Low affinity to other receptors
        • H1 (Ki = 205 nM)
        • H4 (Ki = 210 nM)
        • D1 (Ki = 232 nM)
        • D5 (Ki = 105 nM)
      • Reduced autistic behavior in male BTBR T+tf/J mice.4344
        • ST-713 (3-(2-chloro-10H-phenothiazin-10-yl)-N-methyl-N-(4-(3-(3-(piperidin-1-yl)propoxy)benzyl)propan-1-amine) improved at 2.5, 5 and 10 mg/kg, i.p. dose-dependent
          • social deficits
          • repetitive/compulsive behaviors
          • disturbed states of anxiety
          • but not the hyperactivity of the mice tested
          • 5 mg attenuated the increased protein levels in the hippocampus and cerebellum of
            • NF-κB p65
            • COX-2
            • iNOS
          • Simultaneous administration of an HR agonist or an anticholinergic drug abolished the improvement in social parameters
    • ST-22234546
      • H3R/D2R/D3R receptor antagonist
      • 2.5, 5 and 10 mg/kg, i.p. significantly and dose-dependently attenuated social deficits and disturbed anxiety in BTBR mice
      • Increase of histamine in
        • Cerebellum
        • Striatum
      • Increase in dopamine in
        • PFC
        • Striatum
      • Increase in acetylcholine in
        • PFC
        • Striatum
        • Hippocampus

5.4. H4 histamine receptor

  • High histamine affinity1
  • Can be found at4
    • Immune cells
      • Mast cells
      • Eosinphils
      • Dendritic cells
    • Microglia (brain)
    • Not yet detected on neurons (presumably problem with detection methods)
  • Homology to H3R of approx. 40 %4
  • Signal paths:4
    • Gi/Go-coupled GPCRs
      • Reduces cAMP accumulation
    • Increases Ca2+ mobilization
    • Activates kinases (ERK, PI3K and MAPK)
    • Activates the transcription factor activating protein-1
  • Regulatory area
    • Mast cell chemotaxis
    • H4R stimulation increases histamine and cytokine production1
  • Agonists
    • Histamine
    • 4-Methylhistamine
    • Immepip47
  • Antagonists
    • Thioperamide
    • JNJ 7777120
      • Inhibits pro-inflammatory microglia and Parkinson’s development in rat model48

5.5. Histamine receptor heteromers

Receptor heteromers are receptor networks consisting of several receptors of different neurotransmitters.
Histamine receptors form heteromers, among other things, as:49
H3R/Dopamine D2R
H3R/Dopamine D1R
H3R/Adenosine A2A

6. Effect of histamine

Histamine influences

  • In the brain4
    • Arousal (arousal)
    • Awakening
    • Maintaining vigilance29
    • Learning and memory4
      • Histamine appears to play an important role in learning and memory via H1R and H2R
      • HDC-deficient mice show
        • specific changes in task-related learning and memory
        • improved performance in passive avoidance and fear memory
        • gender-specific deficiency or an improvement in the recognition of water labyrinths and new locations
      • Pharmacological blockade of H1R impairs
        • the spatial memory
        • the consolidation of object recognition memory
        • the avoidance memory
      • H2R antagonists block the consolidation of object recognition memory and inhibitory avoidance memory
      • Histamine improves both spatial working and reference memory functions through H1R and H2R in the radial maze task after scopolamine-induced memory deficits
      • H1R or H2R knockout mice show
        • Impairments in object recognition
        • Impairments in the acquisition of spatial memory
        • Improvements in auditory and contextual freezing
      • H1R knockout mice also show
        • severe impairment of memory for temporal sequences
        • Feeding
        • Energy
      • H3R antagonists
        • Show protective effect in various cognitive impairments in Y-maze, object recognition, passive avoidance, radial arm maze and water maze tests
        • Improved detection by the H3R antagonist ciproxifan appears to be at least partially dependent on H1R and H2R
        • Showed memory improvements in Alzheimer’s disease
  • Immune system50
    • Histamine stimulates recruitment of mast cells and basophils at the site of inflammation
      • Reaction to various stimuli such as allergens, pathogens and stress
    • In the innate immune system
      • Intensifies inflammatory reaction
      • Can promote the development of chronic inflammation, apparently via H1R
    • In the adaptive immune response
      • Histamine regulates adaptive immune responses at a systemic level (i.e. vascular system, respiratory tract, gut, microbiota, skin and nervous system)
  • The digestive system50
    • H2R antagonists have a gastroprotective effect
      • Gastritis
      • Gastroesophageal reflux
      • Prevention and treatment of peptic ulcers / bleeding when taking non-steroidal anti-inflammatory drugs (NSAIDs)
  • Social behavior
    • H3R antagonists showed improvements in autistic behaviors
  • Migraine
    • Histamine has a vasodilating effect
    • Migraine attacks are less frequent in the evening, correlating with lower central histaminergic arousal
    • Histamine is involved in migraine pathogenesis via inflammatory pathways
    • Plays a decisive role via an inflammatory pathway
      • H3R agonists are said to have anti-nociceptive and anti-neurogenic anti-inflammatory effects
  • Motivation and stress response51
    • Acute immobilization stress acts via histamine signaling at glutamatergic synapses of D1 receptor-expressing [D1(+)] medium spiny neurons (MSNs) in the nucleus accumbens nucleus
    • Histamine inhibits excitatory gain on D1(+) MSNs via presynaptic H3 receptor-dependent long-term depression, which requires Gβγ-driven Akt-GSK3β signaling
    • Histamine asymmetrically regulates glutamatergic transmission from the PFC and mediodorsal thalamus, with inputs from the PFC cortex undergoing robust long-term depression by histamine
    • Acute immobilization stress attenuates this long-term depression by recruiting endogenous H3 receptor signaling in the nucleus accumbens at glutamatergic synapses on D1(+) MSNs

7. Disorders of the histamine system

Malfunctions are associated with neuropathological disorders, e.g.12

  • Narcolepsy

    • A significantly reduced brain histamine level was found in narcolepsy35
  • Hallucinations

  • Schizophrenia-like conditions
    An influence of histamine on schizophrenia itself has not yet been proven.52

  • Sleep problems

    • Histamine neurons4
      • Stop firing during the transition from wakefulness to sleep
      • Silence during slow-wave sleep and REM sleep
      • Start firing again after the transition to the waking state
      • Lowest firing in the awake idle state
      • Moderate firing during active wakefulness
      • Maximum firing with high vigilance
    • HDC knockout mice (impaired histamine synthesis) show
      • Sleep fragmentation
      • Increased REM sleep during the light period
      • Significant alertness deficits at the onset of darkness
    • H1 receptor antagonists can help with insomnia52
    • H1R antagonists promote sleep
    • Pathways of histamine on sleep:4
      • The ventrolateral preoptic nucleus (VLPO) is associated with the promotion of sleep
      • Histamine inhibits VLPO neurons indirectly by activating GABAergic interneurons, which in turn disinhibits histaminergic neurons to promote wakefulness
      • Histaminergic axons release paracrine GABA in the neocortex to prevent overactivation by histamine and regulate the level of alertness
      • Histamine can also directly modulate glutamatergic neurons of the thalamus and induce a general excitatory effect in several brain regions
      • Histamine triggers cortical activation through:
        • Activation of cholinergic neurons in the basal forebrain
        • Activation of cholinergic neurons in the mesopontine tegmentum
        • Activation of serotonergic neurons in the dorsal raphe nucleus
        • Direct projections into the cortex
      • The hypocretin system maintains wakefulness largely via histaminergic neurons
      • HCRT neurons and histaminergic neurons
        • Lie next to each other in the human hypothalamus
        • Overlap in their projections
        • HCRT neurons directly excite histaminergic neurons via the HCRT receptor 2
      • Histamine possibly regulates HCRT neurons via the H1R
        • The H1R antagonist pyrilamine inhibits HCRT-induced arousal in rats
        • H1R knockout mice show no HCRT-induced increase in alertness
  • Tourette (rare)

  • Alzheimer’s and Parkinson’s

    • High histamine levels in the substantia nigra correlate with a reduced number of dopaminergic cells
    • The H1 receptor appears to be affected52
  • Huntington

  • Depression

    • Reduced H1 receptor binding

7.1. Histamine deficiency

  • Tourette syndrome5354
    Histidine decarboxylase knockout (HDC-KO) mice exhibit stereotypic locomotor behaviors that reflect the core phenomenology of Tourette’s.55
  • Rare gene variants of the histamine receptor gene appear to be involved in Tourette’s and autism spectrum disorders.5657

7.2. Histamine excess / histamine intolerance

7.2.1. Development of histamine intolerance

A very good description of histamine intolerance can be found at www.histaminintoleranz.ch.58

Histamine obesity can have various causes:

  • Excessive histamine intake (food, fish poisoning)
  • Too little histamine breakdown
    • Mostly diamine oxidase deficiency (peripheral)
    • HNMT deficiency (CNS)
  • Smoking appears to increase histamine levels5960616263
    Histamine moderates reactions to cigarette smoke.64 However, reports that smoking reduces histamine levels65 or leaves them unchanged66 are the exception. However, it is conceivable that smoking increases the reaction to histamine.67686970
  • The cause may also be a systemic mast cell activation disease (MCAD)
    Pathologically altered mast cells (mastocytes, a type of immune cell used to defend against foreign bodies) produce histamine and other messenger substances (mediators). The incidence of MCAD is estimated to be between 1 and 17%.71
    Very good presentation of MCAD at https://www.mastzellaktivierung.info/72
    MCAD works primarily, but not only, by means of histamine.
    • Types of MCAD:
      • Mast cell activation syndrome (MCAS)
      • Systemic mastocytosis (SM) (rare)
      • Mast cell leukemia (MCL) (rare)
    • Mechanisms of action of an MCAD:73
      Percentages indicate the consensus that the mediators mentioned play a role in MCAD.
      • Histamine
        • Headache
        • Low blood pressure
        • Hives (red wheals, urticaria)
          • With or without angioedema (rapidly developing painless swelling)
        • Itching (pruritus)
        • Diarrhea
      • Prostaglandin-D2 (PGD2) (95 %)
        • Mucus secretion
        • Constricted airways (bronchoconstriction)
          • In interaction with thromboxane and PGF2α
        • Vascular instability (dilation of the blood vessels)
        • Sleep inducing
        • Lowering body temperature
        • Possible cause of hereditary hair loss in men together with the steroid hormone dihydrotestosterone (DHT)74
      • PAF2 (platelet-activating factor) (90 %)
        • Abdominal cramps
        • Pulmonary edema
        • Urticaria
        • Bronchoconstriction
        • Hypotension
        • Cardiac arrhythmia
      • Proinflammatory cytokines (80 %)
        • Local inflammation
        • Edema formation
        • Leukocyte migration 80 %
      • LTC4 and LTD4 (80 %)
        • Mucus secretion
        • Edema formation
        • Vascular instability
      • Chemokines (70 %)
        • Acute inflammation
        • Leukocyte recruitment
        • Leukocyte migration
      • Tryptase (65 %)
        • Endothelial activation with subsequent inflammatory reactions
      • Leukotrienes75
        • Allergic reactions
        • Inflammatory reactions

An excessive histamine level causes pseudoallergic symptoms. These vary greatly from person to person, making a diagnosis based on a list of symptoms very difficult.

7.2.2. Frequency of histamine intolerance

The prevalence is 1% of the population. 80 % of those affected are women, 20 % men.
More recent studies arrive at higher prevalence values.

7.2.3. Possible symptoms of histamine intolerance

  • Skin
    • Reddening of the skin
    • Hives
    • Eczema
    • Itching[7]
  • Head
    • Headache
    • Feeling of heat
    • Migraine
    • Dizziness
  • Airways
    • Narrowed or runny nose
    • Breathing difficulties
    • Bronchial asthma
    • Sore throat
  • Digestive system
    • Flatulence (bloating)
    • Diarrhea
    • Constipation
    • Nausea/vomiting
    • Abdominal pain
    • Stomach stitches
    • Heartburn
  • Cardiovascular system
    • Changes in blood pressure
      • High blood pressure (hypertension)
      • Low blood pressure (hypotension)
    • Palpitations (tachycardia)
    • Cardiac arrhythmia
  • Urology
    • Menstrual cramps (dysmenorrhea)
    • Cystitis
    • Urethritis
    • Irritation of the mucous membranes of the female genitals
  • Fabric
    • Water retention (edema)
    • Bone marrow edema (BME)
    • Joint pain
  • Energy balance
    • States of exhaustion
    • Seasickness
    • Tiredness
    • Sleep disorders
  • Mental symptoms
    • Confusion
    • Nervousness
    • Depressive moods

7.2.4. Foods that increase histamine

A list of histamine-increasing foods can be found at 58
There are various ways in which food can increase histamine levels.

7.2.4.1. Effects of the histamine increase
7.2.4.1.1. Containing histamine

Foods that contain histamine increase the histamine level.

7.2.4.1.2. Histamine liberators

Some foods cause an increased release of histamine from the salivary vesicles.

7.2.4.1.3. DAO inhibition

Certain substances inhibit the breakdown of histamine by diamine oxidase (DAO).

7.2.4.1.4. DAO mining competitors

Some foods contain substances that also require diamine oxidase (DAO) to break down histamine, which means that less DAO is available to break down histamine.

7.2.4.1.5. Increase in intestinal permeability for histamine

Substances that increase the permeability of the intestinal wall also increase the absorption of histamine.

7.2.4.2. List of triggers for histamine intolerance and MCAD

A very good compilation of MCAD triggers can be found at https://www.mastzellaktivierung.info/76

Foods with high histamine levels are listed in Quade, Bailly, Bartling, Bliesener, Springer: Histamine intolerance.77 This presentation only concerns foods with a high histamine content, not e.g. histamine liberators or DAO degradation competitors.

7.2.5. Treatment of histamine intolerance

The treatment of first choice is a low histamine diet.

A strict low-histamine diet for one month often helps to completely empty the histamine stores. After that, a limited consumption of individual histamine-increasing foods is usually possible. Smoking increases the histamine level considerably and thus undermines the histamine diet.60

In addition, the missing DAO enzyme can be taken 15 to 30 minutes before meals. Taking DAO can only compensate for individual “sins”, but cannot fundamentally avoid a diet.

8. Histamine and ADHD

There is hardly any positive knowledge about a correlation between histamine intolerance and ADHD. NCBI / Pubmed did not find a single article under “histamine intolerance adhd”.78

A large cohort study found that taking antihistamines (especially first-generation antihistamines) in the first years of life significantly increased the risk of ADHD later in life. A disturbance of REM sleep was cited as a possible cause, which secondarily impaired brain maturation.79
According to another study, former use of antihistamines in neurodermatitis sufferers increased ADHD symptoms.80

Food additives (here: Sun yellow, carmoisine, tartrazine, ponceau 4R; quinoline yellow, allura red, sodium benzoate) can cause histamine release from circulating basophils. This is not allergic, i.e. not dependent on immunoglobulin E. The increased release of histamine can increase ADHD symptoms in carriers of certain gene variants of the genes that encode histamine-degrading enzymes81

A report on 4 individual cases of children with ADHD with learning disabilities describes a very large improvement in ADHD symptoms with antihistamines.82

The H3 histamine receptor is thought to be involved in arousal, control of pituitary hormone release, cognitive function, motivation, goal-directed behavior, memory and sleep-wake cycles. However, clinical trials of H3 receptor drugs for ADHD (MK-0249, Bavisant, PF-03654746) were unsuccessful in phase 2 trials or were discontinued in phase 2 (betahistine).83

8.1. Histamine often elevated in ADHD

8.1.1. Histamine degradation by DAO often reduced in ADHD

A Spanish study found a genetically reduced diamine oxidase (DAO) activity in 82.1% of 40 children with ADHD and a greatly reduced DAO activity in 15.2% (i.e. reduced histamine degradation and consequently a (peripherally) increased histamine level).50 The study is to be extended to 200 children with ADHD and 100 controls. DAO is also called ABP1.

8.1.2. Histamine degradation by HNMT often reduced in ADHD

Furthermore, a correlation between ADHD and reduced histamine N-methyltransferase has been described, which also leads to reduced histamine degradation and increased histamine.11
Various HNMT gene polymorphisms are relevant in ADHD and other disorders:

  • Thr105Ile (rs11558538) showed81
    • reduced thermal stability
    • reduced activity of HNMT84, consequently less histamine degradation
    • more adverse effect of food additives on ADHD symptoms in 3- and 8/9-year-old children
    • a reduced risk of Parkinson’s disease85
  • T939C showed
    • more hyperactivity81
    • more adverse effect of food additives on ADHD symptoms in 3- and 8/9-year-old children81

Other gene variants and their influences on HNMT enzyme activity are known, but without reports of an ADHD correlation:

  • G179A showed
    • impaired HNMT enzyme activity
    • mental disability86
  • C314T showed
    • reduced HNMT enzyme activity
    • Km value for histamine increased 1.3-fold (= lower binding affinity)
    • Km value for SAM increased 1.8-fold (= lower binding affinity)
    • rather no correlation with asthma or rhinitis
  • T632C showed
    • impaired HNMT enzyme activity
    • mental disability86
  • A939G (also known as C939T/ rs1050891)81 shows
    • increased HNMT mRNA stability
    • increased HNMT enzyme activity87

8.2. Histamine and dopamine

Animal studies found a correlation between high histamine levels in the substantia nigra and a breakdown of dopaminergic cells, which causes a reduced dopamine level.12 So far, no therapeutic benefit of H3 antagonists (which reduce histamine levels and increase dopamine levels) on Alzheimer’s or ADHD has been found.52

Receptor heteromers:4

  • H3R - D1R - Heteromers
    • D1R activation inhibits the production of cAMP instead of stimulating it as usual
    • H3R activation should reduce D13 affinity
  • H3R - D2R - Heteromers
    • only appear to reduce the dopamine affinity of the receptor

The histaminergic system appears to be closely linked to the dopaminergic system and synaptic transmission in the striatum (collection according to Hu, Chen (2017), unless another source is given):4

  • Histaminergic neurons can also release dopamine (or GABA) as well
  • Over 85% of D1R- and D2R-expressing MSNs in the dorsal and ventral striatum contain H3R88
    • The synergistic effects of ciproxifan and halperidol suggest a direct, functional H3/D2 receptor interaction in striatopallidal neurons, so that H3R antagonists could be useful tools to improve the symptomatic treatment of schizophrenia
  • Histamine can selectively activate microglia, leading to increased inflammation characteristic of PD pathology, and damage dopaminergic neurons of the SNc
  • The H3R agonist Immepip alleviates apomorphine-induced turning behavior in 6-hydroxydopamine (6-OHDA)-lesioned rats
  • Simultaneous administration of Immepip or Imetit with L-DOPA alleviates L-DOPA-induced dyskinesia or chorea, but not dystonia
  • Intranigral injection of Immepip increases turning after systemic apomorphine administration in rats
  • The H3R antagonist thioperamide alleviates apomorphine-induced stereotypic behavior in rats with 6-OHDA lesions
  • The H3R antagonist pitolisant alleviates excessive daytime sleepiness in patients with Parkinson’s disease; motor performance remains unchanged.
  • Histamine H2R levels in the striatum are unchanged in patients with Parkinson’s disease. Nevertheless, the H2R antagonists famotidine and ranitidine enhance the antiparkinsonian effect of L-DOPA in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson’s disease in macaques and in the 6-OHDA model in rats. H2R antagonists could therefore serve as an aid in the treatment of Parkinson’s disease.
  • The TMN contains a group of dopaminergic neurons in the TMN that share electrophysiological properties with histaminergic neurons
  • H3Rs are expressed on GABAergic input terminals from the substantia nigra reticulata. Stimulation of these receptors reduces GABA release, resulting in increased excitation of dopaminergic cells in the substantia nigra pars compacta
  • Histamine inhibits dopamine release in the mouse striatum via H3R, unlike H3R antagonists
  • Histamine inhibits both cortical and thalamic excitatory projections to MSNs via presynaptic H3Rs
  • Histamine can selectively modulate the dynamics of thalamostriatal synapses to facilitate thalamic inputs
  • Histamine depolarizes both MSN classes via the effect of the H2R
  • Histamine suppresses lateral feedback inhibition between MSN through the H3R or H2R
  • The effect of H2R may be indirectly caused by the activation of H2R in cholinergic interneurons to increase acetylcholine release in the striatum
  • H3R agonists modulated in D2R-SPNs (but not in D1R-SPNs)88
    • Akt (serine/threonine PKB) glycogen synthase kinase 3 beta signaling in response to D2R activation via a β-arrestin 2 dependent mechanism
    • The phosphorylation of mitogen- and stress-activated protein kinase 1 and rpS6 (ribosomal protein S6) remained unchanged
  • The selective H3R agonist R-(-)-α-methylhistamine dihydrobromide attenuates movement activity and stereotypical behavior triggered by D2R agonists88
  • H1R and H2R are co-localized on D1R- and D2R-expressing MSNs and mediate histamine-induced excitation of the two neuron types89
  • Histamine excites dopamine D1R- and D2R-expressing neurons in the striatum equally strongly. This takes place via postsynaptic H1R and H2R.89
  • Genetic histamine deficiency causes upregulation of dopamine neurotransmission90
  • Increased histaminergic innervation of the entopeduncular nucleus (EPN) in a mouse model of Parkinson’s disease activates parvalbumin neurons (PV) of the EPN that project to the thalamic motor nucleus via hyperpolarization-activated cyclic nucleotide-gated channels (HCN) coupled to postsynaptic H2R. Presynaptic H3R activation in the glutamatergic neurons of the subthalamic nucleus (STN), which project to the EPN, inhibits this. Activation of both receptor types improves the motor dysfunction associated with parkinsonism. Pharmacologic activation of H2R such as genetic upregulation of HCN2 in EPNPV neurons, which reduce neuronal burst firing, improves parkinsonism-associated motor dysfunction independent of changes in neuronal firing rate. Optogenetic inhibition of EPNPV neurons and pharmacological activation or genetic upregulation of H3R in EPN-projecting STNGlu neurons ameliorate parkinsonism-associated motor dysfunction by reducing firing rate rather than altering the firing pattern of EPNPV neurons91
  • In embryos, histamine appears to decrease dopaminergic gene transcription by altering several epigenetic components associated with DNA and histone modifications, affecting the development of mDA neurons during development. Histamine showed a long-term effect on the formation of the nigrostriatal and mesolimbic/mesocortical pathways. Histamine caused:92
    • A significant decrease in TH immunoreactivity in the midbrain
    • Changes in dopaminergic neuronal fibers
    • A significantly smaller TH-positive area in the forebrain with whole-body staining
  • The H3R agonist α-methylhistamine decreased dopamine release in the ventral (but not in the dorsal) striatum by reducing the activity of striatal cholinergic interneurons93
  • HDC-KO mice lack the histamine-producing enzyme histidine decarboxylase (HDC) and therefore also histamine. HDC-KO mice showed:94
    • A pronounced pattern of behavior when exploring a new environment, in particular
    • An increased frequency of sitting up against the wall, jumping and head/body shaking.
    • Reduced dopamine and serotonin levels in the striatum
    • Increased DOPAC dopamine metabolite levels
    • Reduced gene expression of dynorphin and enkephalin
    • Increased striatal dopamine turnover after treatment with the dopamine precursor l-dopa
  • An H3R antagonist95
    • Attenuated the D1R-induced cell death signaling and neuronal degeneration that exists in Huntington’s disease
    • Reduced cognitive and motor learning deficits and loss of D1R-H3R heteromer expression in the Huntington’s disease mouse model
  • H3R antagonists (GT-233, 1 mg/kg s.c. and ciproxifan, 3 mg/kg s.c.) significantly and dose-dependently improved the learning performance of SHR pups, as did methylphenidate (1 and 3 mg/kg s.c.) and ABT-418 (agonist at nicotinic acetylcholine receptors, 0.03 mg/kg s.c.). The H3R agonist (R)-alpha-methylhistamine (3 mg/kg s.c.) blocked the cognition-enhancing effect of ciproxifan.96
  • H3R regulate the striatum97
  • An HNMT inhibitor reduced dopamine and histamine turnover in the striatum, nucleus accumbens and hypothalamus98
  • H3R are co-expressed with D1R by striato-nigral medium spiny GABAergic neurons, where they functionally antagonize D1R-mediated responses47
  • H3R-KO fish show reduced dopamine and serotonin levels99

8.3. Habenula, ADHD and histamine

Early childhood lesions of the habenula cause behavioral and brain changes similar to those in ADHD.100
Histamine H3 receptor antagonists eliminate these symptoms.101

The habenula

  • Transmits limbic information to the midbrain monoamine system
    • Is thereby involved in the regulation of monoamine release in target brain areas such as the striatum, where some of the biological substrates process time perception.
  • Is part of the circadian rhythm network and involved in sleep regulation

ADHD often shows changes in the circadian rhythm, sleep disorders and time perception.

8.4. Histamine in spontaneously hypertensive rat (SHR)

The SHR is the most widely used model animal for ADHD.
Compared to WKY, the SHR:102

  • Histamine
    • increased in
      - Hypothalamus (anterior and posterior) of young and adult SHR
      - Brain stem of young SHR
    • unchanged in
      • Cortex-midbrain
        • but higher in adult WKY and SHR than in young
    • reduced histamine metabolism in:103
      • Hypothalamus
      • Brain stem
      • Chronic L-histidine administration did not affect the hypertension of the SHR
        • different: Chronic administration of L-histidine (100 mg / kg twice daily for 4 weeks) to young SHR prevented the age-related increase in blood pressure and urine norepinephrine levels typical of SHR.104
    • increased histamine release from mast cells105106
      • with reduced nitric oxide release from mast cells105
  • Histidine decarboxylase activity
    • unchanged in
      • Posterior hypothalamus of young and adult SHR
      • Medulla oblongata of young and adult SHR
      • Hypothalamus anterior young SHR
      • Cortex-midbrain and in the brainstem of adult SHR (but higher than in young SHR and WKY)
    • slightly increased
      • Hypothalamus anterior adult SHR
    • increased
      • in the midbrain cortex of young and adult SHR
  • Histamine N-methyltransferase
    • increased
      • in the midbrain cortex of young SHR
    • reduced
      • in the cortex-midbrain of adult SHR
  • H3 receptor107
    • reduced number of amplificates
    • increasing H3 receptor density in the cortex with age as the number of expressed amplicons decreases
    • despite the decrease in the number of expressed amplicons of the H3 receptor, the expression of the larger amplicon (~500 bp) increased

Depletion of histamine in young SHR caused a delay in the age-typical rise in blood pressure.108
Chronic administration of L-histidine (100 mg / kg twice daily for 4 weeks) to young SHR prevented the age-related increase in blood pressure and urine norepinephrine levels typical of SHR.104
Histamine appears to lower blood pressure in SHR by attenuating sympathetic production via the central histamine H3 receptor. The antihypertensive effects of L-histidine correlated with an increase in nitric oxide in the rostral ventrolateral medulla.

H3R antagonists (GT-233, 1 mg/kg s.c. and ciproxifan, 3 mg/kg s.c.) (which have the effect of increasing histamine) significantly and dose-dependently improved the learning performance of SHR pups, as did methylphenidate (1 and 3 mg/kg s.c.) and ABT-418 (agonist at nicotinic acetylcholine receptors, 0.03 mg/kg s.c.). The H3R agonist (R)-alpha-methylhistamine (3 mg/kg s.c.) blocked the cognition-enhancing effects of ciproxifan.96

In SHR, the histamine doses that make the blood-brain barrier more permeable deviate upwards or downwards.109

8.5. Almost all ADHD medications increase histamine

All common ADHD medications increase histamine. MPH and AMP also increase the breakdown of histamine by DAO.
Only viloxazine does not appear to increase histamine.

More on this under Histamine intolerance / mast cell activation syndrome In the article Choice of medication for ADHD or ADHD with comorbidity


  1. Blasco-Fontecilla H (2023): Is Histamine and Not Acetylcholine the Missing Link between ADHD and Allergies? Speer Allergic Tension Fatigue Syndrome Re-Visited. J Clin Med. 2023 Aug 17;12(16):5350. doi: 10.3390/jcm12165350. PMID: 37629392; PMCID: PMC10455974.

  2. Mohsen, Yoshikawa, Miura, Nakamura, Naganuma, Shibuya, Iida, Harada, Okamura, Watanabe, Yanai (2014): Mechanism of the histamine H3 receptor-mediated increase in exploratory locomotor activity and anxiety-like behaviours in mice, Neuropharmacology, Volume 81, 2014, Pages 188-194, ISSN 0028-3908, https://doi.org/10.1016/j.neuropharm.2014.02.003.

  3. Provensi, Costa, Izquierdo, Blandina, Passani (2018): Brain histamine modulates recognition memory: possible implications in major cognitive disorders. Br J Pharmacol. 2018 Aug 21. doi: 10.1111/bph.14478.

  4. Hu W, Chen Z (2017): The roles of histamine and its receptor ligands in central nervous system disorders: An update. Pharmacol Ther. 2017 Jul;175:116-132. doi: 10.1016/j.pharmthera.2017.02.039. PMID: 28223162. REVIEW

  5. Fujita, Bonnavion, Wilson, Mickelsen, Bloit, Lecea, Jackson (2017): Hypothalamic Tuberomammillary Nucleus Neurons: Electrophysiological Diversity and Essential Role in Arousal Stability. Journal of Neuroscience 27 September 2017, 37 (39) 9574-9592; DOI: 10.1523/JNEUROSCI.0580-17.2017

  6. Takahashi, Lin, Sakai (2006): Neuronal Activity of Histaminergic Tuberomammillary Neurons During Wake–Sleep States in the Mouse. Journal of Neuroscience 4 October 2006, 26 (40) 10292-10298; DOI: https://doi.org/10.1523/JNEUROSCI.2341-06.2006

  7. Khateb, Fort, Pegna, Jones, Mühlethaler (1995): Cholinergic nucleus basalis neurons are excited by histamine in vitro, Neuroscience, Volume 69, Issue 2, 1995, Pages 495-506, ISSN 0306-4522, https://doi.org/10.1016/0306-4522(95)00264-J

  8. Xu, Michelsen, Wu, Morozova, Panula, Alreja (2004): Histamine innervation and activation of septohippocampal GABAergic neurones: involvement of local ACh release. The Journal of Physiology, 561: 657-670. doi:10.1113/jphysiol.2004.071712

  9. Korotkova, Haas, Brown (2002): Histamine excites GABAergic cells in the rat substantia nigra and ventral tegmental area in vitro, Neuroscience Letters, Volume 320, Issue 3, 2002, Pages 133-136, ISSN 0304-3940, https://doi.org/10.1016/S0304-3940(02)00050-2

  10. Korotkova, Sergeeva, Ponomarenko, Haas (2005): Histamine excites noradrenergic neurons in locus coeruleus in rats. Neuropharmacology, Volume 49, Issue 1, 2005, Pages 129-134, ISSN 0028-3908, https://doi.org/10.1016/j.neuropharm.2005.03.001

  11. Yoshikawa T, Nakamura T, Yanai K (2019): Histamine N-Methyltransferase in the Brain. Int J Mol Sci. 2019 Feb 10;20(3):737. doi: 10.3390/ijms20030737. PMID: 30744146; PMCID: PMC6386932. REVIEW

  12. Shan, Bao, Swaab (2017): Changes in Histidine Decarboxylase, Histamine N-Methyltransferase and Histamine Receptors in Neuropsychiatric Disorders. In: Hattori Y., Seifert R. (eds) Histamine and Histamine Receptors in Health and Disease. Handbook of Experimental Pharmacology, vol 241.

  13. Yoshikawa T, Nakamura T, Yanai K (2018): [Analysis of brain histamine clearance using genetically engineered mice]. Nihon Yakurigaku Zasshi. 2018;152(1):16-20. Japanese. doi: 10.1254/fpj.152.16. PMID: 29998947.

  14. Biegański T, Kusche J, Lorenz W, Hesterberg R, Stahlknecht CD, Feussner KD (1983): Distribution and properties of human intestinal diamine oxidase and its relevance for the histamine catabolism. Biochim Biophys Acta. 1983 Mar 31;756(2):196-203. doi: 10.1016/0304-4165(83)90092-2. PMID: 6403048.

  15. Maintz L, Schwarzer V, Bieber T, van der Ven K, Novak N (2008): Effects of histamine and diamine oxidase activities on pregnancy: a critical review. Hum Reprod Update. 2008 Sep-Oct;14(5):485-95. doi: 10.1093/humupd/dmn014. PMID: 18499706. REVIEW

  16. McGrath AP, Hilmer KM, Collyer CA, Shepard EM, Elmore BO, Brown DE, Dooley DM, Guss JM (2009): Structure and inhibition of human diamine oxidase. Biochemistry. 2009 Oct 20;48(41):9810-22. doi: 10.1021/bi9014192. PMID: 19764817; PMCID: PMC2791411.

  17. Maintz L, Novak N (2007): Histamine and histamine intolerance. Am J Clin Nutr. 2007 May;85(5):1185-96. doi: 10.1093/ajcn/85.5.1185. PMID: 17490952. REVIEW

  18. Huszti Z (2003): Histamine uptake into non-neuronal brain cells. Inflamm Res. 2003 Apr;52 Suppl 1:S03-6. doi: 10.1007/s000110300028. PMID: 12755385. REVIEW

  19. Yoshikawa T, Naganuma F, Iida T, Nakamura T, Harada R, Mohsen AS, Kasajima A, Sasano H, Yanai K (2013): Molecular mechanism of histamine clearance by primary human astrocytes. Glia. 2013 Jun;61(6):905-16. doi: 10.1002/glia.22484. PMID: 23505051.

  20. Naganuma F, Yoshikawa T (2021): Organic Cation Transporters in Brain Histamine Clearance: Physiological and Psychiatric Implications. Handb Exp Pharmacol. 2021;266:169-185. doi: 10.1007/164_2021_447. PMID: 33641029. (REVIEW)

  21. Gasser PJ, Lowry CA (2018): Organic cation transporter 3: A cellular mechanism underlying rapid, non-genomic glucocorticoid regulation of monoaminergic neurotransmission, physiology, and behavior. Horm Behav. 2018 Aug;104:173-182. doi: 10.1016/j.yhbeh.2018.05.003. PMID: 29738736; PMCID: PMC7137088. REVIEW

  22. Engel K, Zhou M, Wang J (2004): Identification and characterization of a novel monoamine transporter in the human brain. J Biol Chem. 2004 Nov 26;279(48):50042-9. doi: 10.1074/jbc.M407913200. PMID: 15448143.

  23. Adamsen D, Ramaekers V, Ho HT, Britschgi C, Rüfenacht V, Meili D, Bobrowski E, Philippe P, Nava C, Van Maldergem L, Bruggmann R, Walitza S, Wang J, Grünblatt E, Thöny B (2014): Autism spectrum disorder associated with low serotonin in CSF and mutations in the SLC29A4 plasma membrane monoamine transporter (PMAT) gene. Mol Autism. 2014 Aug 13;5:43. doi: 10.1186/2040-2392-5-43. PMID: 25802735; PMCID: PMC4370364.

  24. Duan H, Wang J (2013): Impaired monoamine and organic cation uptake in choroid plexus in mice with targeted disruption of the plasma membrane monoamine transporter (Slc29a4) gene. J Biol Chem. 2013 Feb 1;288(5):3535-44. doi: 10.1074/jbc.M112.436972. PMID: 23255610; PMCID: PMC3561572.

  25. Koepsell H, Lips K, Volk C (2007): Polyspecific organic cation transporters: structure, function, physiological roles, and biopharmaceutical implications. Pharm Res. 2007 Jul;24(7):1227-51. doi: 10.1007/s11095-007-9254-z. PMID: 17473959. REVIEW

  26. Tabarean (2016): Histamine receptor signaling in energy homeostasis, Neuropharmacology, Volume 106, 2016, Pages 13-19, ISSN 0028-3908, https://doi.org/10.1016/j.neuropharm.2015.04.011.

  27. Provensi, Blandina, Passani (2016): The histaminergic system as a target for the prevention of obesity and metabolic syndrome, Neuropharmacology, Volume 106, 2016, Pages 3-12, ISSN 0028-3908, https://doi.org/10.1016/j.neuropharm.2015.07.002.

  28. Zhang, Zhuang, Li, Wu, Yung, Zhu, Wang (2016): Selective Modulation of Histaminergic Inputs on Projection Neurons of Cerebellum Rapidly Promotes Motor Coordination via HCN Channels. Mol Neurobiol. 53:1386-401.

  29. Thakkar (2011): Histamine in the regulation of wakefulness, Sleep Medicine Reviews, Volume 15, Issue 1, 2011, Pages 65-74, ISSN 1087-0792, https://doi.org/10.1016/j.smrv.2010.06.004.

  30. Márquez-Valadez B, Aquino-Miranda G, Quintero-Romero MO, Papacostas-Quintanilla H, Bueno-Nava A, López-Rubalcava C, Díaz NF, Arias-Montaño JA, Molina-Hernández A (2019): The Systemic Administration of the Histamine H1 Receptor Antagonist/Inverse Agonist Chlorpheniramine to Pregnant Rats Impairs the Development of Nigro-Striatal Dopaminergic Neurons. Front Neurosci. 2019 Apr 16;13:360. doi: 10.3389/fnins.2019.00360. PMID: 31040765; PMCID: PMC6476962.

  31. Wright C, Shin JH, Rajpurohit A, Deep-Soboslay A, Collado-Torres L, Brandon NJ, Hyde TM, Kleinman JE, Jaffe AE, Cross AJ, Weinberger DR (2017): Altered expression of histamine signaling genes in autism spectrum disorder. Transl Psychiatry. 2017 May 9;7(5):e1126. doi: 10.1038/tp.2017.87. PMID: 28485729; PMCID: PMC5534955.

  32. Hino N, Marumo T, Kotani M, Shimazaki T, Kaku-Fukumoto A, Hikichi H, Karasawa JI, Tomishima Y, Komiyama H, Tatsuda E, Nozawa D, Nakamura T, Chaki S (2020): A Novel Potent and Selective Histamine H3 Receptor Antagonist Enerisant: In Vitro Profiles, In Vivo Receptor Occupancy, and Wake-Promoting and Procognitive Effects in Rodents. J Pharmacol Exp Ther. 2020 Nov;375(2):276-285. doi: 10.1124/jpet.120.000185. PMID: 32862143.

  33. Abdulrazzaq YM, Bastaki SMA, Adeghate E (2022): Histamine H3 receptor antagonists - Roles in neurological and endocrine diseases and diabetes mellitus. Biomed Pharmacother. 2022 Jun;150:112947. doi: 10.1016/j.biopha.2022.112947. PMID: 35447544.

  34. Krief S, Berrebi-Bertrand I, Nagmar I, Giret M, Belliard S, Perrin D, Uguen M, Robert P, Lecomte JM, Schwartz JC, Finance O, Ligneau X (2021): Pitolisant, a wake-promoting agent devoid of psychostimulant properties: Preclinical comparison with amphetamine, modafinil, and solriamfetol. Pharmacol Res Perspect. 2021 Oct;9(5):e00855. doi: 10.1002/prp2.855. PMID: 34423920; PMCID: PMC8381683.

  35. Naganuma F, Yoshikawa T (2021): Organic Cation Transporters in Brain Histamine Clearance: Physiological and Psychiatric Implications. Handb Exp Pharmacol. 2021;266:169-185. doi: 10.1007/164_2021_447. PMID: 33641029. REVIEW

  36. Mena-Avila E, Márquez-Gómez R, Aquino-Miranda G, Nieto-Alamilla G, Arias-Montaño JA (2018): Clobenpropit, a histamine H3 receptor antagonist/inverse agonist, inhibits [3H]-dopamine uptake by human neuroblastoma SH-SY5Y cells and rat brain synaptosomes. Pharmacol Rep. 2018 Feb;70(1):146-155. doi: 10.1016/j.pharep.2017.08.007. PMID: 29414147.

  37. Esbenshade TA, Browman KE, Miller TR, Krueger KM, Komater-Roderwald V, Zhang M, Fox GB, Rueter L, Robb HM, Radek RJ, Drescher KU, Fey TA, Bitner RS, Marsh K, Polakowski JS, Zhao C, Cowart MD, Hancock AA, Sullivan JP, Brioni JD (2012): Pharmacological properties and procognitive effects of ABT-288, a potent and selective histamine H3 receptor antagonist. J Pharmacol Exp Ther. 2012 Oct;343(1):233-45. doi: 10.1124/jpet.112.194126. PMID: 22815533.

  38. Fox GB, Esbenshade TA, Pan JB, Radek RJ, Krueger KM, Yao BB, Browman KE, Buckley MJ, Ballard ME, Komater VA, Miner H, Zhang M, Faghih R, Rueter LE, Bitner RS, Drescher KU, Wetter J, Marsh K, Lemaire M, Porsolt RD, Bennani YL, Sullivan JP, Cowart MD, Decker MW, Hancock AA (2005): Pharmacological properties of ABT-239 [4-(2-{2-[(2R)-2-Methylpyrrolidinyl]ethyl}-benzofuran-5-yl)benzonitrile]: II. Neurophysiological characterization and broad preclinical efficacy in cognition and schizophrenia of a potent and selective histamine H3 receptor antagonist. J Pharmacol Exp Ther. 2005 Apr;313(1):176-90. doi: 10.1124/jpet.104.078402. PMID: 15608077.

  39. Cowart M, Faghih R, Curtis MP, Gfesser GA, Bennani YL, Black LA, Pan L, Marsh KC, Sullivan JP, Esbenshade TA, Fox GB (2005): Hancock AA. 4-(2-[2-(2(R)-methylpyrrolidin-1-yl)ethyl]benzofuran-5-yl)benzonitrile and related 2-aminoethylbenzofuran H3 receptor antagonists potently enhance cognition and attention. J Med Chem. 2005 Jan 13;48(1):38-55. doi: 10.1021/jm040118g. PMID: 15634000.

  40. Esbenshade TA, Fox GB, Krueger KM, Baranowski JL, Miller TR, Kang CH, Denny LI, Witte DG, Yao BB, Pan JB, Faghih R, Bennani YL, Williams M, Hancock AA (2004): Pharmacological and behavioral properties of A-349821, a selective and potent human histamine H3 receptor antagonist. Biochem Pharmacol. 2004 Sep 1;68(5):933-45. doi: 10.1016/j.bcp.2004.05.048. PMID: 15294456.

  41. Esbenshade TA, Krueger KM, Miller TR, Kang CH, Denny LI, Witte DG, Yao BB, Fox GB, Faghih R, Bennani YL, Williams M, Hancock AA (2003): Two novel and selective nonimidazole histamine H3 receptor antagonists A-304121 and A-317920: I. In vitro pharmacological effects. J Pharmacol Exp Ther. 2003 Jun;305(3):887-96. doi: 10.1124/jpet.102.047183. PMID: 12606603.

  42. Fox GB, Pan JB, Radek RJ, Lewis AM, Bitner RS, Esbenshade TA, Faghih R, Bennani YL, Williams M, Yao BB, Decker MW, Hancock AA (2003): Two novel and selective nonimidazole H3 receptor antagonists A-304121 and A-317920: II. In vivo behavioral and neurophysiological characterization. J Pharmacol Exp Ther. 2003 Jun;305(3):897-908. doi: 10.1124/jpet.102.047241. PMID: 12606600.

  43. Venkatachalam K, Eissa N, Awad MA, Jayaprakash P, Zhong S, Stölting F, Stark H, Sadek B (2021): The histamine H3R and dopamine D2R/D3R antagonist ST-713 ameliorates autism-like behavioral features in BTBR T+tf/J mice by multiple actions. Biomed Pharmacother. 2021 Jun;138:111517. doi: 10.1016/j.biopha.2021.111517. PMID: 33773463.

  44. Eissa N, Awad MA, Thomas SD, Venkatachalam K, Jayaprakash P, Zhong S, Stark H, Sadek B (2022): Simultaneous Antagonism at H3R/D2R/D3R Reduces Autism-like Self-Grooming and Aggressive Behaviors by Mitigating MAPK Activation in Mice. Int J Mol Sci. 2022 Dec 28;24(1):526. doi: 10.3390/ijms24010526. PMID: 36613969; PMCID: PMC9820264.

  45. Eissa N, Venkatachalam K, Jayaprakash P, Falkenstein M, Dubiel M, Frank A, Reiner-Link D, Stark H, Sadek B (2021): The Multi-Targeting Ligand ST-2223 with Histamine H3 Receptor and Dopamine D2/D3 Receptor Antagonist Properties Mitigates Autism-Like Repetitive Behaviors and Brain Oxidative Stress in Mice. Int J Mol Sci. 2021 Feb 16;22(4):1947. doi: 10.3390/ijms22041947. PMID: 33669336; PMCID: PMC7920280.

  46. Eissa N, Venkatachalam K, Jayaprakash P, Yuvaraju P, Falkenstein M, Stark H, Sadek B (2022): Experimental Studies Indicate That ST-2223, the Antagonist of Histamine H3 and Dopamine D2/D3 Receptors, Restores Social Deficits and Neurotransmission Dysregulation in Mouse Model of Autism. Pharmaceuticals (Basel). 2022 Jul 27;15(8):929. doi: 10.3390/ph15080929. PMID: 36015079; PMCID: PMC9414676.

  47. Avila-Luna A, Ríos C, Gálvez-Rosas A, Montes S, Arias-Montaño JA, Bueno-Nava A (2019): Chronic administration of the histamine H3 receptor agonist immepip decreases L-Dopa-induced dyskinesias in 6-hydroxydopamine-lesioned rats. Psychopharmacology (Berl). 2019 Jun;236(6):1937-1948. doi: 10.1007/s00213-019-5182-y. PMID: 30762089.

  48. Zhou P, Homberg JR, Fang Q, Wang J, Li W, Meng X, Shen J, Luan Y, Liao P, Swaab DF, Shan L, Liu C (2019): Histamine-4 receptor antagonist JNJ7777120 inhibits pro-inflammatory microglia and prevents the progression of Parkinson-like pathology and behaviour in a rat model. Brain Behav Immun. 2019 Feb;76:61-73. doi: 10.1016/j.bbi.2018.11.006. PMID: 30408497.

  49. Łażewska D, Kieć-Kononowicz K. Progress in the development of histamine H3 receptor antagonists/inverse agonists: a patent review (2013-2017). Expert Opin Ther Pat. 2018 Mar;28(3):175-196. doi: 10.1080/13543776.2018.1424135. PMID: 29334795.

  50. Blasco Fontecilla H, Wang P, Li C, Duelo A, Ruiz-Casares E, Perucho T (2022): PREVALENCIA Y PERFIL CLÍNICO DE LA DEFICIENCIA DE DIAMINO OXIDASA (DAO) EN PACIENTES CON TRASTORNO POR DÉFICIT DE ATENCIÓN E HIPERACTIVIDAD (TDAH); REVISTA DE PSIQUIATRÍA INFANTO-JUVENIL, Volumen 39, Suplemento 1, (2022), Page 104.

  51. Manz KM, Becker JC, Grueter CA, Grueter BA (2020): Histamine H3 Receptor Function Biases Excitatory Gain in the Nucleus Accumbens. Biol Psychiatry. 2021 Mar 15;89(6):588-599. doi: 10.1016/j.biopsych.2020.07.023. PMID: 33012522; PMCID: PMC7865000.

  52. Higuchi, Yanai, Okamura, Meguro, Arai, Itoh, Iwata, Ido, Watanabe, Sasaki (2000): Histamine H1 receptors in patients with Alzheimer’s disease assessed by positron emission tomography, Neuroscience, Volume 99, Issue 4, 2000, Pages 721-729, ISSN 0306-4522, https://doi.org/10.1016/S0306-4522(00)00230-X.

  53. Ercan-Sencicek, Stillman, Ghosh, Bilguvar, O’Roak, Mason, Abbott, Gupta, King, Pauls, Tischfield, Heiman, Singer, Gilbert, Hoekstra, Morgan, Loring, Yasuno, Fernandez, Sanders, Louvi, Cho, Mane, Colangelo, Biederer, Lifton, Gunel, State (2010): L-histidine decarboxylase and Tourette’s syndrome. N Engl J Med. 2010 May 20;362(20):1901-8. doi: 10.1056/NEJMoa0907006. . PMID: 20445167; PMCID: PMC2894694.

  54. Karagiannidis, Dehning, Sandor, Tarnok, Rizzo, Wolanczyk, Madruga-Garrido, Hebebrand, Nöthen, Lehmkuhl, Farkas, Nagy, Szymanska, Anastasiou, Stathias, Androutsos, Tsironi, Koumoula, Barta, Zill, Mir, Müller, Barr, Paschou (2013): Support of the histaminergic hypothesis in Tourette syndrome: association of the histamine decarboxylase gene in a large sample of families. J Med Genet. 2013 Nov;50(11):760-4. doi: 10.1136/jmedgenet-2013-101637. PMID: 23825391.

  55. Rapanelli, Frick, Bito, Pittenger (2017): Histamine modulation of the basal ganglia circuitry in the development of pathological grooming. Proc Natl Acad Sci U S A. 2017;114(25):6599-6604. doi:10.1073/pnas.1704547114

  56. Fernandez, Sanders, Yurkiewicz, Ercan-Sencicek, Kim, Fishman, Raubeson, Song, Yasuno, Ho, Bilguvar, Glessner, Chu, Leckman, King, Gilbert, Heiman, Tischfield, Hoekstra, Devlin, Hakonarson, Mane, Günel, State (2012): Rare Copy Number Variants in Tourette Syndrome Disrupt Genes in Histaminergic Pathways and Overlap with Autism, Biological Psychiatry, Volume 71, Issue 5, 2012, Pages 392-402, ISSN 0006-3223, https://doi.org/10.1016/j.biopsych.2011.09.034

  57. Wright, Shin, Rajpurohit, Deep-Soboslay, Collado-Torres, Brandon, Hyde, Kleinman, Jaffe, Cross, Weinberger (2017): Altered expression of histamine signaling genes in autism spectrum disorder. Transl Psychiatry. 2017 May 9;7(5):e1126. doi: 10.1038/tp.2017.87. PMID: 28485729; PMCID: PMC5534955.

  58. https://www.histaminintoleranz.ch/

  59. Bertl, Haririan, Laky, Matejka, Andrukhov, Rausch-Fan (2012): Smoking influences salivary histamine levels in periodontal disease. Oral Dis. 2012 May;18(4):410-6. doi: 10.1111/j.1601-0825.2011.01891.x.

  60. Wilhelm (2017): Anmerkung zu Maintz, Bieber, Novak, in Heft 51–52/2006: Die verschiedenen Gesichter der Histaminintoleranz – Konsequenzen für die Praxis: Tabakrauch ist bedeutende Histaminquelle. Dtsch Arztebl 2007; 104(24): A-1758 / B-1553 / C-1492

  61. Du (1991): [The effects of passive smoking on health].[Article in Chinese] Zhonghua Jie He He Hu Xi Za Zhi. 1991 Apr;14(2):76-8, 126.

  62. Kalenderian, Raju, Roth, Schwartz, Gruber, Janoff (1988): Elevated histamine and tryptase levels in smokers’ bronchoalveolar lavage fluid. Do lung mast cells contribute to smokers’ emphysema? Chest. 1988 Jul;94(1):119-23.

  63. Walter, Walter (1982): Mast cell density in isolated monkey lungs on exposure to cigarette smoke. Thorax. 1982 Sep;37(9):699-702.

  64. Gertner, Bromberger, Traystman, Menkes (1982): Histamine and pulmonary responses to cigarette smoke in periphery of the lung. J Appl Physiol Respir Environ Exerc Physiol. 1982 Sep;53(3):582-8.

  65. Della Rovere, Granata, Familiari, Zirilli, Cimino, Tomaino (2006): Histamine and selenium in lung cancer. Anticancer Res. 2006 Jul-Aug;26(4B):2937-42.

  66. Lewis, Nicholls (1972) Effect of inhaled cigarette smoke on content, release and breakdown of histamine in guinea-pig lung. Life Sci II. 1972 Dec 8;11(23):1167-71.

  67. Daffonchio, Hernandez, Gallico, Omini (1990): Airway hyperreactivity induced by active cigarette smoke exposure in guinea-pigs: possible role of sensory neuropeptides. Pulm Pharmacol. 1990;3(3):161-6.

  68. Keller, Doyle (1976): A mechanism for tobacco smoke-induced allergy. J Allergy Clin Immunol. 1976 Mar;57(3):278-82.

  69. Rijcken, Schouten, Mensinga, Weiss, De Vries, Van der Lende (1993): Factors associated with bronchial responsiveness to histamine in a population sample of adults. Am Rev Respir Dis. 1993 Jun;147(6 Pt 1):1447-53.

  70. Turner, Palmer, Rye, Gibson, Young, Goldblatt, Landau, Le Souëf (2005): Determinants of airway responsiveness to histamine in children. Eur Respir J. 2005 Mar;25(3):462-7.

  71. https://www.mastzellaktivierung.info/de/einleitung_kurzfassung.html#zusammenfassung

  72. https://www.mastzellaktivierung.info/

  73. Valent, Akin, Arock, Brockow, Butterfield, Carter, Castells, Escribano, Hartmann, Lieberman, Nedoszytko, Orfao, Schwartz, Sotlar, Sperr, Triggiani, Valenta, Horny, Metcalfe (2012): Definitions, criteria and global classification of mast cell disorders with special reference to mast cell activation syndromes: a consensus proposal. Int Arch Allergy Immunol. 2012;157(3):215-25. doi: 10.1159/000328760.

  74. Garza, Liu, Yang, Alagesan, Lawson, Norberg, Loy, Zhao, Blatt, Stanton, Carrasco, Ahluwalia. Fischer. FitzGerald, Cotsarelis (2012): Prostaglandin D2 Inhibits Hair Growth and Is Elevated in Bald Scalp of Men with Androgenetic Alopecia. Science Translational Medicine 21 Mar 2012: Vol. 4, Issue 126, pp. 126ra34. DOI: 10.1126/scitranslmed.3003122

  75. Mastozytose: Krankheit mit vielen Gesichtern. Pharmazeutische Zeitung Ausgabe 33/2014

  76. https://www.mastzellaktivierung.info/de/therapie_ausloesermeiden.html

  77. Quade, Bailly, Bartling, Bliesener, Springer: Histamin-Unverträglichkeit

  78. Abruf 17.03.2022

  79. Fuhrmann, Tesch, Romanos, Abraham, Schmitt (2020): ADHD in school-age children is related to infant exposure to systemic H1-antihistamines. Allergy. 2020 Nov;75(11):2956-2957. doi: 10.1111/all.14411. PMID: 32441335. n = 41.484

  80. Schmitt, Buske-Kirschbaum, Tesch, Trikojat, Stephan, Abraham, Bauer, Nemat, Plessow, Roessner (2018): Increased attention-deficit/hyperactivity symptoms in atopic dermatitis are associated with history of antihistamine use. Allergy. 2018 Mar;73(3):615-626. doi: 10.1111/all.13326.

  81. Stevenson J, Sonuga-Barke E, McCann D, Grimshaw K, Parker KM, Rose-Zerilli MJ, Holloway JW, Warner JO (2010): The role of histamine degradation gene polymorphisms in moderating the effects of food additives on children’s ADHD symptoms. Am J Psychiatry. 2010 Sep;167(9):1108-15. doi: 10.1176/appi.ajp.2010.09101529. PMID: 20551163. RCT

  82. Levinson (1991): Dramatic favorable responses of children with learning disabilities or dyslexia and attention deficit disorder to antimotion sickness medications: four case reports. Percept Mot Skills. 1991 Dec;73(3 Pt 1):723-38. doi: 10.2466/pms.1991.73.3.723. PMID: 1686492.

  83. Nazarova VA, Sokolov AV, Chubarev VN, Tarasov VV, Schiöth HB (2022): Treatment of ADHD: Drugs, psychological therapies, devices, complementary and alternative methods as well as the trends in clinical trials. Front Pharmacol. 2022 Nov 17;13:1066988. doi: 10.3389/fphar.2022.1066988. PMID: 36467081; PMCID: PMC9713849.

  84. Pang YP, Zheng XE, Weinshilboum RM (2001): Theoretical 3D model of histamine N-methyltransferase: insights into the effects of a genetic polymorphism on enzymatic activity and thermal stability. Biochem Biophys Res Commun. 2001 Sep 14;287(1):204-8. doi: 10.1006/bbrc.2001.5570. PMID: 11549275.

  85. Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, Agúndez JAG (2016): Thr105Ile (rs11558538) polymorphism in the histamine N-methyltransferase (HNMT) gene and risk for Parkinson disease: A PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore). 2016 Jul;95(27):e4147. doi: 10.1097/MD.0000000000004147. PMID: 27399132; PMCID: PMC5058861.

  86. Heidari A, Tongsook C, Najafipour R, Musante L, Vasli N, Garshasbi M, Hu H, Mittal K, McNaughton AJ, Sritharan K, Hudson M, Stehr H, Talebi S, Moradi M, Darvish H, Arshad Rafiq M, Mozhdehipanah H, Rashidinejad A, Samiei S, Ghadami M, Windpassinger C, Gillessen-Kaesbach G, Tzschach A, Ahmed I, Mikhailov A, Stavropoulos DJ, Carter MT, Keshavarz S, Ayub M, Najmabadi H, Liu X, Ropers HH, Macheroux P, Vincent JB (2015): Mutations in the histamine N-methyltransferase gene, HNMT, are associated with nonsyndromic autosomal recessive intellectual disability. Hum Mol Genet. 2015 Oct 15;24(20):5697-710. doi: 10.1093/hmg/ddv286. PMID: 26206890; PMCID: PMC4581600.

  87. Kim SH, Kang YM, Kim SH, Cho BY, Ye YM, Hur GY, Park HS (2009): Histamine N-methyltransferase 939A>G polymorphism affects mRNA stability in patients with acetylsalicylic acid-intolerant chronic urticaria. Allergy. 2009 Feb;64(2):213-21. doi: 10.1111/j.1398-9995.2008.01795.x. PMID: 19178400.

  88. Xu J, Pittenger C (2023): The histamine H3 receptor modulates dopamine D2 receptor-dependent signaling pathways and mouse behaviors. J Biol Chem. 2023 Apr;299(4):104583. doi: 10.1016/j.jbc.2023.104583. PMID: 36871761; PMCID: PMC10139999.

  89. Zhuang QX, Xu HT, Lu XJ, Li B, Yung WH, Wang JJ, Zhu JN (2018): Histamine Excites Striatal Dopamine D1 and D2 Receptor-Expressing Neurons via Postsynaptic H1 and H2 Receptors. Mol Neurobiol. 2018 Oct;55(10):8059-8070. doi: 10.1007/s12035-018-0976-1. PMID: 29498008.

  90. Koski SK, Leino S, Panula P, Rannanpää S, Salminen O (2020): Genetic lack of histamine upregulates dopamine neurotransmission and alters rotational behavior but not levodopa-induced dyskinesia in a mouse model of Parkinson’s disease. Neurosci Lett. 2020 Jun 11;729:134932. doi: 10.1016/j.neulet.2020.134932. PMID: 32224226.

  91. Peng JY, Qi ZX, Yan Q, Fan XJ, Shen KL, Huang HW, Lu JH, Wang XQ, Fang XX, Mao L, Ni J, Chen L, Zhuang QX (2023): Ameliorating parkinsonian motor dysfunction by targeting histamine receptors in entopeduncular nucleus-thalamus circuitry. Proc Natl Acad Sci U S A. 2023 Apr 25;120(17):e2216247120. doi: 10.1073/pnas.2216247120. PMID: 37068253; PMCID: PMC10151461.

  92. Vargas-Romero F, González-Barrios R, Guerra-Calderas L, Escobedo-Avila I, Cortés-Pérez D, López-Ornelas A, Rocha L, Soto-Reyes E, Velasco I (2019): Histamine Modulates Midbrain Dopamine Neuron Differentiation Through the Regulation of Epigenetic Marks. Front Cell Neurosci. 2019 May 21;13:215. doi: 10.3389/fncel.2019.00215. PMID: 31178697; PMCID: PMC6536891.

  93. Varaschin RK, Osterstock G, Ducrot C, Leino S, Bourque MJ, Prado MAM, Prado VF, Salminen O, Rannanpää Née Nuutinen S, Trudeau LE (2018): Histamine H3 Receptors Decrease Dopamine Release in the Ventral Striatum by Reducing the Activity of Striatal Cholinergic Interneurons. Neuroscience. 2018 Apr 15;376:188-203. doi: 10.1016/j.neuroscience.2018.01.027. PMID: 29374538.

  94. Abdurakhmanova S, Semenova S, Piepponen TP, Panula P (2019): Abnormal behavior, striatal dopamine turnover and opioid peptide gene expression in histamine-deficient mice. Genes Brain Behav. 2019 Nov;18(8):e12595. doi: 10.1111/gbb.12595. PMID: 31216095.

  95. Moreno-Delgado D, Puigdellívol M, Moreno E, Rodríguez-Ruiz M, Botta J, Gasperini P, Chiarlone A, Howell LA, Scarselli M, Casadó V, Cortés A, Ferré S, Guzmán M, Lluís C, Alberch J, Canela EI, Ginés S, McCormick PJ (2020): Modulation of dopamine D1 receptors via histamine H3 receptors is a novel therapeutic target for Huntington’s disease. Elife. 2020 Jun 9;9:e51093. doi: 10.7554/eLife.51093. PMID: 32513388; PMCID: PMC7282811.

  96. Fox GB, Pan JB, Esbenshade TA, Bennani YL, Black LA, Faghih R, Hancock AA, Decker MW (2002): Effects of histamine H(3) receptor ligands GT-2331 and ciproxifan in a repeated acquisition avoidance response in the spontaneously hypertensive rat pup. Behav Brain Res. 2002 Apr 1;131(1-2):151-61. doi: 10.1016/s0166-4328(01)00379-5. PMID: 11844582.

  97. Rapanelli M (2017): The magnificent two: histamine and the H3 receptor as key modulators of striatal circuitry. Prog Neuropsychopharmacol Biol Psychiatry. 2017 Feb 6;73:36-40. doi: 10.1016/j.pnpbp.2016.10.002. PMID: 27773554. REVIEW

  98. Kitanaka N, Hall FS, Kobori S, Kushihara S, Oyama H, Sasaoka Y, Takechi M, Tanaka KI, Tomita K, Igarashi K, Nishiyama N, Sato T, Uhl GR, Kitanaka J (2021): Metoprine, a histamine N-methyltransferase inhibitor, attenuates methamphetamine-induced hyperlocomotion via activation of histaminergic neurotransmission in mice. Pharmacol Biochem Behav. 2021 Oct;209:173257. doi: 10.1016/j.pbb.2021.173257. PMID: 34418452.

  99. Puttonen HAJ, Sundvik M, Semenova S, Shirai Y, Chen YC, Panula P (2018): Knockout of histamine receptor H3 alters adaptation to sudden darkness and monoamine levels in the zebrafish. Acta Physiol (Oxf). 2018 Mar;222(3). doi: 10.1111/apha.12981. PMID: 29044927.

  100. Lee, Goto (2013): Habenula and ADHD: Convergence on time. Neuroscience and biobehavioral reviews (2013), 37(8): 1801-1809

  101. Kim, Goto, Lee (2018): Histamine H3 receptor antagonists ameliorate attention deficit/hyperactivity disorder-like behavioral changes caused by neonatal habenula lesion. Behav Pharmacol. 2018 Feb;29(1):71-78. doi: 10.1097/FBP.0000000000000343.

  102. Prast H, Gujrati V, Walser S, Philippu A (1988): Histamine, histidine decarboxylase and histamine-N-methyltransferase in brain areas of spontaneously hypertensive rats. Naunyn Schmiedebergs Arch Pharmacol. 1988 Nov;338(5):573-6. doi: 10.1007/BF00179332. PMID: 3244396.

  103. Oishi R, Itoh Y, Nishibori M, Saeki K (1985): Decrease in histamine turnover in the brain of spontaneously hypertensive rats. Brain Res. 1985 Sep 16;343(1):180-3. doi: 10.1016/0006-8993(85)91175-8. PMID: 4041852.

  104. Toba H, Nakamori A, Tanaka Y, Yukiya R, Tatsuoka K, Narutaki M, Tokitaka M, Hariu H, Kobara M, Nakata T (2010): Oral L-histidine exerts antihypertensive effects via central histamine H3 receptors and decreases nitric oxide content in the rostral ventrolateral medulla in spontaneously hypertensive rats. Clin Exp Pharmacol Physiol. 2010 Jan;37(1):62-8. doi: 10.1111/j.1440-1681.2009.05227.x. PMID: 19566844.

  105. Masini E, Mannaioni PF, Pistelli A, Salvemini D, Vane J (1991): Impairment of the L-arginine-nitric oxide pathway in mast cells from spontaneously hypertensive rats. Biochem Biophys Res Commun. 1991 Jun 28;177(3):1178-82. doi: 10.1016/0006-291x(91)90664-s. PMID: 1711845.

  106. Amon EU, Ennis M, Lorenz W, Schnabel M, Schneider C (1990): Histamine release induced by radiographic contrast media. Comparison between pulmonary and peritoneal mast cells derived from normotensive and spontaneously hypertensive rats. Int Arch Allergy Appl Immunol. 1990;92(2):203-8. PMID: 1700769.

  107. Shaw JB, Cai Q, Mtshali C, Myles EL, Washington B (2007): Heterogeneity of histamine H3 receptor genomic expression in the cerebral cortex of spontaneously hypertensive rat. Cell Mol Biol (Noisy-le-grand). 2007 May 15;53(4):45-50. PMID: 17531160.

  108. Prast H, Philippu A (1991): Does brain histamine contribute to the development of hypertension in spontaneously hypertensive rats? Naunyn Schmiedebergs Arch Pharmacol. 1991 Mar;343(3):307-10. doi: 10.1007/BF00251131. PMID: 1865929.

  109. Domer FR, Boertje SB, Bing EG, Reddix I (1983): Histamine- and acetylcholine-induced changes in the permeability of the blood-brain barrier of normotensive and spontaneously hypertensive rats. Neuropharmacology. 1983 May;22(5):615-9. doi: 10.1016/0028-3908(83)90153-3. PMID: 6308494.

Diese Seite wurde am 26.02.2024 zuletzt aktualisiert.