Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2015 Jan 12;172(5):1165–1178. doi: 10.1111/bph.12989

Histamine H4 receptors in the gastrointestinal tract

A Deiteren 1, J G De Man 1, P A Pelckmans 1,2, B Y De Winter 1,
PMCID: PMC4337694  PMID: 25363289

Abstract

Histamine is a well-established mediator involved in a variety of physiological and pathophysiological mechanisms and exerts its effect through activation of four histamine receptors (H1–H4). The histamine H4 receptor is the newest member of this histamine receptor family, and is expressed throughout the gastrointestinal tract as well as in the liver, pancreas and bile ducts. Functional studies using a combination of selective and non-selective H4 receptor ligands have rapidly increased our knowledge of H4 receptor involvement in gastrointestinal processes both under physiological conditions and in models of disease. Strong evidence points towards a role for H4 receptors in the modulation of immune-mediated responses in gut inflammation such as in colitis, ischaemia/reperfusion injury, radiation-induced enteropathy and allergic gut reactions. In addition, data have emerged implicating H4 receptors in gastrointestinal cancerogenesis, sensory signalling, and visceral pain as well as in gastric ulceration. These studies highlight the potential of H4 receptor targeted therapy in the treatment of various gastrointestinal disorders such as inflammatory bowel disease, irritable bowel syndrome and cancer.

Tables of Links

TARGETS
GPCRsa
Histamine H1 receptor
Histamine H2 receptor
Histamine H3 receptor
Histamine H4 receptor
Enzymesb
COX-2
LIGANDS
4-Methylhistamine Immepip
Cimetidine JNJ10191584
Clobenpropit JNJ7777120
Clozapine Ketotifen
Dimaprit Pyrilamine
Histamine Thioperamide
Imetit VUF8430

These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,bAlexander et al., 2013a,b).

Introduction

Histamine (2-[4-imidazole]-ethylamine) is a short-acting endogenous amine, involved in several physiological and pathophysiological processes (Jutel et al., 2009). It is present in virtually all bodily organs, with high concentrations reported in the stomach, lymph nodes and thymus (Kumar et al., 1968; Zimmermann et al., 2011). Histamine is synthetized from L-histidine by L-histidine decarboxylase and is stored in the granules of mast cells (MCs) and basophils, the main sources of histamine (Endo, 1982; Jones and Kearns, 2011). Enterochromaffin-like cells, histaminergic neurons, lymphocytes, monocytes, platelets and neutrophils also express L-histidine decarboxylase and are capable of producing, but not storing, high amounts of histamine (Snyder and Epps, 1968; Vanhala et al., 1994; Bencsath et al., 1998; Jutel et al., 2009; Alcaniz et al., 2013). Histamine exerts its actions by binding to four GPCRs that are differentially expressed throughout the body and designated as the H1, H2, H3 and H4 receptors. Histamine H1 receptors mediate sensorineural signalling, vascular dilatation and permeability and airway smooth muscle contraction, and are involved in allergic rhinitis, atopic dermatitis, conjunctivitis, urticaria, asthma and anaphylaxis (Togias, 2003; Simons and Simons, 2011). Histamine H2 receptors are well-known for their role in gastric acid secretion, but also exert immune modulatory properties (Black et al., 1972; Jutel et al., 2009). Histamine H3 receptors are most abundantly present in the CNS and are implicated in sleep–wake disorders, attention-deficient hyperactivity disorder, epilepsy, cognitive impairment and obesity (Kuhne et al., 2011; Singh and Jadhav, 2013). Finally, histamine H4 receptors are predominantly expressed on immune cells, such as lymphocytes, MCs and dendritic cells, and are currently mainly under evaluation for immune-mediated disorders such as allergic rhinitis, asthma and pruritus (Liu, 2014). However, new roles for this receptor subtype are continuously being discovered. Here we provide an overview of the current evidence of H4 receptor involvement in multiple gastrointestinal physiological and pathophysiological processes.

H4 receptors

In the early 2000s, several groups reported on the discovery and cloning of a fourth histamine receptor (Nakamura et al., 2000; Oda et al., 2000; Liu et al., 2001a; Morse et al., 2001; Nguyen et al., 2001; Zhu et al., 2001). The H4 receptor is encoded by a single copy on chromosome 18q11.2 and demonstrates an overall homology of 23% to H1 receptors, 22% to H2 receptors and 37% to H3 receptors (Oda et al., 2000; Coge et al., 2001). The human full-length receptor consists of 390 amino acids, which form seven transmembrane helices, three extracellular loops and three intracellular loops, with an extracellular N-terminal and an intracellular C-terminal peptide (Leurs et al., 2009). H4 receptors couple to Gαi/0 proteins, inhibiting downstream adenylyl cyclase and forskolin-induced cAMP (Morse et al., 2001; Zhu et al., 2001). They are mainly present in immune cells and highly expressed in bone marrow and spleen; varying expression levels were also reported in gastrointestinal tissues, testes, kidney, lung, prostrate and brain (Nakamura et al., 2000; Oda et al., 2000; Coge et al., 2001; Strakhova et al., 2009). Tissue distribution is quite similar across species (Liu et al., 2001b; Oda et al., 2005). There is high homology in the amino acid sequence between human and monkey H4 receptors (92%), whereas this is 72% between human and pig and 65–70% between human and rodent H4 receptors (Liu et al., 2001b; Oda et al., 2002; 2005,). These differences in amino acid sequence also affect the binding profile of histamine towards H4 receptors with high affinity for human and guinea pig H4 receptors (KD 4.8 and 6 nM) compared with rat and mouse H4 receptors (136 and 42 nM) (Liu et al., 2001b). Compared with H1 and H2 receptors, histamine displays high affinity for H4 receptors in both human and rodents (Table 2006).

Table 1.

Ligands for the human H4 receptor

Compound H4R (pKi) H1R (pKi) H2R (pKi) H3R (pKi)
Agonists
 Histamine 7.8 4.2 4.3 8.0
 4-methylhistamine 7.3 <5.0 5.1 5.2
 Clozapine 6.7 9.4 6.6
 Clobenpropit 8.1 8.6
 Dimaprit 6.5 4.6 7.3
 Imetit 8.2 8.8
 Immepip 7.7 9.3
 OUP-16 6.9 5.7
VUF10460 8.2 5.8
 VUF6884 7.6 5.0
 VUF8430 7.5 6.0
Antagonists
 A-943931 8.3
 JNJ10191584* 7.6
 JNJ39758979 7.9 <6 <6 <6
 JNJ7777120 7.8 <5.0 <5.0 5.3
 ZPL3893787 8.6 6.7
 Thioperamide 6.9 7.3
 UR-63325 7.8 <5.4 <5.4 <5.4
*

Former VUF6002. Based on Andaloussi et al. (2013); Alfon et al. (2011); Coruzzi et al. (2007; 2011,); Cowart et al. (2008); Leurs et al. (2009); Lim et al. (2009); Mowbray et al. (2011); Oda et al. (2000); Salcedo et al. (2013); Smits et al. (2009); Thurmond et al. (2004; 2014). Of note, ligand affinity may differ among species. Data presented as Ki value (nM) for the human histamine H4 and H3 receptors.

Soon after its discovery and cloning, attempts were made to elucidate the pharmacological profile of H4 receptors and identify (selective) ligands to stimulate or inhibit H4 receptor signalling. Early assessments indicated that several H3 receptor ligands demonstrated significant affinity for H4 receptors, such as clozapine, imetit and immepip (H3 and H4 receptor agonists) and clobenpropit (H3 receptor antagonist, H4 receptor agonist) (Table 2006) (Leurs et al., 2009; Smits et al., 2009). Since then, several new compounds have been developed targeting H4 receptors such as 4-methylhistamine, VUF8430 and OUP-16 (selective agonists) and A-943931, JNJ7777120 and VUF6002 (selective antagonists; Table 2006) (Leurs et al., 2009; Smits et al., 2009). However, it was recently demonstrated that in addition to inhibition of Gαi/0 proteins, many H4 receptor antagonists can also exert a partial agonist effect at certain species H4 receptor orthologues via β-arrestin recruitment and ERK activation (Rosethorne and Charlton, 2011), which may contribute to some of the species differences that have been reported for H4 receptor ligands (Liu et al., 2001b; Seifert et al., 2011; Nijmeijer et al., 2013; Salcedo et al., 2013).

Histamine and histamine receptors in the gastrointestinal tract

In the gastrointestinal tract, histamine participates in multiple physiological processes among which immunological responses, visceral nociception, modulation of intestinal motility and gastric acid secretion (Black et al., 1972; Poli et al., 2001; Dawicki and Marshall, 2007; Takagaki et al., 2009; Simon et al., 2011; van Diest et al., 2012). Histamine is also involved in several gastrointestinal disorders such as inflammatory bowel diseases (IBD), irritable bowel syndrome (IBS), malignancies, systemic mastocytosis, food allergy and gastric ulcers (Black et al., 1972; He, 2004; Wood, 2004; Barbara et al., 2006; Sokol et al., 2010; Kennedy et al., 2012). All four histamine receptors are expressed in the gastrointestinal tract, although the presence of H3 receptors in the human gut remains controversial (Poli et al., 2001). Human H1 receptors are abundantly expressed throughout the gastrointestinal tract on enterocytes as well as connective tissue cells, immune cells, blood vessels, myocytes and enteric nerves (Sander et al., 2006). H2 receptors are present on gastric parietal cells, enterocytes, immunocytes such as lymphocytes, myenteric ganglia and smooth muscle cells (Fukushima et al., 1999; Sander et al., 2006). H3 receptors were reported to be expressed in gastrointestinal tissue of guinea pig and functional data located them on nerve terminals in the myenteric plexus and on pre- and post-ganglionic cholinergic and non-adrenergic, non-cholinergic fibres (Poli et al., 2001). However, human intestine seems to be devoid of H3 receptors (Hemedah et al., 2001; Poli et al., 2001; Cianchi et al., 2005; Sander et al., 2006).

Using a variety of techniques, several groups demonstrated expression of H4 receptors throughout the gastrointestinal tract and in the pancreas, liver and bile ducts, not only in humans, but also in other species such as rodents, pigs, dogs and monkeys (Table 2005). Sander et al. (2006) reported similar distribution of H4 receptors along the human duodenum, colon, sigmoid and rectum. More specifically, H4 receptors were present on lamina propria mononuclear cells and intestinal MCs, on leucocytes in mucosal and submucosal blood vessels and to a lesser extent on tissue resident leucocytes. In addition, H4 receptor immunoreactivity was seen in intraepithelial cells considered to be neuroendocrine cells, in myenteric ganglion cell somata and neuronal fibres, and on enterocytes in the crypt of Lieberkühn (Sander et al., 2006; Chazot et al., 2007). Expression of H4 receptors on colonic enterocytes was later confirmed by others, who also reported limited staining of non-specified submucosal and connective tissue cells (Boer et al., 2008; Fang et al., 2011). A caveat must be made when interpreting data obtained by immunohistochemistry. Recently the selectivity of commercially available H4 receptor-antibodies was questioned as several of these antibodies failed to yield a specific signal when evaluated in transfected or H4 receptor−/− cells (Beermann et al., 2012).

Table 2.

Expression of H4 receptors in the gastrointestinal tract of different species

Tissue and species Technique Expression profile Reference
Oesophagus
 Guinea pig Immunofluorescence MCs and eosinophils Yu et al. (2008)
Stomach
 Human RNase protection assay Liu et al. (2001a)
Northern blot Morse et al. (2001)
RT-PCR Mucosa Zhang et al. (2012)
Western blot Mucosa Zhang et al. (2012)
Immunofluorescence Mucosal cells Zhang et al. (2012)
 Rat Immunohistochemistry Ganglion cell somata and neuronal fibres in the myenteric but not the submucous plexus; A-like cells in the fundic epithelium Chazot et al. (2007); Morini et al. (2008)
Duodenum
 Human RT-PCR Sander et al. (2006)
Small intestine
 Human RNase protection assay Liu et al. (2001a)
Northern blot Morse et al. (2001)
RT-PCR Coge et al. (2001); Nakamura et al. (2000); Oda et al. (2000)
 Dog RT-PCR Jiang et al. (2008)
 Rat Immunohistochemistry Ganglion cell somata and neuronal fibres in the myenteric plexus Chazot et al. (2007)
Colon
 Human RNase protection assay Liu et al. (2001a)
Northern blot Morse et al. (2001)
RT-PCR Lamina propria mononuclear cells and MCs, mucosa Boer et al. (2008); Cianchi et al. (2005); Fang et al. (2011); Oda et al. (2000); Sander et al. (2006)
Western blot Mucosa Boer et al. (2008); Fang et al. (2011)
Immunohistochemistry Neuroendocrine-like cells, lamina propria, intravascular granulocytes, enterocytes, non-epithelial mucosal cells, submucosal connective tissue cells Boer et al. (2008); Fang et al. (2011); Sander et al. (2006)
 Dog RT-PCR Eisenschenk et al. (2011)
 Rat RT-PCR Deiteren et al. (2014)
Immunohistochemistry Ganglion cell somata and neuronal fibres in the myenteric, but not the submucous plexus Chazot et al. (2007)
 Mouse RT-PCR Sutton et al. (2008)
 Monkey RT-PCR Longitudinal muscle Kim et al. (2011); Oda et al. (2005)
 Pig RT-PCR Oda et al. (2002)
Pancreas
 Human Northern blot Morse et al. (2001)
Liver
 Human RNase protection assay Liu et al. (2001a)
Northern blot Morse et al. (2001)
RT-PCR Coge et al. (2001); Nakamura et al. (2000)
 Dog RT-PCR Eisenschenk et al. (2011); Jiang et al. (2008)
Bile ducts
 Human RT-PCR Francis et al. (2012)
Western blot Francis et al. (2012)
Immunohistochemistry Cholangiocytes Francis et al. (2012); Meng et al. (2011)

A caveat must be made when interpreting H4 receptor expression data obtained by immunohistochemistry: recently the selectivity of commercially available antibodies for the H4 receptor was questioned as several of these antibodies failed to yield a specific signal when evaluated in transfected or H4 receptor −/− cells (Beermann et al., 2012).

Interestingly, gastrointestinal H4 receptor expression is altered in several disease states. Decreased H4 receptor expression was reported in gastric cancer specimens, whereas overexpression was demonstrated in cholangiocarcinoma and both enhanced and decreased expression levels have been reported in colorectal cancer (Cianchi et al., 2005; Boer et al., 2008; Fang et al., 2011; Meng et al., 2011; Francis et al., 2012; Zhang et al., 2012). Colonic inflammation seems to enhance H4 receptor expression as in two experimental models of IBD, namely murine trinitrobenzene sulphonic acid (TNBS)-induced colitis and spontaneous colitis in Giα2 protein-deficient mice, active inflammation was associated with an increase in colonic H4 receptor mRNA (Sutton et al., 2008; Kumawat et al., 2010). Also after complete resolution of TNBS-colitis, colonic H4 receptor mRNA levels remained increased (Deiteren et al., 2014). However, in colonic biopsies of IBS patients with concomitant food allergy, no alterations in H4 receptor mRNA levels were reported (Sander et al., 2006).

H4 receptors and gastrointestinal inflammation

Because of the high levels of expression of H4 receptors on immunocytes, the immune modulatory potential of this receptor subtype attracted much attention, culminating in clinical trials with H4 receptor antagonists in immune-mediated disorders such as asthma and allergic rhinitis. Also in the gastrointestinal tract, their immune modulatory properties have been studied using models of colitis, ischaemia/reperfusion injury and allergic gut reactions (Table 2012).

Table 3.

Preclinical in vivo experiments with H4 receptor ligands in models of inflammation

Model Species In vitro/in vivo Ligand Effect Ref
TNBS-induced colitis Rat In vivo JNJ7777120 JNJ7777120 and JNJ10191584 reduced TNBS-induced colitis Varga et al. (2005)
JNJ10191584
TNBS-induced colitis Rat In vivo JNJ10191584 JNJ10191584 reduced TNBS-induced colitis Dunford et al. (2006b)
TNBS-induced colitis Rat In vivo Thioperamide Thioperamide reduced TNBS-induced colitis Fogel et al. (2007)
Acetic acid-induced colitis Rat In vivo Thioperamide Thioperamide reduced acetic acid-induced colitis Fogel et al. (2005)
Ischaemia/reperfusion intestinal injury Mouse In vivo Thioperamide Thioperamide reduced reperfusion injury Ghizzardi et al. (2009)
Ischaemia/reperfusion liver injury Rat In vivo Dimaprit Histamine, dimaprit and clozapine reduced liver injury Adachi et al. (2006)
Clozapine
Thioperamide Thioperamide reversed the protective effect of histamine and dimaprit
Ischaemia/reperfusion liver injury Rat In vivo Clozapine Histamine and clozapine prevented reperfusion injury El-Mahdy et al. (2013)
Thioperamide Thioperamide reversed the protective effect of histamine
Radiation-induced small intestinal damage Rat In vivo JNJ7777120 JNJ7777120 reduced radiation-induced intestinal damage Martinel Lamas et al. (2013)
Allergen challenge in sensitized oesophagus Guinea pig In vivo Thioperamide Thioperamide inhibited MC and eosinophil migration Yu et al. (2008)

Clozapine, H3 and H4 receptor agonist; dimaprit, H2 and H4 receptor agonist; JNJ10191584, H4 receptor antagonist; JNJ7777120, H4 receptor antagonist; thioperamide, H3 and H4 receptor antagonist.

MCs, an important source of gastrointestinal histamine, are key players of both the innate and adaptive immune systems and congregate at the interface between the internal and external milieu (such as the gut mucosa), where they exert immune modulatory effects. Alterations in MC numbers and activation state with excessive release of histamine have been reported in patients with IBD (Knutson et al., 1990; Bischoff et al., 1996; Farhadi et al., 2007). Moreover, treatment with the MC stabilizer ketotifen prevented chemically induced colitis in animal models and improved disease activity in a small group of IBD patients; however, the underlying mechanism of action was not investigated further (Eliakim et al., 1992; Jones et al., 1998; Marshall and Irvine, 1998; Fogel et al., 2005). Ketotifen stabilizes MCs (in addition to H1 receptor antagonist properties), and thus inhibits the release of histamine in the gut; this may indirectly beneficially affect H4 receptor-mediated pathways activated by histamine.

The selective H4 receptor antagonists JNJ7777120 and JNJ10191584 and the H3/H4 receptor antagonist thioperamide also reduced chemically induced colitis in different rat models for IBD (Fogel et al., 2005; 2007,; Varga et al., 2005; Dunford et al., 2006b). More specifically, treatment with these antagonists reduced macroscopic colonic injury, neutrophil influx and myeloperoxidase levels (a marker for myeloid cell infiltration) (Fogel et al., 2005; 2007,; Varga et al., 2005; Dunford et al., 2006b). This is in line with previous evidence demonstrating that blockade of H4 receptors impedes neutrophil recruitment and cytokine release in other models of inflammation, such as zymosan-induced pleuritis and allergic airway inflammation (Takeshita et al., 2003; Thurmond et al., 2004; Dunford et al., 2006a). The anti-inflammatory effect of H4 receptor antagonism resulted – at least partly – from inhibition of aberrant Toll-like receptor signalling via dendritic cells leading to reduced production of TNF-α and IL-6 (Fogel et al., 2005; Varga et al., 2005; Dunford et al., 2006b). In addition, colonic H4 receptor expression was reported to be increased in the colon of mice with TNBS-induced colitis and during spontaneous colitis in Gαi2 protein-deficient mice (Sutton et al., 2008; Kumawat et al., 2010). Whether H4 receptor expression is also increased in IBD patients is an interesting question that has not been investigated to our knowledge.

Data have also emerged, suggesting a possible role for H4 receptors in mediating gastrointestinal inflammation in ischaemia/reperfusion models. However, in most of these studies non-selective antagonists were used, making it difficult to ascertain that this effect was indeed solely mediated by the H4 receptor subtype. In a mouse model of mesenteric ischaemia/reperfusion injury treatment with the H3/H4 receptor antagonist thioperamide significantly reduced myeloperoxidase activity (Ghizzardi et al., 2009). In contrast, the opposite effect was seen on hepatic ischaemia/reperfusion damage: histamine, the H2/H4 receptor agonist dimaprit and the H3/H4 receptor agonist clozapine reduced post-ischemic liver damage, as shown by a reduction in serum transaminases (Adachi et al., 2006). This protective effect was abolished by the H3/H4 receptor antagonist thioperamide but remained unaffected by the selective H2 receptor antagonist cimetidine, suggesting a beneficial influence of H4 receptor stimulation in the prevention of ischaemia/reperfusion liver damage. Recently, the mechanism of action was further elucidated by El-Mahdy et al. (2013). They found that liver damage was significantly reduced by pretreatment with histamine, remained unaffected by a selective H1 or H2 receptor antagonist, was abolished by the H3/H4 receptor antagonist thioperamide and was reproduced by the H3/H4 receptor agonist clozapine. The protective effect of histamine and clozapine was mediated by attenuating TNF-α and IL-12 secretion and consequently reduced reactive oxygen species (El-Mahdy et al., 2013). As H3 receptors were absent from adult mouse liver tissue (Heron et al., 2001), it seems reasonable to assume that the protective effect of histamine and clozapine was indeed mediated by H4 receptors. However, it is important to exclude the possibility that hepatic ischaemia/reperfusion does not induce H3 receptor expression to be sure that the effect is due to H4 receptor modulation.

Pronounced gastrointestinal inflammation is seen after radiation and results from reactive oxygen/nitrate species, apoptosis and clonogenic cell death, mucosal breakdown and transcription of proinflammatory cytokines, chemokines and growth factors (Francois et al., 2013). In view of the promising results of H4 receptor blockade on gastrointestinal inflammation in other animal models, Martinel Lamas et al. (2013) evaluated the radioprotective potential of JNJ7777120, a selective H4 receptor antagonist. Preventive treatment with JNJ7777120 preserved the villi and the number of crypts in the small intestine and diminished mucosal atrophy after radiation by reducing apoptosis and DNA damage in enterocytes (Martinel Lamas et al., 2013).

Finally, preliminary evidence also points towards a possible involvement of H4 receptors in allergic gut reactions (Yu et al., 2008). Actively sensitized guinea pigs were exposed to inhaled 0.1% ovalbumin; MC and eosinophil infiltration into the oesophagus was assessed 1 h later. Pretreatment with the H3/H4 receptor antagonist thioperamide inhibited migration of both cell types to the oesophageal epithelium (Yu et al., 2008). As both MCs and eosinophils did not express H3 receptors, the effect was ascribed to blockade of H4 receptors, which seems consistent with previous reports of H4 receptor-mediated chemotaxis of these cell types (Hofstra et al., 2003; Thurmond et al., 2004; Yu et al., 2008).

In conclusion, these in vivo experiments suggest that H4 receptors participate in mediating gastrointestinal inflammation and immune responses in a variety of animal models. These findings are in line with previous preclinical observations from immune-mediated disorders in other organ systems and underline the immunomodulatory role of H4 receptors. However, further research confirming these findings using highly selective ligands for H4 receptors are much needed before clinical trials can be initiated for gastrointestinal inflammation and immune-mediated disorders.

H4 receptors and carcinogenesis

Enhanced expression of L-histidine decarboxylase and high histamine producing and secreting capabilities have been reported in malignancies, such as melanoma, breast, colorectal and pancreatic carcinoma both in experimental models and in human tumour biopsies (Medina and Rivera, 2010; Kennedy et al., 2012). Histamine, released by the malignant cells themselves or by other histamine-secreting cells in the environment such as MCs, acts as a growth factor in an autocrine or paracrine fashion, regulating angiogenesis, cell invasion, migration, differentiation, apoptosis and immune suppression (Medina and Rivera, 2010). These results suggest an important role for histamine in tumour development and progression. Histamine-induced cell proliferation seems to be mediated via H2 receptors as antagonists for these receptors induced apoptosis in human colorectal and gastric cancer cell lines and in experimental models (Rajendra et al., 2004; Jiang et al., 2010). These findings culminated in clinical trials evaluating the effect of H2 receptor-targeted therapy in colorectal cancer, indicating a beneficial effect when H2 receptor antagonists were given as therapy, adjuvant to curative surgical resection (Deva and Jameson, 2012). Interestingly, H2 receptor expression was comparable in colorectal cancer and adjacent normal mucosal specimens, whereas H1 receptor and H4 receptor expression were significantly reduced in tumour tissue (Boer et al., 2008; Fang et al., 2011). These findings suggest that carcinogenesis might benefit from loss of H4 receptors (and H1 receptors). A potential antiproliferative action of H4 receptors in colorectal cancer was further substantiated by in vitro experiments demonstrating that stimulation of H4 receptors induced a cell cycle arrest in the G1 phase via a cAMP-dependent pathway, resulting in reduced cell proliferation and tumour growth (Table 2013) (Fang et al., 2011). This antiproliferative action was only present in H4 receptor-expressing colorectal cancer cell lines, but not in mock-transfected cells and could be prevented by pretreatment with the selective H4 receptor antagonist JNJ7777120, further corroborating involvement of these receptors. In addition, H4 receptor stimulation enhanced apoptosis induced by the chemotherapeutic agent 5-fluorouracil (Fang et al., 2011). In contrast, Cianchi et al. (2005) found that H4 receptor expression was increased in colorectal cancer specimens. Moreover, histamine-exposure stimulated cell proliferation and VEGF levels, which were reduced by the H4 receptor antagonist JNJ7777120 (and the H2 receptor antagonist cimetidine). This proliferative effect of H4 receptor stimulation was mediated by COX 2-induced PGE2 as it was only evident in those cell lines that expressed COX 2 (Cianchi et al., 2005). In addition, JNJ7777120 only reduced histamine-induced cell proliferation, but did not affect basal (non-histamine stimulated) cell growth (Coruzzi et al., 2012).

Table 4.

Preclinical in vitro and in vivo experiments with H4 receptor ligands on carcinogenesis

Model Species In vitro/in vivo Ligand Effect Ref
Colorectal cancer cell line Human In vitro Clozapine Clozapine and clobenpropit reduced cell growth Fang et al. (2011)
Clobenpropit Clozapine enhanced 5-FU induced apoptosis, which was reversed by JNJ7777120
JNJ7777120
Colorectal cancer cell line Human In vitro JNJ7777120 JNJ7777120 prevented histamine-induced COX-2 expression/activity, cell proliferation and VEGF production Cianchi et al. (2005)
Gastric cancer cell line Human In vitro Clobenpropit JNJ7777120 abolished clobenpropit-induced cell growth Zhang et al. (2012)
JNJ7777120
Pancreatic duct carcinoma cell line Human In vitro Clobenpropit Clobenpropit stimulation reduces cell growth Cricco et al. (2008)
Cholangiocarcinoma cell line Human In vitro Clobenpropit Clobenpropit inhibited cell proliferation and metastatic potential Meng et al. (2011)
Cholangiocarcinoma cell line Human In vitro Thioperamide No effect on histamine secretion and cell growth Francis et al. (2012)
Xenograft cholangiocarcinoma Mouse In vivo Clobenpropit Clobenpropit inhibited tumour growth Meng et al. (2011)

5-FU, 5-fluorouracil; clobenpropit, H3 receptor antagonist, H4 receptor agonist; clozapine, H3 and H4 receptor agonist; JNJ7777120, H4 receptor antagonist; thioperamide, H3 and H4 receptor antagonist.

Attenuated H4 receptor expression was reported in human gastric cancer specimens and was most prominent in advanced malignancies (Zhang et al., 2012). Similarly to what was previously demonstrated in colorectal cancer, reduced H4 receptor expression was linked to enhanced cell proliferation as H4 receptor stimulation with clobenpropit and histamine reduced the growth of gastric cancer cells (Zhang et al., 2012). Although neither ligand is an exclusive H4 receptor agonist, the involvement of H4 receptors was inferred from the fact that pretreatment with the selective H4 receptor antagonist JNJ7777120 completely abolished agonist-induced responses (Zhang et al., 2012). In line with this, clobenpropit reduced tumour cell proliferation in a pancreatic duct carcinoma cell line (Cricco et al., 2008).

In contrast, H4 receptor expression was enhanced in malignant cholangiocytes from patients with proven cholangiocarcinoma (Meng et al., 2011). H4 receptor stimulation with the H3 receptor antagonist/H4 receptor agonist clobenpropit dose-dependently reduced proliferation of several cholangiocarcinoma cell lines in vitro (Meng et al., 2011). This cytostatic effect resulted from reduced growth potential and disruption of the invading capacity of the cells. As the effect of clobenpropit was maintained in in vitro experiments in which H3 receptors were knocked down, this indicates that the effects were indeed mediated via H4 receptors. Importantly, in an elegant in vivo design, the authors demonstrated the clinical potential of H4 receptor-modulation in this tumour type as treatment with clobenpropit inhibited tumour growth and disrupted its invasive potential in a xenographic cholangiocarcinoma mouse model (Meng et al., 2011). However, in another study, inhibition of H4 receptors by the H3/H4 receptor antagonist thioperamide did not affect cholangiocarcinoma cell line proliferation (Francis et al., 2012).

Overall, these findings indicate that depending on the type of tumour (gastric vs. colorectal vs. cholangiocarcinoma) H4 receptor-expression can either be decreased or enhanced. It is unclear whether H4 receptor expression differs in early versus advanced stages, and this would be interesting to investigate further. In addition, although the data gathered from in vitro experiments using different cell lines strongly indicate that H4 receptors can potently modulate tumour growth and progression, the results are not univocal. To complement these in vitro findings and increase our understanding of the role of H4 receptors in gastrointestinal carcinogenesis, additional research and in vivo experiments using selective ligands seem crucial.

H4 receptors and visceral sensory signalling

Visceral hypersensitivity refers to an enhanced perception of stimuli originating from the internal organs and is believed to contribute to abdominal pain in multiple gastrointestinal disorders among which IBD, IBS and functional dyspepsia (Vermeulen et al., 2014). Sensitization of afferent nerve endings in the gut wall is thought to underlie visceral hypersensitivity (Anand et al., 2007). Several lines of evidence indicate that histamine is involved in this process (Buhner and Schemann, 2012; van Diest et al., 2012). For instance, supernatant from IBS colonic biopsies contains increased levels of histamine (Barbara et al., 2007). When applied to human submucous neurons, this supernatant increased neuronal activity and the degree of activation correlated with histamine levels in the supernatant (Buhner et al., 2009). In addition, histamine induced murine jejunal afferent firing and excited primary sensory neurons (Kreis et al., 1998; Brunsden and Grundy, 1999). The pro-nociceptive effect of histamine seems to be mediated – at least partially – by H1 receptors expressed on sensory afferents, which is consistent with the finding that excitation of rat jejunal afferents by IBS supernatant can be reduced by application of the H1 receptor antagonist pyrilamine (Barbara et al., 2007). In addition, a role for H4 receptors in mediating visceral sensory signalling and nociception has emerged (Table 2013a).

Table 5.

Preclinical in vitro and in vivo experiments with H4 receptor ligands in visceral sensory signalling and nociception

Model Species In vitro/in vivo Ligand Effect Ref
Submucous plexus neurons Human In vitro 4-methylhistamine 4-methylhistamine-induced excitation reduced by JNJ7777120 Breunig et al. (2007)
JNJ7777120
Jejunal afferent firing Rat In vitro Thioperamide Thioperamide reduced histamine-induced jejunal afferent firing Brunsden and Grundy (1999)
Jejunal afferent firing Rat In vivo Thioperamide No effect on histamine-induced jejunal afferent firing Kreis et al. (1998)
Post-inflammatory visceral hypersensitivity Rat In vivo JNJ7777120 JNJ7777120 dose-dependently reversed visceral hypersensitivity Deiteren et al. (2014)

4-methylhistamine, H4 receptor agonist; JNJ7777120, H4 receptor antagonist; thioperamide, H3 and H4 receptor antagonist.

Breunig et al. (2007) reported that the H4 receptor agonist 4-methylhistamine excited human submucous plexus neurons, an effect that was inhibited by the selective H4 receptor antagonist JNJ7777120. Also, in vitro jejunal afferent excitation by histamine was reversed by the H3/H4 receptor antagonist thioperamide (Brunsden and Grundy, 1999), although these results are in contrast to earlier reports in a similar set-up (Kreis et al., 1998). Recently, our group provided in vivo evidence of reduced visceral nociception after blockade of H4 receptors. Post-inflammatory visceral hypersensitivity was dose-dependently reduced by JNJ7777120 in a rat model of post-inflammatory IBS, without affecting visceral sensitivity in healthy controls (Deiteren et al., 2014). Although increased colonic expression of H4 receptor mRNA in hypersensitive rats points towards a peripheral mechanism of action, it remains to be determined whether the antinociceptive effect is mediated by blockade of H4 receptors on sensory afferents directly or indirectly by modulation of H4 receptors expressed elsewhere in the gut wall (Deiteren et al., 2014). Nevertheless, these findings coincide with previous reports of antinociceptive and analgesic effects of H4 receptor antagonists in models of somatic and neuropathic pain (independent of their anti-inflammatory properties) (Coruzzi et al., 2007; Hsieh et al., 2010) and emphasize that H4 receptors are also attractive targets in the modulation of visceral pain.

H4 receptors and intestinal contractility

Histaminergic control of gastrointestinal contractility and motility is complex and involves all histamine receptor subtypes. H1 receptors, located in smooth muscle cells, contribute to contractility by increasing calcium availability at the sarcoplasmic level whereas H2 receptors mainly facilitate cholinergic and non-cholinergic excitatory transmission in intramural neurons (Poli et al., 2001). Although H3 receptors inhibit the release of excitatory and inhibitory neurotransmitters from the myenteric plexus, their involvement in enteric peristalsis remains unclear, as no effect of H3 receptor ligands on gastrointestinal transit was seen in in vivo models and the presence of H3 receptors in the human digestive tract remains controversial (Hemedah et al., 2001; Poli et al., 2001; Cianchi et al., 2005; Sander et al., 2006). Recently, H4 receptors were reported to be present on murine myenteric neurons (Chazot et al., 2007). In addition, 4-methylhistamine excited human submucous plexus neurons, which could be blocked by the H4 receptor antagonist JNJ7777120 (Breunig et al., 2007). As the enteric plexus in highly involved in the regulation of reflex behaviour, peristalsis and intestinal secretion, these findings suggest that H4 receptors could be involved in gut motility and transit (Table 2013b). However, the H4 receptor agonist VUF8430 did not affect twitch responses induced by electrical field stimulation in rat duodenum (Pozzoli et al., 2009). In addition, no effect was seen from H4 receptor stimulation on the membrane potential of murine small intestinal interstitial cells of Cajal, the enteric pacemaker cells and conductors of electrical slow waves in intestinal smooth muscle (Kim et al., 2013). In a recent study, longitudinal smooth muscle preparations with an intact myenteric plexus were harvested from guinea pig ileum and exposed to supernatants prepared from colonic biopsies from IBS patients. This supernatant enhanced cholinergic twitch contractions; however, the responses were not affected by a mixture containing antagonists for H1–H4 receptors (Balestra et al., 2012). The H4 receptor agonist 4-methylhistamine increased contractile forces only in longitudinal smooth muscle strips of monkey colon (Kim et al., 2011). However, as these effects were only present when high doses were used, these results need to be interpreted with caution.

Table 6.

Preclinical in vitro experiments with H4 receptor ligands in gastrointestinal contractility and transit

Model Species In vitro/in vivo Ligand Effect Ref
Submucous plexus neurons Human In vitro 4-methylhistamine 4-methylhistamine-induced excitation reduced by JNJ7777120 Breunig et al. (2007)
JNJ7777120
Whole mount duodenum segments Rat In vitro VUF8430 No effect on contractions Pozzoli et al. (2009)
Longitudinal smooth muscle incl. myenteric plexus Guinea pig In vitro Thioperamide No effect on contractions induced by IBS supernatant Balestra et al. (2012)
Colonic smooth muscle strips Monkey In vitro 4-methyl-histamine 4-methylhistamine increased contractile force Kim et al. (2011)
Cultured small intestine interstitial cells of Cajal Mouse In vitro 4-methyl-histamine No effect on pace maker potentials Kim et al. (2013)

4-methylhistamine, H4 receptor agonist; IBS, irritable bowel syndrome; JNJ7777120, H4 receptor antagonist; thioperamide, H3 and H4 receptor antagonist; VUF8430, H4 receptor agonist.

H4 receptors and gastric acid secretion and ulceration

Histamine is a potent activator of the acid secreting cells of the stomach (Kopic and Geibel, 2010). Binding of histamine to basolateral H4 receptors activates adenylyl cyclase resulting in accumulation of cAMP and H+ secretion. Before the development of proton pump inhibitors, pharmacological blockade of H2 receptors was the cornerstone of the treatment of acid-related gastrointestinal disorders (Kopic and Geibel, 2010). In addition to H2 receptor antagonists, H3 receptor stimulation also exerted gastroprotective effects via increased mucus production in animal models (Coruzzi et al., 2001; Barocelli and Ballabeni, 2003). The homology between H3 and H4 receptors subsequently spurred interest in a possible role for H4 receptors in gastric acid secretion (Table 2011). Overall, the data gathered to date suggest that H4 receptors do not participate in gastric acid secretion under physiological conditions as neither H4 receptor agonists such as VUF8430 and VUF10460 nor H4 receptor antagonists such as JNJ7777120 and VUF5949 affected basal acid production or the macroscopic appearance of the stomach (Lim et al., 2009; Coruzzi et al., 2011; Adami et al., 2012). However, when the mucosal integrity was compromised such as in models of chemically induced gastric ulceration, damage was significantly enhanced by H4 receptor stimulation and markedly reduced by its blockade (Adami et al., 2005; 2012,; Coruzzi et al., 2009; 2011,). In addition, enhanced chemically induced mucosal damage by H4 receptor agonists could be prevented by concomitant H4 receptor antagonists and vice versa (Coruzzi et al., 2009; 2011,). However, the findings are not fully consistent as the H4 receptor agonists VUF8430 and VUF10460 had no effect on indomethacin/bethanecol-induced lesions in a mouse model whereas the HCl-induced damage in rats was enhanced by both agonists, and in contrast, indomethacin-induced ulcerations were reduced by VUF10460 (Coruzzi et al., 2009; 2011,; Adami et al., 2012). It was hypothesized that species and strain differences might contribute to the differential effects as JNJ7777120 effectively reduced indomethacin/bethanecol-induced lesions in CD-1, NMRI and BALB/c, but not in C57BL/6J mice (Adami et al., 2012). However, it should be kept in mind that several of the compounds used also display considerable affinity for the H3 receptor, such as VUF8430 [pKi for rat H4 receptors of 6.9 vs. 6.5 for rat H3 receptors (Lim et al., 2009); Table 2006 ], again underscoring the need for selective H4 receptor ligands. Although these data seem promising, more research is needed to further elucidate the effect of H4 receptor modulation on gastric ulcer disease. If a beneficial effect of H4 receptor blockade on gastric ulceration could be confirmed, this would be a major advantage in terms of drug development, as H4 receptor antagonists are already under evaluation for their anti-inflammatory and analgesic properties.

Table 7.

Preclinical in vivo experiments with H4 receptor ligands on gastric acid secretion and ulceration

Model Species In vitro/in vivo Ligand Effect Ref
Gastric acid secretion Rat In vivo Dimaprit Dimaprit potently induced gastric secretion, whereas VUF8430 only marginally increased secretion Lim et al. (2009)
VUF8430
JNJ7777120 Induced gastric acid secretion was not affected by JNJ7777120
Indomethacin/bethanechol-induced gastric ulceration Mouse In vivo JNJ7777120 JNJ7777120 reduces lesions in CD-1, NMRI and BALB/c, but not in C57BL/6J mice. Adami et al. (2012; Coruzzi et al. (2009)
VUF10460 No effect of VUF10460 and VUF8430 on gastric lesions
VUF8430
VUF10460 abolished the protective effect of JNJ7777120
Indomethacin-induced gastric ulceration Rat In vivo VUF5949 VUF5949 and JNJ7777120 reduced indomethacin-induced lesions Adami et al. (2005); Coruzzi et al. (2009)
JNJ7777120
VUF10460 VUF10460 reduced lesions
VUF8430 VUF8430 only reduced lesions in the presence of a H2 receptor antagonist
HCl-induced gastric ulceration Rat In vivo Immepip Immepip, VUF8430 and VUF10460 enhanced HCl-induced gastric lesions Coruzzi et al. (2011)
VUF8430
VUF10460
JNJ7777120 JNJ7777120 abolished the effect of immepip, but not of VUF8430 and VUF10460

Dimaprit, H2 receptor agonist, H4 receptor agonist; Immepip, H3 and H4 receptor agonist; JNJ7777120, H4 receptor antagonist; VUF10460, H4 receptor agonist; VUF5949, H4 receptor antagonist; VUF8430, H4 receptor agonist.

Clinical development

To date, no clinical trials with H4 receptor ligands have been initiated in the field of gastroenterology. However, several H4 receptor antagonists have already progressed to phase II clinical trials for immune-mediated disorders such as rheumatoid arthritis, asthma, atopic dermatitis and allergic rhinitis (Table 2007). Currently registered clinical trials (http://clinicaltrials.gov) include compounds from Johnson & Johnson (JNJ39758979 and JNJ38518168), Ziarco Pharma (ZPL3893787) and Palau Pharma (UR-63325).

Table 8.

Current clinical trials with H4 receptor ligands

Compound Phase Population Status Trial number
JNJ39758979 I Healthy volunteers Completed NCT01081821
I Histamine-induced itch in healthy volunteers Completed NCT01068223
I Rheumatoid arthritis Completed NCT01442545
II Asthma Completed NCT00946569
II Atopic dermatitis Terminated NCT01497119
II Rheumatoid arthritis Terminated NCT01480388
II Asthma Withdrawn NCT01493882
JNJ38518168 I Healthy volunteers Completed NCT01442532
I Patients with normal or mild to moderate hepatic impairment Completed NCT01863784
I Healthy volunteers Completed NCT01970020
I Healthy volunteers on ketonazole Completed NCT01690286
I Rheumatoid arthritis Completed NCT01450982
II Rheumatoid arthritis Recruiting NCT01862224
II Rheumatoid arthritis Active, not recruiting NCT01679951
II Rheumatoid arthritis Terminated* NCT00941707
II Asthma Recruiting NCT01823016
ZPL3893787§ I Healthy volunteers Completed NCT00992342
I Asthma Completed NCT00856687
UR63325 II Allergic rhinitis Completed NCT01260753
*

Terminated because of a single, unexpected serious event.

Terminated because of two cases of agranulocytosis.

Terminated/withdrawn because of cases of agranulocytosis in trial NCT01497119.

§

Former PF03893787. Clinical trials as registered on http://clinicaltrials.gov on 11 June 2014.

JNJ39758979, derived from the H4 receptor antagonist JNJ7777120, showed promising results in initial phase I trials, with good pharmacokinetics upon oral dosing with a plasma half-life of 124–157 h after a single oral dose (Thurmond et al., 2014). In addition, the compound was well-tolerated up to 1200 mg in single ascending dose studies and up to 300 mg bid in a multiple ascending dose study; dose-dependent gastrointestinal symptoms were the main adverse events (abnormal faeces, nausea, vomiting and abdominal pain) (Thurmond et al., 2014). A single dose of 600 mg effectively reduced histamine-induced itch in 23 healthy volunteers (Kollmeier et al., 2014). However, a subsequent phase II trial in patients with atopic dermatitis was discontinued due to two cases of drug-induced agranulocytosis, leading to the termination of JNJ39758979 (Thurmond et al., 2014). The agranulocytosis was reported to be related to the chemical structure of JNJ39758979 and not to the H4 receptor antagonism (Kollmeier et al., 2014; Liu, 2014; Thurmond et al., 2014); further details are expected to be released in the near future (Thurmond et al., 2014). Therefore, the development of other H4 receptor antagonists is currently being pursued such as JNJ38518168, which has progressed to phase II for rheumatoid arthritis and asthma. However, one of these trials was terminated because of a single, unexpected serious event, which was not specified further. Details on the underlying mechanisms (H4 receptor-related or compound-specific) are not yet available.

ZPL3893787 (former PF03893787) is a lead compound of Ziarco Pharma and successfully completed phase I single ascending dose and 14 days multiple ascending dose studies in healthy volunteers (Liu, 2014). No results have been published yet; however, Ziarco Pharma communicated on their website that the compound displayed an excellent pharmacokinetic and safety profile. Results from a subsequent proof of concept trial in patients with asthma have not yet been disclosed.

The Palau Pharma compound UR-63325 successfully completed single and multiple dose ascending studies demonstrating a linear pharmacokinetic profile and no safety concerns according to Salcedo et al. (2013); in addition, a phase II clinical trial in allergic rhinitis patients was recently completed and the data are eagerly awaited.

Conclusions

Since their discovery and cloning almost 15 years ago, knowledge on the role of H4 receptors has increased rapidly. The expression of H4 receptors on immune cells has spurred interest in H4 receptor antagonists as a potential new class of anti-inflammatory drugs in the treatment of rheumatoid arthritis and asthma among others. Also in the gastrointestinal tract, there is now strong preclinical evidence that H4 receptors modulate the inflammatory process, indicating that these receptors could be interesting new targets in the treatment of IBD, ischaemia/reperfusion injury, radiation-induced enteropathy and allergic intestinal reactions. It would be interesting to investigate whether genetic polymorphisms and copy gene number variations for H4 receptors are linked to gastrointestinal inflammation as was previously reported for other immune-mediated disorders such asthma and atopic dermatitis (Yu et al., 2010; Simon et al., 2012; Chen et al., 2013). In addition, recent data indicate that H4 receptors also participate in carcinogenesis and gastric ulceration and in mediating IBS-like visceral pain. The preliminary data gathered so far seem promising, but the effects of pharmacological H4 receptor modulation will need to be confirmed using highly selective ligands, that are devoid of biased signalling and are extensively evaluated in both in vitro and in vivo settings. In addition, the results of ongoing trials with H4 receptor antagonists for immune-mediated disorders are eagerly awaited and will be crucial for the future of any therapy targeted at H4 receptors.

Acknowledgments

A Deiteren is an aspirant of the Fund for Scientific Research (FWO), Flanders. This work was supported financially by the FWO (G.0341.13 and G.0249.09N).

Glossary

Abbreviations

IBD

inflammatory bowel disease

IBS

irritable bowel syndrome

MC

mast cell

TNBS

trinitrobenzene sulphonic acid

Conflicts of interest

The authors report no conflicts of interest.

References

  1. Adachi N, Liu K, Motoki A, Nishibori M, Arai T. Suppression of ischemia/reperfusion liver injury by histamine H4 receptor stimulation in rats. Eur J Pharmacol. 2006;544:181–187. doi: 10.1016/j.ejphar.2006.06.053. [DOI] [PubMed] [Google Scholar]
  2. Adami M, Coruzzi G, Guaita E, de Esch I, Leurs R. 2005. Antiinflammatory, analgesic and gastroprotective effects of the novel and selective histamine H4-receptor antagonist VUF5949. 34th Meeting of the European Histamine Research Society p. 47. [DOI] [PubMed]
  3. Adami M, Pozzoli C, Menozzi A, Bertini S, Passeri B, Cantoni AM, et al. Effects of histamine H4 receptor ligands in a mouse model of gastric ulceration. Pharmacology. 2012;89:287–294. doi: 10.1159/000337736. [DOI] [PubMed] [Google Scholar]
  4. Alcaniz L, Vega A, Chacon P, El Bekay R, Ventura I, Aroca R, et al. Histamine production by human neutrophils. FASEB. 2013;27:2902–2910. doi: 10.1096/fj.12-223867. [DOI] [PubMed] [Google Scholar]
  5. Alexander SP, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol. 2013a;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL. Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Enzymes. Br J Pharmacol. 2013b;170:1797–1867. doi: 10.1111/bph.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Alfon J, Sanchez-Gomez S, Salcedo C, Fernandez E, Gil-Torregrosa B, Ardanaz N, et al. Efficacy of UR-63325, a new histamine H4 receptor antagonist in house dust mite-induced mouse asthma models. Am J Respir Crit Care Med. 2011;183:A1302. [Google Scholar]
  8. Anand P, Aziz Q, Willert R, van Oudenhove L. Peripheral and central mechanisms of visceral sensitization in man. Neurogastroenterol Motil. 2007;19(1 Suppl):29–46. doi: 10.1111/j.1365-2982.2006.00873.x. [DOI] [PubMed] [Google Scholar]
  9. Andaloussi M, Lim HD, van der Meer T, Sijm M, Poulie CB, de Esch IJ, et al. A novel series of histamine H4 receptor antagonists based on the pyrido[3,2-d]pyrimidine scaffold: comparison of hERG binding and target residence time with PF-3893787. Bioorg Med Chem Lett. 2013;23:2663–2670. doi: 10.1016/j.bmcl.2013.02.091. [DOI] [PubMed] [Google Scholar]
  10. Balestra B, Vicini R, Cremon C, Zecchi L, Dothel G, Vasina V, et al. Colonic mucosal mediators from patients with irritable bowel syndrome excite enteric cholinergic motor neurons. Neurogastroenterol Motil. 2012;24:1118–e1570. doi: 10.1111/nmo.12000. [DOI] [PubMed] [Google Scholar]
  11. Barbara G, Stanghellini V, De Giorgio R, Corinaldesi R. Functional gastrointestinal disorders and mast cells: implications for therapy. Neurogastroenterol Motil. 2006;18:6–17. doi: 10.1111/j.1365-2982.2005.00685.x. [DOI] [PubMed] [Google Scholar]
  12. Barbara G, Wang B, Stanghellini V, de Giorgio R, Cremon C, Di Nardo G, et al. Mast cell-dependent excitation of visceral-nociceptive sensory neurons in irritable bowel syndrome. Gastroenterology. 2007;132:26–37. doi: 10.1053/j.gastro.2006.11.039. [DOI] [PubMed] [Google Scholar]
  13. Barocelli E, Ballabeni V. Histamine in the control of gastric acid secretion: a topic review. Pharmacol Res. 2003;47:299–304. doi: 10.1016/s1043-6618(03)00009-4. [DOI] [PubMed] [Google Scholar]
  14. Beermann S, Seifert R, Neumann D. Commercially available antibodies against human and murine histamine H(4)-receptor lack specificity. Naunyn Schmiedebergs Arch Pharmacol. 2012;385:125–135. doi: 10.1007/s00210-011-0700-4. [DOI] [PubMed] [Google Scholar]
  15. Bencsath M, Paloczi K, Szalai C, Szenthe A, Szeberenyi J, Falus A. Histidine decarboxylase in peripheral lymphocytes of healthy individuals and chronic lymphoid leukemia patients. Pathol Oncol Res. 1998;4:121–124. doi: 10.1007/BF02904705. [DOI] [PubMed] [Google Scholar]
  16. Bischoff SC, Wedemeyer J, Herrmann A, Meier PN, Trautwein C, Cetin Y, et al. Quantitative assessment of intestinal eosinophils and mast cells in inflammatory bowel disease. Histopathology. 1996;28:1–13. doi: 10.1046/j.1365-2559.1996.262309.x. [DOI] [PubMed] [Google Scholar]
  17. Black JW, Duncan WA, Durant CJ, Ganellin CR, Parsons EM. Definition and antagonism of histamine H2-receptors. Nature. 1972;236:385–390. doi: 10.1038/236385a0. [DOI] [PubMed] [Google Scholar]
  18. Boer K, Helinger E, Helinger A, Pocza P, Pos Z, Demeter P, et al. Decreased expression of histamine H1 and H4 receptors suggests disturbance of local regulation in human colorectal tumours by histamine. Eur J Cell Biol. 2008;87:227–236. doi: 10.1016/j.ejcb.2007.12.003. [DOI] [PubMed] [Google Scholar]
  19. Breunig E, Michel K, Zeller F, Seidl S, Weyhern CW, Schemann M. Histamine excites neurones in the human submucous plexus through activation of H1, H2, H3 and H4 receptors. J Physiol. 2007;583(Pt 2):731–742. doi: 10.1113/jphysiol.2007.139352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Brunsden AM, Grundy D. Sensitization of visceral afferents to bradykinin in rat jejunum in vitro. J Physiol. 1999;521(Pt 2):517–527. doi: 10.1111/j.1469-7793.1999.00517.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Buhner S, Schemann M. Mast cell-nerve axis with a focus on the human gut. Biochim Biophys Acta. 2012;1822:85–92. doi: 10.1016/j.bbadis.2011.06.004. [DOI] [PubMed] [Google Scholar]
  22. Buhner S, Li Q, Vignali S, Barbara G, De Giorgio R, Stanghellini V, et al. Activation of human enteric neurons by supernatants of colonic biopsy specimens from patients with irritable bowel syndrome. Gastroenterology. 2009;137:1425–1434. doi: 10.1053/j.gastro.2009.07.005. [DOI] [PubMed] [Google Scholar]
  23. Chazot PL, Shenton FC, Waldvogel HJ, Grandi D, Morini G. 2007. The H4 histamine receptor is expressed in both the human CNS and rodent PNS. 36th Annual Meeting of the European Histamine Research Society, Florence, Italy, p. O4.
  24. Chen B, Ye T, Shao Y, Zhang J, Zhong Q, Hu X, et al. Association between copy-number variations of the human histamine H4 receptor gene and atopic dermatitis in a Chinese population. Clin Exp Dermatol. 2013;38:295–300. doi: 10.1111/ced.12117. , quiz 300-301. [DOI] [PubMed] [Google Scholar]
  25. Cianchi F, Cortesini C, Schiavone N, Perna F, Magnelli L, Fanti E, et al. The role of cyclooxygenase-2 in mediating the effects of histamine on cell proliferation and vascular endothelial growth factor production in colorectal cancer. Clin Cancer Res. 2005;11(19 Pt 1):6807–6815. doi: 10.1158/1078-0432.CCR-05-0675. [DOI] [PubMed] [Google Scholar]
  26. Coge F, Guenin SP, Rique H, Boutin JA, Galizzi JP. Structure and expression of the human histamine H4-receptor gene. Biochem Biophys Res Commun. 2001;284:301–309. doi: 10.1006/bbrc.2001.4976. [DOI] [PubMed] [Google Scholar]
  27. Coruzzi G, Morini G, Adami M, Grandi D. Role of histamine H3 receptors in the regulation of gastric functions. J Physiol Pharmacol. 2001;52(4 Pt 1):539–553. [PubMed] [Google Scholar]
  28. Coruzzi G, Adami M, Guaita E, de Esch IJ, Leurs R. Antiinflammatory and antinociceptive effects of the selective histamine H4-receptor antagonists JNJ7777120 and VUF6002 in a rat model of carrageenan-induced acute inflammation. Eur J Pharmacol. 2007;563:240–244. doi: 10.1016/j.ejphar.2007.02.026. [DOI] [PubMed] [Google Scholar]
  29. Coruzzi G, Adami M, Pozzoli C, Smits R, De Esch I, Leurs R. 2009. Gastroprotective effects of histamine H4 receptor ligands in rodent ulcer models. Presented at the British Pharmacological Society, London, UK.
  30. Coruzzi G, Adami M, Pozzoli C, de Esch IJ, Smits R, Leurs R. Selective histamine H(3) and H(4) receptor agonists exert opposite effects against the gastric lesions induced by HCl in the rat stomach. Eur J Pharmacol. 2011;669:121–127. doi: 10.1016/j.ejphar.2011.07.038. [DOI] [PubMed] [Google Scholar]
  31. Coruzzi G, Adami M, Pozzoli C. Role of histamine H4 receptors in the gastrointestinal tract. Front Biosci (Schol Ed) 2012;4:226–239. doi: 10.2741/264. [DOI] [PubMed] [Google Scholar]
  32. Cowart MD, Altenbach RJ, Liu H, Hsieh GC, Drizin I, Milicic I, et al. Rotationally constrained 2,4-diamino-5,6-disubstituted pyrimidines: a new class of histamine H4 receptor antagonists with improved druglikeness and in vivo efficacy in pain and inflammation models. J Med Chem. 2008;51:6547–6557. doi: 10.1021/jm800670r. [DOI] [PubMed] [Google Scholar]
  33. Cricco GP, Mohamad NA, Sambuco LA, Genre F, Croci M, Gutierrez AS, et al. Histamine regulates pancreatic carcinoma cell growth through H3 and H4 receptors. Inflamm Res. 2008;57(Suppl. 1):S23–S24. doi: 10.1007/s00011-007-0611-5. [DOI] [PubMed] [Google Scholar]
  34. Dawicki W, Marshall JS. New and emerging roles for mast cells in host defence. Curr Opin Immunol. 2007;19:31–38. doi: 10.1016/j.coi.2006.11.006. [DOI] [PubMed] [Google Scholar]
  35. Deiteren A, De Man JG, Ruyssers NE, Moreels TG, Pelckmans PA, De Winter BY. Histamine H4 and H1 receptors contribute to postinflammatory visceral hypersensitivity. Gut. 2014;63:1873–1882. doi: 10.1136/gutjnl-2013-305870. [DOI] [PubMed] [Google Scholar]
  36. Deva S, Jameson M. Histamine type 2 receptor antagonists as adjuvant treatment for resected colorectal cancer. Cochrane Database Syst Rev. 2012;(8) doi: 10.1002/14651858.CD007814.pub2. CD007814. [DOI] [PubMed] [Google Scholar]
  37. van Diest SA, Stanisor OI, Boeckxstaens GE, de Jonge WJ, van den Wijngaard RM. Relevance of mast cell-nerve interactions in intestinal nociception. Biochim Biophys Acta. 2012;1822:74–84. doi: 10.1016/j.bbadis.2011.03.019. [DOI] [PubMed] [Google Scholar]
  38. Dunford PJ, O'Donnell N, Riley JP, Williams KN, Karlsson L, Thurmond RL. The histamine H4 receptor mediates allergic airway inflammation by regulating the activation of CD4 + T cells. J Immunol. 2006a;176:7062–7070. doi: 10.4049/jimmunol.176.11.7062. [DOI] [PubMed] [Google Scholar]
  39. Dunford PJ, Varga C, Thurmond RL, Whittle BJ. Histamine H4 receptor antagonism attenuates toll-like receptor signaling and inhibits experimental colitis in the rat. Gastroenterology. 2006b;130(4 Suppl. 2):A689. [Google Scholar]
  40. Eisenschenk MN, Torres SM, Oliveira S, Been CS. The expression of histamine H4 receptor mRNA in the skin and other tissues of normal dogs. Vet Dermatol. 2011;22:396–400. doi: 10.1111/j.1365-3164.2011.00959.x. [DOI] [PubMed] [Google Scholar]
  41. El-Mahdy NA, El-Sisi AE, Dewidar BI, El-Desouky KI. Histamine protects against the acute phase of experimentally-induced hepatic ischemia/re-perfusion. J Immunotoxicol. 2013;10:9–16. doi: 10.3109/1547691X.2012.684158. [DOI] [PubMed] [Google Scholar]
  42. Eliakim R, Karmeli F, Okon E, Rachmilewitz D. Ketotifen effectively prevents mucosal damage in experimental colitis. Gut. 1992;33:1498–1503. doi: 10.1136/gut.33.11.1498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Endo Y. Simultaneous induction of histidine and ornithine decarboxylases and changes in their product amines following the injection of Escherichia coli lipopolysaccharide into mice. Biochem Pharmacol. 1982;31:1643–1647. doi: 10.1016/0006-2952(82)90394-x. [DOI] [PubMed] [Google Scholar]
  44. Fang Z, Yao W, Xiong Y, Li J, Liu L, Shi L, et al. Attenuated expression of HRH4 in colorectal carcinomas: a potential influence on tumor growth and progression. BMC Cancer. 2011;11:1–11. doi: 10.1186/1471-2407-11-195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Farhadi A, Keshavarzian A, Fields JZ, Jakate S, Shaikh M, Banan A. Reduced immunostaining for c-kit receptors in mucosal mast cells in inflammatory bowel disease. J Gastroenterol Hepatol. 2007;22:2338–2343. doi: 10.1111/j.1440-1746.2007.05011.x. [DOI] [PubMed] [Google Scholar]
  46. Fogel WA, Wagner W, Sasiak K, Stasiak A. The role of histamine in experimental ulcerative colitis in rats. Inflamm Res. 2005;54(Suppl. 1):S68–S69. doi: 10.1007/s00011-004-0431-9. [DOI] [PubMed] [Google Scholar]
  47. Fogel WA, Jochem J, Lewinski A. Influence of the H3/H4 receptor antagonist, thioperamide on regional haemodynamics in rats with trinitrobenzene sulfonic acid-induced colitis. Inflamm Res. 2007;56(Suppl. 1):S21–S22. doi: 10.1007/s00011-006-0510-1. [DOI] [PubMed] [Google Scholar]
  48. Francis H, DeMorrow S, Venter J, Onori P, White M, Gaudio E, et al. Inhibition of histidine decarboxylase ablates the autocrine tumorigenic effects of histamine in human cholangiocarcinoma. Gut. 2012;61:753–764. doi: 10.1136/gutjnl-2011-300007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Francois A, Milliat F, Guipaud O, Benderitter M. Inflammation and immunity in radiation damage to the gut mucosa. Biomed Res Int. 2013;2013:123241. doi: 10.1155/2013/123241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Fukushima Y, Ohmachi Y, Asano T, Nawano M, Funaki M, Anai M, et al. Localization of the histamine H(2) receptor, a target for antiulcer drugs, in gastric parietal cells. Digestion. 1999;60:522–527. doi: 10.1159/000007701. [DOI] [PubMed] [Google Scholar]
  51. Ghizzardi P, Gobbetti T, Bertoni S, Saccani F, Flammini L, Ballabeni V, Barocelli E. Histamine H4 receptor antagonism and mesenteric ischemia/reperfusion injury in mice. Gastroenterology. 2009;136(5 Suppl. 1):A397. [Google Scholar]
  52. He SH. Key role of mast cells and their major secretory products in inflammatory bowel disease. World J Gastroenterol. 2004;10:309–318. doi: 10.3748/wjg.v10.i3.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hemedah M, Loiacono R, Coupar IM, Mitchelson FJ. Lack of evidence for histamine H3 receptor function in rat ileum and human colon. Naunyn Schmiedebergs Arch Pharmacol. 2001;363:133–138. doi: 10.1007/s002100000345. [DOI] [PubMed] [Google Scholar]
  54. Heron A, Rouleau A, Cochois V, Pillot C, Schwartz JC, Arrang JM. Expression analysis of the histamine H(3) receptor in developing rat tissues. Mech Dev. 2001;105:167–173. doi: 10.1016/s0925-4773(01)00389-6. [DOI] [PubMed] [Google Scholar]
  55. Hofstra CL, Desai PJ, Thurmond RL, Fung-Leung WP. Histamine H4 receptor mediates chemotaxis and calcium mobilization of mast cells. J Pharmacol Exp Ther. 2003;305:1212–1221. doi: 10.1124/jpet.102.046581. [DOI] [PubMed] [Google Scholar]
  56. Hsieh GC, Chandran P, Salyers AK, Pai M, Zhu CZ, Wensink EJ, et al. H4 receptor antagonism exhibits anti-nociceptive effects in inflammatory and neuropathic pain models in rats. Pharmacol Biochem Behav. 2010;95:41–50. doi: 10.1016/j.pbb.2009.12.004. [DOI] [PubMed] [Google Scholar]
  57. Jiang CG, Liu FR, Yu M, Li JB, Xu HM. Cimetidine induces apoptosis in gastric cancer cells in vitro and inhibits tumor growth in vivo. Oncol Rep. 2010;23:693–700. doi: 10.3892/or_00000686. [DOI] [PubMed] [Google Scholar]
  58. Jiang W, Lim HD, Zhang M, Desai P, Dai H, Colling PM, et al. Cloning and pharmacological characterization of the dog histamine H4 receptor. Eur J Pharmacol. 2008;592:26–32. doi: 10.1016/j.ejphar.2008.06.095. [DOI] [PubMed] [Google Scholar]
  59. Jones BL, Kearns GL. Histamine: new thoughts about a familiar mediator. Clin Pharmacol Ther. 2011;89:189–197. doi: 10.1038/clpt.2010.256. [DOI] [PubMed] [Google Scholar]
  60. Jones NL, Roifman CM, Griffiths AM, Sherman P. Ketotifen therapy for acute ulcerative colitis in children: a pilot study. Dig Dis Sci. 1998;43:609–615. doi: 10.1023/a:1018827527826. [DOI] [PubMed] [Google Scholar]
  61. Jutel M, Akdis M, Akdis CA. Histamine, histamine receptors and their role in immune pathology. Clin Exp Allergy. 2009;39:1786–1800. doi: 10.1111/j.1365-2222.2009.03374.x. [DOI] [PubMed] [Google Scholar]
  62. Kennedy L, Hodges K, Meng F, Alpini G, Francis H. Histamine and histamine receptor regulation of gastrointestinal cancers. Transl Gastrointest Cancer. 2012;1:215–227. [PMC free article] [PubMed] [Google Scholar]
  63. Kim BJ, Kwon YK, Kim E, So I. Effects of histamine on cultured interstitial cells of cajal in murine small intestine. Korean J Physiol Pharmacol. 2013;17:149–156. doi: 10.4196/kjpp.2013.17.2.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kim H, Dwyer L, Song JH, Martin-Cano FE, Bahney J, Peri L, et al. Identification of histamine receptors and effects of histamine on murine and simian colonic excitability. Neurogastroenterol Motil. 2011;23:949–e409. doi: 10.1111/j.1365-2982.2011.01760.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Knutson L, Ahrenstedt O, Odlind B, Hallgren R. The jejunal secretion of histamine is increased in active Crohn's disease. Gastroenterology. 1990;98:849–854. doi: 10.1016/0016-5085(90)90006-m. [DOI] [PubMed] [Google Scholar]
  66. Kollmeier A, Francke K, Chen B, Dunford PJ, Greenspan AJ, Xia Y, et al. The histamine H4 receptor antagonist, JNJ 39758979, is effective in reducing histamine-induced pruritus in a randomized clinical study in healthy subjects. J Pharmacol Exp Ther. 2014;350:181–187. doi: 10.1124/jpet.114.215749. [DOI] [PubMed] [Google Scholar]
  67. Kopic S, Geibel JP. Update on the mechanisms of gastric acid secretion. Curr Gastroenterol Rep. 2010;12:458–464. doi: 10.1007/s11894-010-0137-9. [DOI] [PubMed] [Google Scholar]
  68. Kreis ME, Haupt W, Kirkup AJ, Grundy D. Histamine sensitivity of mesenteric afferent nerves in the rat jejunum. Am J Physiol. 1998;275(4 Pt 1):G675–G680. doi: 10.1152/ajpgi.1998.275.4.G675. [DOI] [PubMed] [Google Scholar]
  69. Kuhne S, Wijtmans M, Lim HD, Leurs R, de Esch IJ. Several down, a few to go: histamine H3 receptor ligands making the final push towards the market? Expert Opin Investig Drugs. 2011;20:1629–1648. doi: 10.1517/13543784.2011.625010. [DOI] [PubMed] [Google Scholar]
  70. Kumar V, Laddu AR, Lahiri PK, Sanyal RK. The physiological levels of histamine in human tissues. Int Arch Allergy Appl Immunol. 1968;34:233–236. doi: 10.1159/000230115. [DOI] [PubMed] [Google Scholar]
  71. Kumawat AK, Götlind Y-Y, Fredin MF, Willén R, Strid H, Hultgren-Hornquist E. Expression patterns of histamine receptors in the Gi2alpha-deficient mouse model of colitis. Inflamm Res. 2010;59(4 Suppl):S358–S359. [Google Scholar]
  72. Leurs R, Chazot PL, Shenton FC, Lim HD, de Esch IJ. Molecular and biochemical pharmacology of the histamine H4 receptor. Br J Pharmacol. 2009;157:14–23. doi: 10.1111/j.1476-5381.2009.00250.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Lim HD, Adami M, Guaita E, Werfel T, Smits RA, de Esch IJ, et al. Pharmacological characterization of the new histamine H4 receptor agonist VUF 8430. Br J Pharmacol. 2009;157:34–43. doi: 10.1111/j.1476-5381.2009.00200.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Liu C, Ma X, Jiang X, Wilson SJ, Hofstra CL, Blevitt J, et al. Cloning and pharmacological characterization of a fourth histamine receptor (H(4)) expressed in bone marrow. Mol Pharmacol. 2001a;59:420–426. doi: 10.1124/mol.59.3.420. [DOI] [PubMed] [Google Scholar]
  75. Liu C, Wilson SJ, Kuei C, Lovenberg TW. Comparison of human, mouse, rat, and guinea pig histamine H4 receptors reveals substantial pharmacological species variation. J Pharmacol Exp Ther. 2001b;299:121–130. [PubMed] [Google Scholar]
  76. Liu WL. Histamine H4 receptor antagonists for the treatment of inflammatory disorders. Drug Discov Today. 2014;19:1222–1225. doi: 10.1016/j.drudis.2014.05.007. [DOI] [PubMed] [Google Scholar]
  77. Marshall JK, Irvine EJ. Ketotifen treatment of active colitis in patients with 5-aminosalicylate intolerance. Can J Gastroenterol. 1998;12:273–275. doi: 10.1155/1998/398142. [DOI] [PubMed] [Google Scholar]
  78. Martinel Lamas DJ, Carabajal E, Prestifilippo JP, Rossi L, Elverdin JC, Merani S, et al. Protection of radiation-induced damage to the hematopoietic system, small intestine and salivary glands in rats by JNJ7777120 compound, a histamine H4 ligand. PLoS ONE. 2013;8:e69106. doi: 10.1371/journal.pone.0069106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Medina VA, Rivera ES. Histamine receptors and cancer pharmacology. Br J Pharmacol. 2010;161:755–767. doi: 10.1111/j.1476-5381.2010.00961.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Meng F, Han Y, Staloch D, Francis T, Stokes A, Francis H. The H4 histamine receptor agonist, clobenpropit, suppresses human cholangiocarcinoma progression by disruption of epithelial mesenchymal transition and tumor metastasis. Hepatology. 2011;54:1718–1728. doi: 10.1002/hep.24573. [DOI] [PubMed] [Google Scholar]
  81. Morini G, Becchi G, Shenton FC, Chazot PL, Grandi D. Histamine H3 and H4 receptors are expressed on distinct endocrine cell types in the rat fundic mucosa. Inflamm Res. 2008;57(Suppl. 1):S57–S58. doi: 10.1007/s00011-007-0628-9. [DOI] [PubMed] [Google Scholar]
  82. Morse KL, Behan J, Laz TM, West RE, Jr, Greenfeder SA, Anthes JC, et al. Cloning and characterization of a novel human histamine receptor. J Pharmacol Exp Ther. 2001;296:1058–1066. [PubMed] [Google Scholar]
  83. Mowbray CE, Bell AS, Clarke NP, Collins M, Jones RM, Lane CA, et al. Challenges of drug discovery in novel target space. The discovery and evaluation of PF-3893787: a novel histamine H4 receptor antagonist. Bioorg Med Chem Lett. 2011;21:6596–6602. doi: 10.1016/j.bmcl.2011.07.125. [DOI] [PubMed] [Google Scholar]
  84. Nakamura T, Itadani H, Hidaka Y, Ohta M, Tanaka K. Molecular cloning and characterization of a new human histamine receptor, HH4R. Biochem Biophys Res Commun. 2000;279:615–620. doi: 10.1006/bbrc.2000.4008. [DOI] [PubMed] [Google Scholar]
  85. Nguyen T, Shapiro DA, George SR, Setola V, Lee DK, Cheng R, et al. Discovery of a novel member of the histamine receptor family. Mol Pharmacol. 2001;59:427–433. doi: 10.1124/mol.59.3.427. [DOI] [PubMed] [Google Scholar]
  86. Nijmeijer S, Vischer HF, Sirci F, Schultes S, Engelhardt H, de Graaf C, et al. Detailed analysis of biased histamine H(4) receptor signalling by JNJ 7777120 analogues. Br J Pharmacol. 2013;170:78–88. doi: 10.1111/bph.12117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Oda T, Morikawa N, Saito Y, Masuho Y, Matsumoto S. Molecular cloning and characterization of a novel type of histamine receptor preferentially expressed in leukocytes. J Biol Chem. 2000;275:36781–36786. doi: 10.1074/jbc.M006480200. [DOI] [PubMed] [Google Scholar]
  88. Oda T, Matsumoto S, Masuho Y, Takasaki J, Matsumoto M, Kamohara M, et al. cDNA cloning and characterization of porcine histamine H4 receptor. Biochim Biophys Acta. 2002;1575:135–138. doi: 10.1016/s0167-4781(02)00236-1. [DOI] [PubMed] [Google Scholar]
  89. Oda T, Matsumoto S, Matsumoto M, Takasaki J, Kamohara M, Soga T, et al. Molecular cloning of monkey histamine H4 receptor. J Pharmacol Sci. 2005;98:319–322. doi: 10.1254/jphs.sc0050033. [DOI] [PubMed] [Google Scholar]
  90. Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. NC-IUPHAR. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledge base of drug targets and their ligands. Nucl. Acids Res. 2014;42:D1098–1106. doi: 10.1093/nar/gkt1143. (Database Issue): [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Poli E, Pozzoli C, Coruzzi G. Role of histamine H(3) receptors in the control of gastrointestinal motility. An overview. J Physiol (Paris) 2001;95:67–74. doi: 10.1016/s0928-4257(01)00010-9. [DOI] [PubMed] [Google Scholar]
  92. Pozzoli C, Adami M, Smits RA, Coruzzi G. Effect of histamine H4 receptor ligands on cholinergic neurotransmission of the rat duodenum. Inflamm Res. 2009;58(Suppl. 1):59–60. doi: 10.1007/s00011-009-2012-4. [DOI] [PubMed] [Google Scholar]
  93. Rajendra S, Mulcahy H, Patchett S, Kumar P. The effect of H2 antagonists on proliferation and apoptosis in human colorectal cancer cell lines. Dig Dis Sci. 2004;49:1634–1640. doi: 10.1023/b:ddas.0000043377.30075.ac. [DOI] [PubMed] [Google Scholar]
  94. Rosethorne EM, Charlton SJ. Agonist-biased signaling at the histamine H4 receptor: JNJ7777120 recruits beta-arrestin without activating G proteins. Mol Pharmacol. 2011;79:749–757. doi: 10.1124/mol.110.068395. [DOI] [PubMed] [Google Scholar]
  95. Salcedo C, Pontes C, Merlos M. Is the H4 receptor a new drug target for allergies and asthma? Front Biosci. 2013;5:178–187. doi: 10.2741/e606. [DOI] [PubMed] [Google Scholar]
  96. Sander LE, Lorentz A, Sellge G, Coeffier M, Neipp M, Veres T, et al. Selective expression of histamine receptors H1R, H2R, and H4R, but not H3R, in the human intestinal tract. Gut. 2006;55:498–504. doi: 10.1136/gut.2004.061762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Seifert R, Schneider EH, Dove S, Brunskole I, Neumann D, Strasser A, et al. Paradoxical stimulatory effects of the ‘standard’ histamine H4-receptor antagonist JNJ7777120: the H4 receptor joins the club of 7 transmembrane domain receptors exhibiting functional selectivity. Mol Pharmacol. 2011;79:631–638. doi: 10.1124/mol.111.071266. [DOI] [PubMed] [Google Scholar]
  98. Simon T, Laszlo V, Falus A. Impact of histamine on dendritic cell functions. Cell Biol Int. 2011;35:997–1000. doi: 10.1042/CBI20100844. [DOI] [PubMed] [Google Scholar]
  99. Simon T, Semsei AF, Ungvari I, Hadadi E, Virag V, Nagy A, et al. Asthma endophenotypes and polymorphisms in the histamine receptor HRH4 gene. Int Arch Allergy Immunol. 2012;159:109–120. doi: 10.1159/000335919. [DOI] [PubMed] [Google Scholar]
  100. Simons FE, Simons KJ. Histamine and H1-antihistamines: celebrating a century of progress. J Allergy Clin Immunol. 2011;128:1139–1150. doi: 10.1016/j.jaci.2011.09.005. e1134. [DOI] [PubMed] [Google Scholar]
  101. Singh M, Jadhav HR. Histamine H3 receptor function and ligands: recent developments. Mini Rev Med Chem. 2013;13:47–57. [PubMed] [Google Scholar]
  102. Smits RA, Leurs R, de Esch IJ. Major advances in the development of histamine H4 receptor ligands. Drug Discov Today. 2009;14:745–753. doi: 10.1016/j.drudis.2009.05.007. [DOI] [PubMed] [Google Scholar]
  103. Snyder SH, Epps L. Regulation of histidine decarboxylase in rat stomach by gastrin: the effect of inhibitors of protein synthesis. Mol Pharmacol. 1968;4:187–195. [PubMed] [Google Scholar]
  104. Sokol H, Georgin-Lavialle S, Grandpeix-Guyodo C, Canioni D, Barete S, Dubreuil P, et al. Gastrointestinal involvement and manifestations in systemic mastocytosis. Inflamm Bowel Dis. 2010;16:1247–1253. doi: 10.1002/ibd.21218. [DOI] [PubMed] [Google Scholar]
  105. Strakhova MI, Nikkel AL, Manelli AM, Hsieh GC, Esbenshade TA, Brioni JD, et al. Localization of histamine H4 receptors in the central nervous system of human and rat. Brain Res. 2009;1250:41–48. doi: 10.1016/j.brainres.2008.11.018. [DOI] [PubMed] [Google Scholar]
  106. Sutton TL, Zhao A, Madden KB, Elfrey JE, Tuft BA, Sullivan CA, et al. Anti-Inflammatory mechanisms of enteric Heligmosomoides polygyrus infection against trinitrobenzene sulfonic acid-induced colitis in a murine model. Infect Immun. 2008;76:4772–4782. doi: 10.1128/IAI.00744-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Takagaki K, Osawa S, Horio Y, Yamada T, Hamaya Y, Takayanagi Y, et al. Cytokine responses of intraepithelial lymphocytes are regulated by histamine H(2) receptor. J Gastroenterol. 2009;44:285–296. doi: 10.1007/s00535-009-0019-9. [DOI] [PubMed] [Google Scholar]
  108. Takeshita K, Sakai K, Bacon KB, Gantner F. Critical role of histamine H4 receptor in leukotriene B4 production and mast cell-dependent neutrophil recruitment induced by zymosan in vivo. J Pharmacol Exp Ther. 2003;307:1072–1078. doi: 10.1124/jpet.103.057489. [DOI] [PubMed] [Google Scholar]
  109. Thurmond RL, Desai PJ, Dunford PJ, Fung-Leung WP, Hofstra CL, Jiang W, et al. A potent and selective histamine H4 receptor antagonist with anti-inflammatory properties. J Pharmacol Exp Ther. 2004;309:404–413. doi: 10.1124/jpet.103.061754. [DOI] [PubMed] [Google Scholar]
  110. Thurmond RL, Chen B, Dunford PJ, Greenspan AJ, Karlsson L, La D, et al. Clinical and preclinical characterization of the histamine H4 receptor antagonist JNJ-39758979. J Pharmacol Exp Ther. 2014;349:176–184. doi: 10.1124/jpet.113.211714. [DOI] [PubMed] [Google Scholar]
  111. Togias A. H1-receptors: localization and role in airway physiology and in immune functions. J Allergy Clin Immunol. 2003;112(4 Suppl):S60–S68. doi: 10.1016/s0091-6749(03)01878-5. [DOI] [PubMed] [Google Scholar]
  112. Vanhala A, Yamatodani A, Panula P. Distribution of histamine-, 5-hydroxytryptamine-, and tyrosine hydroxylase-immunoreactive neurons and nerve fibers in developing rat brain. J Comp Neurol. 1994;347:101–114. doi: 10.1002/cne.903470108. [DOI] [PubMed] [Google Scholar]
  113. Varga C, Horvath K, Berko A, Thurmond RL, Dunford PJ, Whittle BJ. Inhibitory effects of histamine H4 receptor antagonists on experimental colitis in the rat. Eur J Pharmacol. 2005;522:130–138. doi: 10.1016/j.ejphar.2005.08.045. [DOI] [PubMed] [Google Scholar]
  114. Vermeulen W, De Man JG, Pelckmans PA, De Winter BY. Neuroanatomy of lower gastrointestinal pain disorders. World J Gastroenterol. 2014;20:1005–1020. doi: 10.3748/wjg.v20.i4.1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Wood JD. Enteric neuroimmunophysiology and pathophysiology. Gastroenterology. 2004;127:635–657. doi: 10.1053/j.gastro.2004.02.017. [DOI] [PubMed] [Google Scholar]
  116. Yu B, Shao Y, Zhang J, Dong XL, Liu WL, Yang H, et al. Polymorphisms in human histamine receptor H4 gene are associated with atopic dermatitis. Br J Dermatol. 2010;162:1038–1043. doi: 10.1111/j.1365-2133.2010.09675.x. [DOI] [PubMed] [Google Scholar]
  117. Yu S, Stahl E, Li Q, Ouyang A. Antigen inhalation induces mast cells and eosinophils infiltration in the guinea pig esophageal epithelium involving histamine-mediated pathway. Life Sci. 2008;82:324–330. doi: 10.1016/j.lfs.2007.12.002. [DOI] [PubMed] [Google Scholar]
  118. Zhang C, Xiong Y, Li J, Yang Y, Liu L, Wang W, et al. Deletion and down-regulation of HRH4 gene in gastric carcinomas: a potential correlation with tumor progression. PLoS ONE. 2012;7:e31207. doi: 10.1371/journal.pone.0031207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Zhu Y, Michalovich D, Wu H, Tan KB, Dytko GM, Mannan IJ, et al. Cloning, expression, and pharmacological characterization of a novel human histamine receptor. Mol Pharmacol. 2001;59:434–441. doi: 10.1124/mol.59.3.434. [DOI] [PubMed] [Google Scholar]
  120. Zimmermann AS, Burhenne H, Kaever V, Seifert R, Neumann D. Systematic analysis of histamine and N-methylhistamine concentrations in organs from two common laboratory mouse strains: C57Bl/6 and Balb/c. Inflamm Res. 2011;60:1153–1159. doi: 10.1007/s00011-011-0379-5. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES