ArticlePDF AvailableLiterature Review

Enigmatic Histamine Receptor H4 for Potential Treatment of Multiple Inflammatory, Autoimmune, and Related Diseases

Authors:
  • Venkateshwar Institute of Pharmacy

Abstract and Figures

The histamine H4 receptor, belonging to the family of G-protein coupled receptors, is an increasingly attractive drug target. It plays an indispensable role in many cellular pathways, and numerous H4R ligands are being studied for the treatment of several inflammatory, allergic, and autoimmune disorders, including pulmonary fibrosis. Activation of H4R is involved in cytokine production and mediates mast cell activation and eosinophil chemotaxis. The importance of this receptor has also been shown in inflammatory models: peritonitis, respiratory tract inflammation, colitis, osteoarthritis, and rheumatoid arthritis. Recent studies suggest that H4R acts as a modulator in cancer, neuropathic pain, vestibular disorders, and type-2 diabetes, however, its role is still not fully understood.
Content may be subject to copyright.
life
Review
Enigmatic Histamine Receptor H4for Potential
Treatment of Multiple Inflammatory, Autoimmune,
and Related Diseases
Pakhuri Mehta 1, Przemysław Miszta 1, Przemysław Rzodkiewicz 2, Olga Michalak 3,
Piotr Krzeczy ´nski 3and Sławomir Filipek 1, *
1Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 02-093 Warsaw,
Poland; pmehta@chem.uw.edu.pl or pakhurimehta@gmail.com (P.M.); pmiszta@chem.uw.edu.pl (P.M.)
2
Department of General and Experimental Pathology, Medical University of Warsaw, 02-091 Warsaw, Poland;
przemyslaw.rzodkiewicz@wum.edu.pl
3Łukasiewicz Research Network-Pharmaceutical Research Institute, 01-793 Warsaw, Poland;
o.michalak@ifarm.eu (O.M.); p.krzeczynski@ifarm.eu (P.K.)
*Correspondence: sfilipek@chem.uw.edu.pl
Received: 31 March 2020; Accepted: 20 April 2020; Published: 24 April 2020


Abstract:
The histamine H
4
receptor, belonging to the family of G-protein coupled receptors, is an
increasingly attractive drug target. It plays an indispensable role in many cellular pathways, and
numerous H
4
R ligands are being studied for the treatment of several inflammatory, allergic, and
autoimmune disorders, including pulmonary fibrosis. Activation of H
4
R is involved in cytokine
production and mediates mast cell activation and eosinophil chemotaxis. The importance of this
receptor has also been shown in inflammatory models: peritonitis, respiratory tract inflammation,
colitis, osteoarthritis, and rheumatoid arthritis. Recent studies suggest that H
4
R acts as a modulator
in cancer, neuropathic pain, vestibular disorders, and type-2 diabetes, however, its role is still not
fully understood.
Keywords:
histamine H
4
receptor; G protein-coupled receptors; allergic diseases; inflammatory
diseases; autoimmune disorders; neuropathic pain; cancer
1. Introduction
Histamine action via distinct receptors (H
1
R–H
4
R) modulates diverse physiological as well as
pathological processes. Due to their dierential receptor pharmacology and signal transduction
properties, histamine has characteristic eects dependent upon the histamine receptor subtype it
is bound to. Histamine receptors H
1
–H
4
are widespread throughout the body but there is limited
knowledge about the H
4
R. The role of H4R in neuropathic pain transmission and other diseases is
still controversial after nearly 20 years since its discovery. This may be due to biased signaling of
histamine and H
4
receptor agonists and dierential eects on multiple signaling pathways in central
and peripheral parts of the sensory nervous system. However, in the last two decades, there was
a particular increment in evidence supporting participation of H
3
R and H
4
R in neuropathic pain
modulation [
1
]. Histamine has also been identified to be responsible for a vascular type headache,
e.g., migraine, hence the antihistamines are regarded as a possible treatment [
2
]. The proper action of
particular subtypes of histamine receptors is of special importance as it has been shown for instance
for the delirium syndrome in which H
1
R and H
2
R antagonists have pro-delirium potential, while H
3
R
antagonists have proved to be beneficial in combating delirium. The H
4
R may also play an indirect
role requiring further intensive exploration [3].
Life 2020,10, 50; doi:10.3390/life10040050 www.mdpi.com/journal/life
Life 2020,10, 50 2 of 17
Pulmonary fibrosis is the most frequent form of interstitial lung disease. Unavailability of eective
therapies has led to the urge of exploiting novel curative approaches. Histamine receptor H
4
has
been recognized as a new target for inflammatory and immune diseases, and H
4
R ligands reduced
inflammation and oxidative stress in lung tissue. It has been shown that poly(ADP-ribose) polymerase
(PARP-1) and H
4
R are both involved in inflammatory and fibrotic responses. Treatment with H
4
R
antagonist JNJ7777120 ((5-chloro-1H-indol-2-yl)(4-methyl-1-piperazinyl)-methanone; CAS Number
459168-41-3; Molecular Weight: 277.8) in a condition of PARP-1 inhibition, provides anti-inflammatory
and anti-fibrotic eects, causing reduction in airway remodeling and bronchoconstriction. Its synergistic
eect with selective PARP-1 inhibitors could be of potential use for the treatment of pulmonary
fibrosis [
4
]. Viral infections can be important contributors to development of asthma and chronic
obstructive pulmonary disease. Pulmonary fibrosis is the main factor leading to pulmonary dysfunction
and quality of life decline in SARS survivors. Gaining a deeper understanding of the interaction
between Coronaviruses and the innate immune system of the host may shed light on the development
and persistence of inflammation in the lungs and can possibly reduce the risk of lung inflammation
caused by CoVs [5].
2. The Histamine Receptors—Localization and Function
Histamine receptors, numbered in the order of their discovery H
1
R-H
4
R, are G protein-coupled
receptors (GPCRs) that constitute the largest family of cell surface receptors in humans and play a key
role in cellular signaling. In the central nervous system (CNS), the histaminergic system is mainly
modulated by histamine, an inflammatory biogenic amine involved in wide range of pathophysiological
eects through interaction with histamine GPCRs which belong to class A (rhodopsin-like) GPCRs.
These GPCRs dier in localization and cellular signaling mechanisms and they even dier in the
level of constitutive activity, i.e., the ability to adopt an active conformation independent of ligand
binding [
6
,
7
]. H
1
R and H
2
R are found in the brain and periphery, H
3
R is abundant in the CNS, while
H
4
R has low expression, if any, in the CNS and is predominantly expressed on a variety of peripheral
immune cells such as eosinophils, dendritic cells, mast cells (HMC-1, LAD-2, and primary cord blood
derived CD34+human mast cells), leukocytes, and T-cells (including
γδ
T, T helper 1, 2, Th17, and CD8
cells) [
6
,
8
12
]. The presence and role of H
4
R in brain nervous tissue is yet elusive and not fully known
but the presence of H
4
R in non-neuronal cells in the brain has been confirmed [
13
,
14
]. Functional
H
4
receptors that increase [
35
S]-GTP
γ
S binding and/or decrease noradrenaline release have not been
identified in human, guinea pig, and mouse cortex [
15
]. In human mast cells, H
4
R mediates release of
cytokines, leukotrienes, and chemokines (TGF-
β
1, TNF-
α
, TNF-
β
, PDGF-BB, TIMP-2, M-CSF, IP-10,
IL-16, IL-6, IL-3, IL-10, MIP-1α, IL-1α, ICAM-1, Eotaxin-2, RANTES, IL-8, MCP-1, and IL-6sR) [10].
Being a member of the most populated class A of the GPCR superfamily, human H
4
R also
contains seven transmembrane helices and a short amphipathic helix that possibly runs parallel to
the cytosolic membrane surface. It consists of 390 amino acid residues possessing all of the highly
conserved sequence motifs [
16
,
17
] of the class A GPCRs including the most evolutionary conserved
residues in each of the transmembrane helices: N1.50, D2.50, R3.50, W4.50, P5.50, P6.50, and P7.50
(Ballesteros–Weinstein numbering [
18
]) indicating the same activation mechanism of H
4
R as that of
the other receptors in class A GPCRs [
19
]. The Ballesteros–Weinstein numbering scheme of GPCRs
provides information about the relative positions of amino acids present in seven transmembrane
helices. Each residue of the receptor is recognized by two numbers separated by a dot; the first number
(1–7) indicates the number of the transmembrane helix where the residue is located while the second
number indicates its position in relation to the most conserved residue, assigned number 50, of the
same helix. The prominent residues such as D3.32 and W7.40, specific for amine-activated GPCRs, are
also present in the H
4
R [
20
]. It has been observed that the two agonists (histamine and OUP-16) exhibit
complementary interactions with residues D3.32, E5.46, and T6.55, while the reference antagonist
JNJ7777120 exhibits interactions with D3.32 and E5.46 only (Figure 1), implicating a dierentiating role
of T6.55 in ligand binding and receptor activation [
21
,
22
]. There are also striking complementarities
Life 2020,10, 50 3 of 17
between the H
4
R binding pocket and the structural properties of most H
4
R antagonists. They consist
of a minimum of one, or preferably two, positively charged groups complementary to two negatively
charged residues in the binding pocket, namely D3.32 and E5.46, and such double interaction is crucial
for the interaction of high anity ligands with H4R [21].
Life 2020, 10, 50 3 of 17
complementary to two negatively charged residues in the binding pocket, namely D3.32 and E5.46,
and such double interaction is crucial for the interaction of high affinity ligands with H4R [21].
Figure 1. The homology model of H4R with docked JNJ7777120 antagonist. The specific ligand–
receptor interactions are shown on the right panel. D3.32 forms both a hydrogen bond and an ionic
interaction with the charged amine group of the ligand.
Among the histamine receptors, H1R and H4R possess 40% amino acid identity in the
transmembrane region and they recognize the same endogenous ligand that is histamine. Due to such
similarity the crystal structure of H1R has been used by many researchers for building the homology
models of H4R. However, there are substantial differences in histamine receptor binding sites. For
instance, N4.57 in H4R is equivalent to W4.56, L5.39 to K5.39, E5.46 to N5.46, and Q7.42 to G7.42 in
H1R. Additionally, the mutations of residues N4.57 and E5.46 resulted in significant alteration of
inhibition constants of JNJ7777120 which was the first reported H4R antagonist [23] and the homology
model of H4R featured two specific hydrogen bonds and ionic interactions of JNJ7777120 to D3.32
and E5.46 [24]. H4R has the highest sequence homology with H3R as it possesses 37% amino acid
identity in protein sequence and 58% identity in the transmembrane region. It is evident that a
number of ligands of H4R also have a high affinity for H3R due to the identical amino acids within
the binding site of both receptors, including E5.46, Y3.33, and Y6.51, involved in ligand binding [25].
These amino acids residues contribute to the similarity between the binding sites of hH3R and hH4R
forcing similar conformations of ligands. This explains the number of ligands which are antagonists
of both receptors. Additionally, various substituted histamine derivatives such as R-(α)-
methylhistamine have significant H4R binding in addition to H3R [6]. Istyastono et al. have shown
that the E5.46Q mutation impaired the binding strength of clobenpropit and its derivatives in both
those receptors [26]. Moreover, the L5.39V and E5.46Q mutations resulted in a decrease of binding of
the reported ligands to H4R. This finding emphasized the importance of the E5.46 residue which
provides a crucial interaction with antagonists [27].
A plethora of studies have related the heterogenic and complex pharmacology of histamine
receptors to various diseases: H1R to the allergic inflammation, anaphylaxis, and motion sickness
[28,29], H2R to the stimulation of gastric acid secretion leading to peptic ulcer, GERD and aspiration
pneumonitis [30,31], H3R to the neurotransmission controlling sleep, cognitive processes,
schizophrenia, epilepsy, and pain [32–37], and H4R to the immune responses (cancers, myocarditis)
and inflammation [38–42] (Figure 2). The H3 and H4 receptors have relatively high affinity for
Figure 1.
The homology model of H
4
R with docked JNJ7777120 antagonist. The specific ligand–receptor
interactions are shown on the right panel. D3.32 forms both a hydrogen bond and an ionic interaction
with the charged amine group of the ligand.
Among the histamine receptors, H
1
R and H
4
R possess 40% amino acid identity in the
transmembrane region and they recognize the same endogenous ligand that is histamine. Due
to such similarity the crystal structure of H
1
R has been used by many researchers for building the
homology models of H
4
R. However, there are substantial dierences in histamine receptor binding sites.
For instance, N4.57 in H
4
R is equivalent to W4.56, L5.39 to K5.39, E5.46 to N5.46, and Q7.42 to G7.42
in H
1
R. Additionally, the mutations of residues N4.57 and E5.46 resulted in significant alteration of
inhibition constants of JNJ7777120 which was the first reported H
4
R antagonist [
23
] and the homology
model of H
4
R featured two specific hydrogen bonds and ionic interactions of JNJ7777120 to D3.32 and
E5.46 [
24
]. H
4
R has the highest sequence homology with H
3
R as it possesses 37% amino acid identity
in protein sequence and 58% identity in the transmembrane region. It is evident that a number of
ligands of H
4
R also have a high anity for H
3
R due to the identical amino acids within the binding
site of both receptors, including E5.46, Y3.33, and Y6.51, involved in ligand binding [
25
]. These amino
acids residues contribute to the similarity between the binding sites of hH
3
R and hH
4
R forcing similar
conformations of ligands. This explains the number of ligands which are antagonists of both receptors.
Additionally, various substituted histamine derivatives such as R-(
α
)-methylhistamine have significant
H
4
R binding in addition to H
3
R [
6
]. Istyastono et al. have shown that the E5.46Q mutation impaired
the binding strength of clobenpropit and its derivatives in both those receptors [
26
]. Moreover, the
L5.39V and E5.46Q mutations resulted in a decrease of binding of the reported ligands to H
4
R. This
finding emphasized the importance of the E5.46 residue which provides a crucial interaction with
antagonists [27].
A plethora of studies have related the heterogenic and complex pharmacology of histamine
receptors to various diseases: H
1
R to the allergic inflammation, anaphylaxis, and motion
Life 2020,10, 50 4 of 17
sickness [
28
,
29
], H
2
R to the stimulation of gastric acid secretion leading to peptic ulcer, GERD and
aspiration pneumonitis [
30
,
31
], H
3
R to the neurotransmission controlling sleep, cognitive processes,
schizophrenia, epilepsy, and pain [
32
37
], and H
4
R to the immune responses (cancers, myocarditis) and
inflammation [
38
42
] (Figure 2). The H
3
and H
4
receptors have relatively high anity for histamine
(5–10 nM) compared to the low anity of H
1
R and H
2
R which is in the
µ
M range [
6
,
43
]. Hence,
the biological response has been linked directly with the local tissue histamine concentration and
functional expression of dierent receptors [6].
Life 2020, 10, 50 4 of 17
histamine (5–10 nM) compared to the low affinity of H1R and H2R which is in the μM range [6,43].
Hence, the biological response has been linked directly with the local tissue histamine concentration
and functional expression of different receptors [6].
Figure 2. Classification of histamine receptors (H1R–H4R) in relation to their functions. H1R–H3R
transduce extracellular signals via q/11, Gαis, and Gαi/o, respectively, while H4R acts through Gαi/o
and β-arrestin. H1R and H2R are low-affinity receptors while H3R and H4R are high-affinity receptors
towards histamine. Ligands of H1R–H4R have therapeutic applications in allergic inflammation,
gastric acid secretion, neurotransmission, and immunomodulation, respectively. The information in
the figure is partially based on [44].
3. Species Differences of H4R
Following the identification of the human H4R (UniProt id: Q9H3N8), various sequences of
mouse, rat, guinea pig, pig, dog, and monkey H4R have been reported and functionally expressed
[38]. Eighty-five protein sequences of H4R orthologues from different species have been extracted
from the UniProt database and aligned to draw the phylogenetic relationship between H4R
orthologues (Scheme 1). The H4 receptors of the chimpanzee, gorilla, and orangutan show the highest
sequence homology (98–99%) with the human orthologue (hH4R). H4 receptors of some species are
highly homologous to hH4R with sequence homology between 78% and 94%, specifically those of
macaques, baboon, drill, Angolan colobus, mangabey, Cebus capucinus imitator, marmoset, and
Philippine tarsier (Table 1). Orthologues in some species were only moderately homologous to hH4R
with sequence homology between 54% and 73% while the least homologous showed homology
ranging from 10% to 47%. Pig, mouse, smooth cauliflower coral, Japanese scallop, turbot, and pig
have each two H4R orthologues while sea cucumber has three orthologues. However, these
orthologues, show only 10–36% homology to hH4R while all others show a substantially higher
homology (>50%). As some of the sequences are still incomplete, changes in the phylogenetic tree are
to be expected. Within these GPCR sequences, the typical aminergic GPCR features (D3.32 in TM3
and E5.46 in TM5) can often be found. Detailed analysis of most of these species variants is however
lacking even though it could provide useful tools to dissect receptor–ligand binding. Using site-
directed mutagenesis Wifling et al. have proved that the F169, located in the second extracellular loop
ECL2, is a crucial amino acid for differential interactions, affinities, and potencies of certain agonists
with the human and mouse H4R orthologues [45]. Receptor sequence differences have implications
even for ligand function as the JNJ7777120 ligand acts as a partial inverse agonist at the human H4R,
but as a partial agonist at the rat and mouse H4R which possess lower constitutive activity than their
Figure 2.
Classification of histamine receptors (H
1
R–H
4
R) in relation to their functions. H
1
R–H
3
R
transduce extracellular signals via G
αq/11
, G
αis
, and G
αi/o
, respectively, while H
4
R acts through G
αi/o
and
β
-arrestin. H
1
R and H
2
R are low-anity receptors while H
3
R and H
4
R are high-anity receptors
towards histamine. Ligands of H
1
R–H
4
R have therapeutic applications in allergic inflammation, gastric
acid secretion, neurotransmission, and immunomodulation, respectively. The information in the figure
is partially based on [44].
3. Species Dierences of H4R
Following the identification of the human H
4
R (UniProt id: Q9H3N8), various sequences of
mouse, rat, guinea pig, pig, dog, and monkey H
4
R have been reported and functionally expressed [
38
].
Eighty-five protein sequences of H
4
R orthologues from dierent species have been extracted from
the UniProt database and aligned to draw the phylogenetic relationship between H
4
R orthologues
(Scheme 1). The H
4
receptors of the chimpanzee, gorilla, and orangutan show the highest sequence
homology (98–99%) with the human orthologue (hH
4
R). H
4
receptors of some species are highly
homologous to hH
4
R with sequence homology between 78% and 94%, specifically those of macaques,
baboon, drill, Angolan colobus, mangabey, Cebus capucinus imitator, marmoset, and Philippine tarsier
(Table 1). Orthologues in some species were only moderately homologous to hH
4
R with sequence
homology between 54% and 73% while the least homologous showed homology ranging from 10%
to 47%. Pig, mouse, smooth cauliflower coral, Japanese scallop, turbot, and pig have each two H
4
R
orthologues while sea cucumber has three orthologues. However, these orthologues, show only
10–36% homology to hH
4
R while all others show a substantially higher homology (>50%). As some of
the sequences are still incomplete, changes in the phylogenetic tree are to be expected. Within these
GPCR sequences, the typical aminergic GPCR features (D3.32 in TM3 and E5.46 in TM5) can often be
Life 2020,10, 50 5 of 17
found. Detailed analysis of most of these species variants is however lacking even though it could
provide useful tools to dissect receptor–ligand binding. Using site-directed mutagenesis Wifling et
al. have proved that the F169, located in the second extracellular loop ECL2, is a crucial amino acid
for dierential interactions, anities, and potencies of certain agonists with the human and mouse
H
4
R orthologues [
45
]. Receptor sequence dierences have implications even for ligand function as
the JNJ7777120 ligand acts as a partial inverse agonist at the human H
4
R, but as a partial agonist
at the rat and mouse H
4
R which possess lower constitutive activity than their human counterpart.
Therefore, dierences in pharmacological activities of H
4
R ligands between dierent species might
hamper preclinical development of future H4R drugs [46].
Life 2020, 10, 50 5 of 17
human counterpart. Therefore, differences in pharmacological activities of H4R ligands between
different species might hamper preclinical development of future H4R drugs [46].
Scheme 1. Phylogenetic tree of H4R orthologues. The sequences were obtained from UniProt [47] and
the sequences were aligned with ClustalW and the cladogram was created with Clustal Omega
service [48].
Table 1. Sequence similarities of species specific H4R to the human orthologue.
Species
Scientific Name
UniProt ID
Similarity to hH
4
R
1
Human
Homo sapiens
Q9H3N8
-
2
Chimpanzee
Pan troglodytes
H2QED2
99%
3
Gorilla
Gorilla
G3QS38
98%
4
Pygmy chimpanzee
Pan paniscus
A0A2R9BQY6
98%
5
Orangutan
Pongo abelii
H2NW27
98%
6
Crab
-
eating macaque
Macaca fascicularis
Q3V8G8
94%
7
Pig
-
tailed
macaque
Macaca nemestrina
A0A2K6D1G7
94%
8
Rhesus macaque
Macaca mulatta
G7NKH9
94%
9
Olive baboon
Papio anubis
A0A096NGN9
94%
Drill
Mandrillus leucophaeus
A0A2K5YBZ5
94%
Scheme 1.
Phylogenetic tree of H
4
R orthologues. The sequences were obtained from UniProt [
47
]
and the sequences were aligned with ClustalW and the cladogram was created with Clustal Omega
service [48].
Life 2020,10, 50 6 of 17
Table 1. Sequence similarities of species specific H4R to the human orthologue.
Species Scientific Name UniProt ID Similarity to hH4R
1
Human Homo sapiens Q9H3N8 -
2
Chimpanzee Pan troglodytes H2QED2 99%
3
Gorilla Gorilla G3QS38 98%
4
Pygmy chimpanzee Pan paniscus A0A2R9BQY6 98%
5
Orangutan Pongo abelii H2NW27 98%
6
Crab-eating macaque Macaca fascicularis Q3V8G8 94%
7
Pig-tailed macaque Macaca nemestrina A0A2K6D1G7 94%
8
Rhesus macaque Macaca mulatta G7NKH9 94%
9
Olive baboon Papio anubis A0A096NGN9 94%
10
Drill Mandrillus leucophaeus A0A2K5YBZ5 94%
11
Angolan colobus Colobus angolensis palliatus A0A2K5HHL6 93%
12
Sooty mangabey Cercocebus atys A0A2K5LQL7 93%
13
Black snub-based monkey Rhinopithecus bieti A0A2K6MXG3 93%
14
Golden snub-based monkey Rhinopithecus roxellana A0A2K6RWF0 93%
15
Green monkey Chlorocebus sabaeus A0A0D9RYY4 90%
16
Ma’s Night monkey Aotus nancymaae A0A2K5CHI5 90%
17
Cebus capucinus imitator Cebus capucinus imitator A0A2K5RKQ4 90%
18
White-tufted-ear marmoset Callithrix jacchus F7IT43 89%
19
Squirrel monkey Saimiri boliviensis A0A2K6TG45 88%
20
Philippine tarsier Tarsius syrichta A0A1U7UM57 78%
21
Small-eared galago Otolemur garnettii H0WYC8 73%
22
Thirteen-lined ground squirrel Ictidomys tridecemlineatus I3MG71 72%
23
Dog Canis lupus familiaris J9P1C3 71%
24
Golden hamster Mesocricetus auratus A0A1U7Q7T1 71%
25
Grizzly bear Ursus arctos horribilis A0A3Q7WBT8 70%
26
Polar bear Ursus maritimus A0A384C2G0 70%
27
Pig Sus scrofa Q8WNV9 (Pig 1) 70%
28
A0A5G2QV28 (Pig 2) 10%
29
Red fox Vulpes vulpes A0A3Q7SYT7 70%
30
Black flying fox Pteropus alecto L5K5C7 69%
31
African elephant Loxodonta africana G3STF1 69%
32
Giant panda Ailuropoda melanoleuca G1M6D3 69%
33
Chinese hamster Cricetulus griseus A0A3L7I1V9 69%
34
Horse Equus caballus F6Z8L3 69%
35
Sea cow Trichechus manatus latirostris A0A2Y9E7N3 69%
36
Rabbit Oryctolagus cuniculus G1TKW6 68%
37
Iberian lynx Lynx pardinus A0A485N8M7 68%
38
Cat Felis catus M3WE71 68%
39
Pacific walrus Odobenus rosmarus divergens A0A2U3WW63 68%
40
Rat Rattus norvegicus Q91ZY1 68%
41
Kangaroo rat Dipodomys ordii A0A1S3F272 68%
42
Hawaiian monk seal Neomonachus schauinslandi A0A2Y9GRV4 68%
43
Northern fur seal Callorhinus ursinus A0A3Q7Q9W4 67%
44
Sea otter Enhydra lutris kenyoni A0A2Y9ITU9 67%
45
Hedgehog Erinaceus europaeus A0A1S3A2Y6 67%
46
European domestic ferret Mustela putorius furo M3Y4H4 67%
47
Mouse Mus musculus Q91ZY2 (Mouse 1) 67%
48
B2ZGH2 (Mouse 2) 66%
49
Goat Capra hircus A0A452DKI0 65%
50
Sheep Ovis aries W5PBL0 65%
51
Sperm whale Physeter macrocephalus A0A2Y9F727 65%
52
Hybrid cattle Bos indicus*Bos taurus A0A4W2DVG0 64%
53
Yak Bos mutus L8IEJ5 64%
54
Bovine Bos taurus E1BBS2 64%
55
Guinea pig Cavia porcellus Q91ZY3 63%
56
Black bear Ursus americanus A0A452QKW6 62%
57
Yangtze river dolphin Lipotes vexillifer A0A340YGS9 61%
58
American mink Neovison vison U6CNR7 61%
59
Beluga whale Delphinapterus leucas A0A2Y9PB56 59%
60
Yangtze finless porpoise Neophocaena asiaeorientalis A0A341CIF8 59%
61
European red deer Cervus elaphus hippelaphus A0A212C702 59%
62
Indo-pacific humpbacked dolphin
Sousa chinensis A0A484GQ08 57%
63
Narwhal Monodon monoceros A0A4U1FGC1 56%
64
Wolverine Gulo gulo A0A3P4RYS2 55%
65
Atlantic bottle-nosed dolphin Tursiops truncatus A0A2U3V3K5 54%
66
Gray short-tailed opossum Monodelphis domestica F6QB56 47%
67
North-Pacific minke whale Balaenoptera acutorostrata scammoni A0A452C640 46%
68
Tasmanian devil Sarcophilus harrisii G3X3P1 45%
69
Weddell seal Leptonychotes weddellii A0A2U3YB28 42%
70
White-tailed sea-eagle Haliaeetus albicilla A0A091PX74 42%
71
Trogon Apaloderma vittatum A0A091NQC4 41%
72
Cuckoo Cuculus canorus A0A091G9T7 40%
73
Turbot Scophthalmus maximus A0A2U9BJT1 (Turbot 1) 36%
74
A0A2U9C3Q1 (Turbot 2) 36%
Life 2020,10, 50 7 of 17
Table 1. Cont.
Species Scientific Name UniProt ID Similarity to hH4R
75
Channel catfish Ictalurus punctatus A0A2D0RQW6 36%
76
Chinese tree shrew Tupaia chinensis L8YD15 35%
77
Rifleman Acanthisitta chloris A0A091MN56 31%
78
Scallop Mizuhopecten yessoensis A0A210PRL2 (Scallop 1) 26%
79
A0A210PS14 (Scallop 2) 22%
80
Oyster Crassostrea gigas K1PU39 24%
81
Coral Stylophora pistillata A0A2B4RTL0 (Coral 1) 17%
82
A0A2B4RX53 (Coral 2) 14%
83
Sea cucumber Stichopus japonicus
A0A2G8KHM7
(Sea cucumber 1) 15%
84
A0A2G8L2L5
(Sea cucumber 2) 13%
85
A0A2G8JXR8
(Sea cucumber 3) 20%
4. The Pharmacological Eects of H4R Ligands
Although the pharmacology of H
4
R ligands is yet not fully elucidated H
4
R has been widely studied
and reviewed since its characterization and cloning in 2000 [
25
,
49
]. The vast body of accumulating
knowledge on physiological and pathophysiological functions associated with H
4
R modulation can be
exploited for therapeutic purposes [
11
]. The properties of H
4
R make this amine receptor and its ligands
of interest to specialists in the field of allergology, neurobiology, gastroenterology, endocrinology, and
also to researchers of cardiovascular functions [
6
,
50
]. The results of research on the role of H
4
R in
various pathophysiological and immunological processes indicate its association with the development
and course of many diseases including a crucial role of H
4
R in airway and dermal inflammation
(Figure 3), pruritus, ocular inflammation, arthritis, systemic lupus erythematosus, Sjogren’s syndrome,
multiple sclerosis, gastric ulcer, cancer, and pain [12,51].
Life 2020, 10, 50 7 of 17
75
Channel catfish
Ictalurus punctatus
A0A2D0RQW6
36%
76
Chinese tree shrew
Tupaia chinensis
L8YD15
35%
77
Rifleman
Acanthisitta chloris
A0A091MN56
31%
78
Scallop Mizuhopecten yessoensis
A0A210PRL2 (Scallop 1)
26%
79
A0A210PS14 (Scallop 2)
22%
80
Oyster
Crassostrea gigas
K1PU39
24%
81
Coral Stylophora pistillata
A0A2B4RTL0 (Coral 1)
17%
82
A0A2B4RX53 (Coral 2)
14%
83
Sea cucumber Stichopus japonicus
A0A2G8KHM7
(Sea cucumber 1) 15%
84
A0A2G8L2L5
(Sea cucumber 2) 13%
85
A0A2G8JXR8
(Sea cucumber 3) 20%
4. The Pharmacological Effects of H4R Ligands
Although the pharmacology of H4R ligands is yet not fully elucidated H4R has been widely
studied and reviewed since its characterization and cloning in 2000 [25,49]. The vast body of
accumulating knowledge on physiological and pathophysiological functions associated with H4R
modulation can be exploited for therapeutic purposes [11]. The properties of H4R make this amine
receptor and its ligands of interest to specialists in the field of allergology, neurobiology,
gastroenterology, endocrinology, and also to researchers of cardiovascular functions [6,50]. The
results of research on the role of H4R in various pathophysiological and immunological processes
indicate its association with the development and course of many diseases including a crucial role of
H4R in airway and dermal inflammation (Figure 3), pruritus, ocular inflammation, arthritis, systemic
lupus erythematosus, Sjogren’s syndrome, multiple sclerosis, gastric ulcer, cancer, and pain [12,51].
Figure 3. Potential role of histamine and histamine H4R-induced recruitment of eosinophils and mast
cells in chronic allergic inflammation. Histamine has been known to be a major mediator of
inflammation. Histamine H4 receptors are expressed on the surface of both eosinophils and mast cells.
Allergen may crosslink immunoglobulin E (IgE) on mast cells to release histamine, lipid mediators,
and cytokines. Antigen is also processed by dendritic cells and macrophages for presentation to T-
helper cells. During this process a local release of histamine and cytokines may occur. Histamine can
act on a variety of cells and at different levels. In asthma histamine can facilitate the recruitment of
inflammatory cells by regulating the chemotaxis of additional dendritic cells, eosinophils, and mast
cells to the airways via the action at H4R. Histamine may additionally affect cytokine release from
CD8+ cells via binding to H4R and from eosinophils, neutrophils, and mast cells through multiple
histamine receptors.
Figure 3.
Potential role of histamine and histamine H
4
R-induced recruitment of eosinophils and
mast cells in chronic allergic inflammation. Histamine has been known to be a major mediator of
inflammation. Histamine H
4
receptors are expressed on the surface of both eosinophils and mast cells.
Allergen may crosslink immunoglobulin E (IgE) on mast cells to release histamine, lipid mediators,
and cytokines. Antigen is also processed by dendritic cells and macrophages for presentation to
T-helper cells. During this process a local release of histamine and cytokines may occur. Histamine can
act on a variety of cells and at dierent levels. In asthma histamine can facilitate the recruitment of
inflammatory cells by regulating the chemotaxis of additional dendritic cells, eosinophils, and mast
cells to the airways via the action at H
4
R. Histamine may additionally aect cytokine release from
CD8
+
cells via binding to H
4
R and from eosinophils, neutrophils, and mast cells through multiple
histamine receptors.
Life 2020,10, 50 8 of 17
4.1. Allergic Diseases
Inflammatory conditions were for a long time thought to be mediated by activation of the histamine
receptor subtype 1. However, the discovery and pharmacological characterization of H
4
R ligands
especially antagonists, (and, to a lesser extent H
3
R and even H
2
R ligands) on mast cells, eosinophils,
and T cells demonstrates the possibility of its involvement in inflammatory conditions/symptoms
such as atopic dermatitis (AD), asthma, allergic rhinitis, rheumatoid arthritis (RA), and pruritus in
humans. This is evident from the results obtained in diverse experimental models of inflammation
including hepatic ischemia-reperfusion, colitis, atopic dermatitis, in which H
4
R antagonists (JNJ7777120,
JNJ10191584, thioperamide) proved to be ecient anti-inflammatory agents with reduced neutrophil
recruitment and release of cytokines [
51
,
52
]. Preclinical and clinical data strongly suggest the regulatory
involvement of H
4
R in the calcium influx and cellular chemotaxis [
53
,
54
], hence establishing a link
between the potential therapeutic application of selectively acting H
4
R ligands to inflammatory
conditions while also indicating involvement of H
4
R in diseases accompanied by itch and pain [
55
].
The investigations of histamine in the inflammation process have led to a development of the first
highly potent and selective non-imidazole H
4
R antagonist JNJ7777120, followed by reexamination and
synthesis of a plethora of H4R-targeted compounds [50,51].
Currently, many H
4
R ligands are known, synthesized, and evaluated [
56
,
57
]. Studies using
selective H
4
R ligands in animal models of pruritus revealed a role for H
4
R in mediating chronic
pruritus associated with conditions such as atopic dermatitis [
51
,
58
]. Antagonists of H
4
R (JNJ7777120,
JNJ39758979, INCB38579, and others) reduced pruritus in a number of animal studies [
59
] as well as
itching sensation in dierent conditions in human patients. Alcaftadine, a topical ophthalmic drug
indicated for the prevention of itching associated with allergic conjunctivitis, is a potent H
1
R and
H
2
R antagonist (in fact, inverse agonist) with weak inverse agonistic activity also towards H
4
R [
60
].
Administration of H
1
R/H
4
R antagonists or co-administration of H
1
R and H
4
R antagonists will probably
be eective also in humans. Such antagonists are more ecacious as compared to olopatadine (H
1
R
antagonist without H
4
R activity) [
61
]. Consequently, these studies indicate that H
4
R is involved in
mediating pruritic responses in humans, and that H
4
R antagonists are ought to be eective in the
treatment of pruritic histamine-mediated conditions, such as AD, acute urticaria, allergic rhinitis, or
allergic conjunctivitis.
The histamine receptor H
4
R was also found on cartilage cells–chondrocytes [
62
,
63
]. As the
presence of the histamine triggering protein (HRF) has been identified in the joints of people with
RA, it seems very likely that H
4
R antagonists will be used in the future in the treatment of RA [
64
].
This receptor may also be important in the pathogenesis of Sjörgen’s syndrome, erythematous lupus
erythematosus, and atopic dermatitis [
65
]. H
4
R activation not only results in phosphorylation of
ERK and PI3K in a time dependent manner but it also leads to enhanced synthesis of inflammatory
mediators associated with allergic reactions. It leads to inflammatory conditions as well as contributes
to postinflammatory visceral hypersensitivity, thus, making H
4
R antagonists important for reducing
inflammation and normalizing postinflammatory visceral hypersensitivity [66].
4.2. Asthma
H
4
R seems to be an interesting pharmacological target in the treatment of asthma [
6
]. Asthma is
a condition typically characterized by involvement of eosinophils and mast cells [
67
69
]. Extensive
studies have provided evidence detailing the functional profile of H
4
R in eosinophil biology [
70
]
and in the chemotaxis and dierentiation of other immune cell types. In experiments carried out
on animal models of inflammation of the airways, it was observed that in mice lacking the H
4
R
gene, there was a significant reduction in the allergic reaction caused by the administration of
a chicken protein-ovalbumin [
71
]. Chemotaxis of eosinophils was shown to be blocked by H
4
R
selective antagonists (JNJ7777120, JNJ39758979, or JNJ10191584) in animal asthma models due to
priming and T cell activation [
51
,
72
] while induced by histamine and selective H
4
R agonists (e.g.,
4-methylhistamine) [
72
]. Some selective H
4
R antagonists in animal models of asthma proved beneficial
Life 2020,10, 50 9 of 17
by mediating lung function and inflammation [
51
,
73
]. In asthma animal models, H
4
R antagonists act
either directly by reducing the number of T cells at the site of inflammation [
74
] or indirectly when it is
involved in dendritic cell function driving the response [
51
]. However, none of the H
4
R antagonists
have been introduced to treat the above disorders.
4.3. Diabetes
The histamine receptor H
4
may also be a therapeutic target in diseases not directly related
to inflammation. For instance, H
4
R is suggested to be important in the pathogenesis of diabetes.
In streptozotocin-induced diabetic rats H
4
R is overexpressed in tubular epithelial cells [
75
], and
administration of a H
4
R antagonist resulted in a decreased blood sugar [
76
]. H
4
R participates in
diabetic nephropathy progression through both a direct eect on tubular reabsorption and an indirect
action on renal tissue architecture via inflammatory cell recruitment. Therefore, H
4
R antagonism
emerges as a possible new multi-mechanism therapeutic approach to counteract development of
diabetic nephropathy [77].
4.4. Parkinson’s and Alzheimer’s Diseases
Evidence about the H
4
R antagonist JNJ7777120 inhibiting propagation of microglial inflammation
by attenuating the release of M1 microglial cells and largely preventing the pathological progression of
Parkinson’s disease-like pathology and motor dysfunction has been provided by the latest research [
78
].
These findings support H
4
R as a promising novel therapeutic target for Parkinson’s disease. For
Alzheimer’s disease the precise mechanism of histamine-induced Alzheimer ’s pathology is not well
known although the increased levels of histamine in plasma and in some areas of the brain are seen in
Alzheimer’s dementia brain [
79
]. It is known that H
3
R can regulate cognitive and memory functions
in the hippocampus so it could be involved in Alzheimer’s pathology [
80
]. Since H
4
R is also present
in the brain and its stimulation regulates neuronal functions, then stimulating H
4
receptors may
have some beneficial eects in the brain of Alzheimer’s disease patients. Recently, it has been found
that clobenpropit, a selective H
3
R antagonist with partial H
4
R agonist property, caused a significant
reduction in amyloid-
β
deposits in a rat model of Alzheimer-like brain pathology. This eect was
accompanied by marked reduction in neuronal or glial reactions so such dual-action compounds may
have neuroprotective properties [81].
High similarity between H
3
R and H
4
R entails considerable similarity in ligand anities and
facilitates simultaneous activation of both receptors. Dual-acting H
3
R/H
4
R ligands may exhibit
therapeutic potential in diverse pathological conditions, such as neuropathic pain, cancer, Parkinson’s,
and inflammatory diseases [
7
,
82
]. Dual H
3
R/H
4
R imidazole containing ligands used so far includes
compounds such as imetit, immepip, clobenpropit, and thioperamide [7].
4.5. Autoimmune Diseases
The characterization of a histamine receptor H
4
R with putative immunomodulating properties
encouraged new hopes for the translational exploitation of this new therapeutic target for the still
unmet medical needs, specifically asthma, autoimmune diseases, and a host defense. Rheumatoid
arthritis (RA), which is a systemic autoimmune disorder, is characterized by chronic synovitis of
peripheral joints, cartilage and bone destruction followed by joint disability. It was found that histamine
and Th17 cytokines induced osteoclast dierentiation from monocytes and JNJ7777120 decreased the
osteoclastogenesis and the osteoclastogenic role of H
4
R has been evident in patients with RA [
83
].
Studies in the animal model of RA have shown that the H
4
R antagonist JNJ7777120 reduces the
degree and severity of joint damage and reduces the number of cells producing IL-17 in the joint,
thus, significantly inhibiting the inflammatory process in joints [
84
]. H
4
R involvement has been also
confirmed in several types of cancers: melanoma [
85
], breast cancer [
86
], pancreatic cancer [
87
], and
colorectal cancer [
88
]. H
4
R can regulate the aging and apoptosis of cancer cells and blocking H
4
R by
antagonists inhibits tumor cell proliferation [
86
]. Histamine receptors play also an important role in
Life 2020,10, 50 10 of 17
the pathogenesis of multiple sclerosis. It turned out that H
1
R and H
2
R play a propathogenic role while
H3R and H4R may reduce the risk of the disease [89].
5. Clinical Trials of Drug Candidates Targeting H4R
Recently, H
4
R research has been gaining a lot of importance and the clinical studies were initiated
for the putative therapeutic exploitation in inflammatory and allergic disorders [
38
] such as atopic
dermatitis (AD) [
59
,
90
], pruritus, asthma, rheumatoid arthritis (RA), as well as in vestibular disease
(Table 2) [
91
]. Toreforant (JNJ38518168), the first oral H
4
R antagonist, has been explored for the
treatment of RA patients with active disease despite concomitant methotrexate therapy (phase 2 trials,
ClinicalTrials.gov database entry NCT01862224 and dose range finding study NCT01679951) [
92
,
93
].
Both studies were prematurely terminated in 2014 because of the lack of ecacy. The similar phase 2
clinical studies for the same compound evaluating ecacy and safety of toreforant in patients with
symptomatic uncontrolled, persistent eosinophilic asthma (NCT01823016) [
94
], and in patients with
moderate to severe plaque-type psoriasis (NCT02295865) [
95
] were completed in 2015 and 2016. In the
former study toreforant (at the dose tested) failed to provide any therapeutic benefit [
96
]. Preclinical
toxicity studies of another H
4
R antagonist, JNJ39758979, provided sucient evidence of an excellent
safe profile encouraging the clinical level testing [
72
]. JNJ39758979 was observed to mitigate RA in the
collagen-induced arthritis models (CIAM) [
59
]. The completed phase 2 clinical trial demonstrating its
safety and eectiveness in human volunteers with persistent asthma (NCT00946569) whereas several
phase 1 studies stating its safety and pharmacokinetics, as well as its eect on histamine-induced itch
(pruritus) (NCT01068223) in healthy male volunteers have successfully been accomplished [
97
,
98
].
Simultaneously, the two phase 2 clinical studies were initiated to find a dose range of JNJ39758979
in patients with RA despite concomitant methotrexate therapy (NCT01480388) and patients with
uncontrolled asthma (NCT01493882) but they were withdrawn in 2014 and 2015, respectively, due to
the same reasons [
99
,
100
]. This adverse eect was predicted to be related with reactive metabolites
of JNJ39758979 and not with H
4
R antagonism. Hence, the significant reduction in the pruritus after
JNJ39758979 administration can be concluded in the way that drug-induced agranulocytosis can
be most likely an o-target eect and other H
4
R antagonists could be beneficial in the treatment
of AD, particularly pruritus, without serious adverse eects [
101
]. In the similar clinical studies,
another oral, potent, and selective H
4
R antagonist ZPL3893787 has completed phase 2 clinical trials
determining its safety, ecacy, and tolerability on pruritus in adult subjects with moderate to severe AD
(NCT02424253) [
102
] and in patients with plaque psoriasis (NCT02618616) [
103
] in 2016 but no results
for both these studies were posted on ClinicalTrials.gov. Results showed that ZPL3893787 improved
inflammatory skin lesions in patients with AD, confirming H
4
R antagonism as a novel therapeutic
option [
90
]. Additionally, in two dierent phase 2 trials, there is an evaluation safety and ecacy
of ZPL3893787 in patients with moderate to severe AD (NCT03517566) [
104
] and the impact of its
concomitant use along with topical corticosteroids (TCS) and/or topical calcineurin inhibitors (TCI) in
patients with AD (NCT03948334) [
105
]. The ecacy of Seliforant (SENS-111) in patients suering from
acute unilateral vestibulopathy is currently under evaluation in Phase 2 trial (NCT03110458) [
106
]. The
above-mentioned observations indicate a wide range of potential clinical applications of H
4
R ligands.
Life 2020,10, 50 11 of 17
Table 2.
Details of compounds which are/have been in clinical trial studies which started/ended/
terminated in the 2014–2019 period.
Compound Clinical
Indications Phase Status ClinicalTrials.Gov
Database Entry Ref.
JNJ38518168 (Toreforant)
Life 2020, 10, 50 10 of 17
5. Clinical Trials of Drug Candidates Targeting H4R
Recently, H4R research has been gaining a lot of importance and the clinical studies were
initiated for the putative therapeutic exploitation in inflammatory and allergic disorders [38] such as
atopic dermatitis (AD) [59,90], pruritus, asthma, rheumatoid arthritis (RA), as well as in vestibular
disease (Table 2) [91]. Toreforant (JNJ38518168), the first oral H4R antagonist, has been explored for
the treatment of RA patients with active disease despite concomitant methotrexate therapy (phase 2
trials, ClinicalTrials.gov database entry NCT01862224 and dose range finding study NCT01679951)
[92,93]. Both studies were prematurely terminated in 2014 because of the lack of efficacy. The similar
phase 2 clinical studies for the same compound evaluating efficacy and safety of toreforant in patients
with symptomatic uncontrolled, persistent eosinophilic asthma (NCT01823016) [94], and in patients
with moderate to severe plaque-type psoriasis (NCT02295865) [95] were completed in 2015 and 2016.
In the former study toreforant (at the dose tested) failed to provide any therapeutic benefit [96].
Preclinical toxicity studies of another H4R antagonist, JNJ39758979, provided sufficient evidence of
an excellent safe profile encouraging the clinical level testing [72]. JNJ39758979 was observed to
mitigate RA in the collagen-induced arthritis models (CIAM) [59]. The completed phase 2 clinical
trial demonstrating its safety and effectiveness in human volunteers with persistent asthma
(NCT00946569) whereas several phase 1 studies stating its safety and pharmacokinetics, as well as its
effect on histamine-induced itch (pruritus) (NCT01068223) in healthy male volunteers have
successfully been accomplished [97,98]. Simultaneously, the two phase 2 clinical studies were
initiated to find a dose range of JNJ39758979 in patients with RA despite concomitant methotrexate
therapy (NCT01480388) and patients with uncontrolled asthma (NCT01493882) but they were
withdrawn in 2014 and 2015, respectively, due to the same reasons [99,100]. This adverse effect was
predicted to be related with reactive metabolites of JNJ39758979 and not with H4R antagonism.
Hence, the significant reduction in the pruritus after JNJ39758979 administration can be concluded
in the way that drug-induced agranulocytosis can be most likely an off-target effect and other H4R
antagonists could be beneficial in the treatment of AD, particularly pruritus, without serious adverse
effects [101]. In the similar clinical studies, another oral, potent, and selective H4R antagonist
ZPL3893787 has completed phase 2 clinical trials determining its safety, efficacy, and tolerability on
pruritus in adult subjects with moderate to severe AD (NCT02424253) [102] and in patients with
plaque psoriasis (NCT02618616) [103] in 2016 but no results for both these studies were posted on
ClinicalTrials.gov. Results showed that ZPL3893787 improved inflammatory skin lesions in patients
with AD, confirming H4R antagonism as a novel therapeutic option [90]. Additionally, in two
different phase 2 trials, there is an evaluation safety and efficacy of ZPL3893787 in patients with
moderate to severe AD (NCT03517566) [104] and the impact of its concomitant use along with topical
corticosteroids (TCS) and/or topical calcineurin inhibitors (TCI) in patients with AD (NCT03948334)
[105]. The efficacy of Seliforant (SENS-111) in patients suffering from acute unilateral vestibulopathy
is currently under evaluation in Phase 2 trial (NCT03110458) [106]. The above-mentioned
observations indicate a wide range of potential clinical applications of H4R ligands.
Table 2. Details of compounds which are/have been in clinical trial studies which
started/ended/terminated in the 2014–2019 period.
Compound Clinical
Indications Phase Status ClinicalTrials.Gov
Database Entry Ref.
JNJ38518168
(Toreforant)
RA 2 T NCT01862224 [92]
RA 2 T NCT01679951 [93]
Asthma 2 C NCT01823016 [94]
Psoriasis 2 C NCT02295865 [95]
JNJ39758979 RA 2 W NCT01480388 [99]
Asthma 2 W NCT01493882 [100]
RA 2 T NCT01862224 [92]
RA 2 T NCT01679951 [93]
Asthma 2 C NCT01823016 [94]
Psoriasis 2 C NCT02295865 [95]
JNJ39758979
Life 2020, 10, 50 11 of 17
ZPL3893787
(Adriforant/PF38937
87/ZPL389)
AD 2 C NCT02424253 [102]
Psoriasis 2 C NCT02618616 [103]
AD 2 R NCT03517566 [104]
AD 2 R NCT03948334 [105]
SENS-111
(Seliforant)
Unilateral
Vestibulopathy
2 R NCT03110458 [106]
*Status: T: terminated; C: completed; R: recruiting; W: withdrawn.
6. Challenges and Perspectives
The H4R research triggered serious concern as to the role of histamine in the regulation of
immune (patho)physiology. It has been established that JNJ7777120 acts as an antagonist in respect
to G protein-dependent signaling, but it also recruits β-arrestin to the receptor in a non-G protein-
dependent manner [107]. Moreover, JNJ7777120 acts as a partial inverse agonist at the human H4R
but as a partial agonist at the rat and mouse H4 receptors [46], which show a lower constitutive
activity than their human counterpart [45,46,108,109]. Frequently generated controversies and even
in vivo misleading results in a variety of experimental models have been the repercussions of these
problems [109]. The clinical development of JNJ7777120 as a prototype experimental tool was
hampered due to several setbacks that surfaced over the past two decades including: localized
concerns over the receptor subtypes, ligand binding and functional selectivity, constitutive and
intrinsic activity and the biased signaling [6,46,50,51,95,110], its short half-life in vivo, and the
hypoadrenocorticism toxicity concerns [50]. Therefore, the experimental findings on the role of H4R
cannot be relied upon and need thorough investigation with caution.
Although GPCR biased signaling significantly complicates drug discovery attempts, it makes a
great promise to design specific ligands with minor side effects [95,111]. The precise drugs have
rapidly become the center of research for therapeutic exploitation in immunopharmacology as well
as clinical immunology [90,112,113]. However, in addition to H4R, significant evidence attributes
some immunomodulatory properties to H2R [90,110], thus, dissection of histamine functions in the
immune system becomes indispensable. Although there are many problems in H4R research, a
significant number of studies focusing on H4R provide an optimistic research perspective for this new
drug target.
Author Contributions: Conceptualization, P.M. (Pakhuri Mehta) and S.F.; writing—original draft preparation,
P.M. (Pakhuri Mehta); writing—review and editing, P.M. (Pakhuri Mehta), P.M. (Przemysław Miszta), P.R.,
O.M., P.K. and S.F.; visualization, P.M. (Pakhuri Mehta); supervision, S.F.; funding acquisition, S.F., P.R. and
O.M. All authors have read and agreed to the published version of the manuscript.
Funding: This research was funded by NATIONAL SCIENCE CENTRE, POLAND, grant OPUS
2017/25/B/NZ7/02788.
RA 2 W NCT01480388 [99]
Asthma 2 W NCT01493882 [100]
ZPL3893787
(Adriforant/PF3893787/ZPL389)
Life 2020, 10, 50 11 of 17
ZPL3893787
(Adriforant/PF38937
87/ZPL389)
AD 2 C NCT02424253 [102]
Psoriasis 2 C NCT02618616 [103]
AD 2 R NCT03517566 [104]
AD 2 R NCT03948334 [105]
SENS-111
(Seliforant)
Unilateral
Vestibulopathy
2 R NCT03110458 [106]
*Status: T: terminated; C: completed; R: recruiting; W: withdrawn.
6. Challenges and Perspectives
The H4R research triggered serious concern as to the role of histamine in the regulation of
immune (patho)physiology. It has been established that JNJ7777120 acts as an antagonist in respect
to G protein-dependent signaling, but it also recruits β-arrestin to the receptor in a non-G protein-
dependent manner [107]. Moreover, JNJ7777120 acts as a partial inverse agonist at the human H4R
but as a partial agonist at the rat and mouse H4 receptors [46], which show a lower constitutive
activity than their human counterpart [45,46,108,109]. Frequently generated controversies and even
in vivo misleading results in a variety of experimental models have been the repercussions of these
problems [109]. The clinical development of JNJ7777120 as a prototype experimental tool was
hampered due to several setbacks that surfaced over the past two decades including: localized
concerns over the receptor subtypes, ligand binding and functional selectivity, constitutive and
intrinsic activity and the biased signaling [6,46,50,51,95,110], its short half-life in vivo, and the
hypoadrenocorticism toxicity concerns [50]. Therefore, the experimental findings on the role of H4R
cannot be relied upon and need thorough investigation with caution.
Although GPCR biased signaling significantly complicates drug discovery attempts, it makes a
great promise to design specific ligands with minor side effects [95,111]. The precise drugs have
rapidly become the center of research for therapeutic exploitation in immunopharmacology as well
as clinical immunology [90,112,113]. However, in addition to H4R, significant evidence attributes
some immunomodulatory properties to H2R [90,110], thus, dissection of histamine functions in the
immune system becomes indispensable. Although there are many problems in H4R research, a
significant number of studies focusing on H4R provide an optimistic research perspective for this new
drug target.
Author Contributions: Conceptualization, P.M. (Pakhuri Mehta) and S.F.; writing—original draft preparation,
P.M. (Pakhuri Mehta); writing—review and editing, P.M. (Pakhuri Mehta), P.M. (Przemysław Miszta), P.R.,
O.M., P.K. and S.F.; visualization, P.M. (Pakhuri Mehta); supervision, S.F.; funding acquisition, S.F., P.R. and
O.M. All authors have read and agreed to the published version of the manuscript.
Funding: This research was funded by NATIONAL SCIENCE CENTRE, POLAND, grant OPUS
2017/25/B/NZ7/02788.
AD 2 C NCT02424253 [102]
Psoriasis 2 C NCT02618616 [103]
AD 2 R NCT03517566 [104]
AD 2 R NCT03948334 [105]
SENS-111 (Seliforant)
Life 2020, 10, 50 11 of 17
ZPL3893787
(Adriforant/PF38937
87/ZPL389)
AD 2 C NCT02424253 [102]
Psoriasis 2 C NCT02618616 [103]
AD 2 R NCT03517566 [104]
AD 2 R NCT03948334 [105]
SENS-111
(Seliforant)
Unilateral
Vestibulopathy
2 R NCT03110458 [106]
*Status: T: terminated; C: completed; R: recruiting; W: withdrawn.
6. Challenges and Perspectives
The H4R research triggered serious concern as to the role of histamine in the regulation of
immune (patho)physiology. It has been established that JNJ7777120 acts as an antagonist in respect
to G protein-dependent signaling, but it also recruits β-arrestin to the receptor in a non-G protein-
dependent manner [107]. Moreover, JNJ7777120 acts as a partial inverse agonist at the human H4R
but as a partial agonist at the rat and mouse H4 receptors [46], which show a lower constitutive
activity than their human counterpart [45,46,108,109]. Frequently generated controversies and even
in vivo misleading results in a variety of experimental models have been the repercussions of these
problems [109]. The clinical development of JNJ7777120 as a prototype experimental tool was
hampered due to several setbacks that surfaced over the past two decades including: localized
concerns over the receptor subtypes, ligand binding and functional selectivity, constitutive and
intrinsic activity and the biased signaling [6,46,50,51,95,110], its short half-life in vivo, and the
hypoadrenocorticism toxicity concerns [50]. Therefore, the experimental findings on the role of H4R
cannot be relied upon and need thorough investigation with caution.
Although GPCR biased signaling significantly complicates drug discovery attempts, it makes a
great promise to design specific ligands with minor side effects [95,111]. The precise drugs have
rapidly become the center of research for therapeutic exploitation in immunopharmacology as well
as clinical immunology [90,112,113]. However, in addition to H4R, significant evidence attributes
some immunomodulatory properties to H2R [90,110], thus, dissection of histamine functions in the
immune system becomes indispensable. Although there are many problems in H4R research, a
significant number of studies focusing on H4R provide an optimistic research perspective for this new
drug target.
Author Contributions: Conceptualization, P.M. (Pakhuri Mehta) and S.F.; writing—original draft preparation,
P.M. (Pakhuri Mehta); writing—review and editing, P.M. (Pakhuri Mehta), P.M. (Przemysław Miszta), P.R.,
O.M., P.K. and S.F.; visualization, P.M. (Pakhuri Mehta); supervision, S.F.; funding acquisition, S.F., P.R. and
O.M. All authors have read and agreed to the published version of the manuscript.
Funding: This research was funded by NATIONAL SCIENCE CENTRE, POLAND, grant OPUS
2017/25/B/NZ7/02788.
Unilateral
Vestibulopathy 2 R NCT03110458 [106]
Status: T: terminated; C: completed; R: recruiting; W: withdrawn.
6. Challenges and Perspectives
The H
4
R research triggered serious concern as to the role of histamine in the regulation of immune
(patho)physiology. It has been established that JNJ7777120 acts as an antagonist in respect to G
protein-dependent signaling, but it also recruits
β
-arrestin to the receptor in a non-G protein-dependent
manner [
107
]. Moreover, JNJ7777120 acts as a partial inverse agonist at the human H
4
R but as a partial
agonist at the rat and mouse H
4
receptors [
46
], which show a lower constitutive activity than their
human counterpart [
45
,
46
,
108
,
109
]. Frequently generated controversies and even
in vivo
misleading
results in a variety of experimental models have been the repercussions of these problems [
109
].
The clinical development of JNJ7777120 as a prototype experimental tool was hampered due to
several setbacks that surfaced over the past two decades including: localized concerns over the
receptor subtypes, ligand binding and functional selectivity, constitutive and intrinsic activity and the
biased signaling [
6
,
46
,
50
,
51
,
95
,
110
], its short half-life
in vivo
, and the hypoadrenocorticism toxicity
concerns [
50
]. Therefore, the experimental findings on the role of H
4
R cannot be relied upon and need
thorough investigation with caution.
Although GPCR biased signaling significantly complicates drug discovery attempts, it makes
a great promise to design specific ligands with minor side eects [
95
,
111
]. The precise drugs have
rapidly become the center of research for therapeutic exploitation in immunopharmacology as well as
clinical immunology [
90
,
112
,
113
]. However, in addition to H
4
R, significant evidence attributes some
immunomodulatory properties to H
2
R [
90
,
110
], thus, dissection of histamine functions in the immune
system becomes indispensable. Although there are many problems in H
4
R research, a significant
number of studies focusing on H
4
R provide an optimistic research perspective for this new drug target.
Author Contributions:
Conceptualization, P.M. (Pakhuri Mehta) and S.F.; writing—original draft preparation,
P.M. (Pakhuri Mehta); writing—review and editing, P.M. (Pakhuri Mehta), P.M. (Przemysław Miszta), P.R., O.M.,
P.K. and S.F.; visualization, P.M. (Pakhuri Mehta); supervision, S.F.; funding acquisition, S.F., P.R. and O.M.
All authors have read and agreed to the published version of the manuscript.
Life 2020,10, 50 12 of 17
Funding:
This research was funded by NATIONAL SCIENCE CENTRE, POLAND, grant OPUS 2017/25/
B/NZ7/02788.
Conflicts of Interest:
The authors declare no conflict of interest. The funders had no role in the design of the
study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to
publish the results.
References
1.
Obara, I.; Telezhkin, V.; Alrashdi, I.; Chazot, P.L. Histamine, histamine receptors, and neuropathic pain relief.
Br. J. Pharmacol. 2020,177, 580–599. [CrossRef]
2.
Worm, J.; Falkenberg, K.; Olesen, J. Histamine and migraine revisited: Mechanisms and possible drug targets.
J. Headache Pain 2019,20, 30. [CrossRef]
3.
Chazot, P.L.; Johnston, L.; McAuley, E.; Bonner, S. Histamine and Delirium: Current Opinion. Front.
Pharmacol. 2019,10, 299. [CrossRef]
4.
Durante, M.; Sgambellone, S.; Lanzi, C.; Nardini, P.; Pini, A.; Moroni, F.; Masini, E.; Lucarini, L. Eects of
PARP-1 Deficiency and Histamine H4 Receptor Inhibition in an Inflammatory Model of Lung Fibrosis in
Mice. Front. Pharmacol. 2019,10, 525. [CrossRef]
5.
Li, G.; Fan, Y.; Lai, Y.; Han, T.; Li, Z.; Zhou, P.; Pan, P.; Wang, W.; Hu, D.; Liu, X.; et al. Coronavirus infections
and immune responses. J. Med. Virol. 2020,92, 424–432. [CrossRef]
6.
Panula, P.; Chazot, P.L.; Cowart, M.; Gutzmer, R.; Leurs, R.; Liu, W.L.; Stark, H.; Thurmond, R.L.; Haas, H.L.
International union of basic and clinical pharmacology. XCVIII. Histamine receptors. Pharmacol. Rev. 2015,
67, 601–655. [CrossRef]
7. Corrêa, M.F.; Fernandes, J.P.d.S. Histamine H4 receptor ligands: Future applications and state of art. Chem.
Biol. Drug Des. 2015,85, 461–480. [CrossRef]
8.
Cataldi, M.; Borriello, F.; Granata, F.; Annunziato, L.; Marone, G. Histamine receptors and antihistamines:
From discovery to clinical applications. Chem. Immunol. Allergy 2014,100, 214–226. [CrossRef]
9.
Cowden, J.M.; Yu, F.; Banie, H.; Farahani, M.; Ling, P.; Nguyen, S.; Riley, J.P.; Zhang, M.; Zhu, J.; Dunford, P.J.;
et al. The histamine H4 receptor mediates inflammation and Th17 responses in preclinical models of arthritis.
Ann. Rheum. Dis. 2014,73, 600–608. [CrossRef]
10.
Jemima, E.A.; Prema, A.; Thangam, E.B. Functional characterization of histamine H4 receptor on human
mast cells. Mol. Immunol. 2014,62, 19–28. [CrossRef]
11.
Schneider, E.H.; Seifert, R. The histamine H4-receptor and the central and peripheral nervous system: A
critical analysis of the literature. Neuropharmacology 2016,106, 116–128. [CrossRef]
12.
Sadek, B.; Stark, H. Cherry-picked ligands at histamine receptor subtypes. Neuropharmacology
2016
,106,
56–73. [CrossRef]
13. Haas, H.L.; Panula, P.P. Histamine receptors. Neuropharmacology 2016,106, 1–2. [CrossRef]
14.
Zhu, J.; Qu, C.; Lu, X.; Zhang, S. Activation of microglia by histamine and substance P. Cell. Physiol. Biochem.
2014,34, 768–780. [CrossRef]
15.
Feliszek, M.; Speckmann, V.; Schacht, D.; von Lehe, M.; Stark, H.; Schlicker, E. A search for functional
histamine H4 receptors in the human, guinea pig and mouse brain. Naunyn Schmiedebergs Arch. Pharmacol.
2015,388, 11–17. [CrossRef]
16. Filipek, S. Molecular switches in GPCRs. Curr. Opin. Struct. Biol. 2019,55, 114–120. [CrossRef]
17.
Zhou, Q.; Yang, D.; Wu, M.; Guo, Y.; Guo, W.; Zhong, L.; Cai, X.; Dai, A.; Jang, W.; Shakhnovich, E.I.; et al.
Common activation mechanism of class A GPCRs. eLife 2019,8, e50279. [CrossRef]
18.
Ballesteros, J.A.; Weinstein, H. Integrated methods for the construction of three-dimensional models and
computational probing of structure-function relations in G protein-coupled receptors. Methods Neurosci.
1995,25, 366–428.
19.
Zhu, Y.; Michalovich, D.; Wu, H.-L.; Tan, K.B.; Dytko, G.M.; Mannan, I.J.; Boyce, R.; Alston, J.; Tierney, L.A.;
Li, X.; et al. Cloning, Expression, and Pharmacological Characterization of a Novel Human Histamine
Receptor. Mol. Pharmacol. 2001,59, 434–441. [CrossRef]
20.
Huang, E.S. Construction of a sequence motif characteristic of aminergic G protein–coupled receptors. Protein
Sci. 2003,12, 1360–1367. [CrossRef]
Life 2020,10, 50 13 of 17
21.
Pappalardo, M.; Shachaf, N.; Basile, L.; Milardi, D.; Zeidan, M.; Raiyn, J.; Guccione, S.; Rayan, A. Sequential
application of ligand and structure based modeling approaches to index chemicals for their hH4R antagonism.
PLoS ONE 2014,9, e109340. [CrossRef]
22.
Kiss, R.; Noszal, B.; Racz, A.; Falus, A.; Eros, D.; Keseru, G.M. Binding mode analysis and enrichment studies
on homology models of the human histamine H4 receptor. Eur. J. Med. Chem.
2008
,43, 1059–1070. [CrossRef]
23.
Lim, H.D.; de Graaf, C.; Jiang, W.; Sadek, P.; McGovern, P.M.; Istyastono, E.P.; Bakker, R.A.; de Esch, I.J.;
Thurmond, R.L.; Leurs, R. Molecular determinants of ligand binding to H4R species variants. Mol. Pharmacol.
2010,77, 734–743. [CrossRef]
24.
Kuhne, S.; Kooistra, A.J.; Bosma, R.; Bortolato, A.; Wijtmans, M.; Vischer, H.F.; Mason, J.S.; de Graaf, C.;
de Esch, I.J.; Leurs, R. Identification of Ligand Binding Hot Spots of the Histamine H1 Receptor following
Structure-Based Fragment Optimization. J. Med. Chem. 2016,59, 9047–9061. [CrossRef]
25.
Liu, C.; Ma, X.-J.; Jiang, X.; Wilson, S.J.; Hofstra, C.L.; Blevitt, J.; Pyati, J.; Li, X.; Chai, W.; Carruthers, N.
Cloning and pharmacological characterization of a fourth histamine receptor (H4) expressed in bone marrow.
Mol. Pharmacol. 2001,59, 420–426. [CrossRef]
26.
Istyastono, E.P.; Kooistra, A.J.; Vischer, H.F.; Kuijer, M.; Roumen, L.; Nijmeijer, S.; Smits, R.A.; de Esch, I.J.P.;
Leurs, R.; de Graaf, C. Structure-based virtual screening for fragment-like ligands of the G protein-coupled
histamine H4 receptor. Med. Chem. Comm. 2015,6, 1003–1017. [CrossRef]
27.
Schultes, S.; Kooistra, A.J.; Vischer, H.F.; Nijmeijer, S.; Haaksma, E.E.J.; Leurs, R.; de Esch, I.J.P.; de Graaf, C.
Combinatorial Consensus Scoring for Ligand-Based Virtual Fragment Screening: A Comparative Case Study
for Serotonin 5-HT3A, Histamine H1, and Histamine H4 Receptors. J. Chem. Inf. Model.
2015
,55, 1030–1044.
[CrossRef]
28.
Jie, Q.; Kodithuwakku, N.D.; Yuan, X.; He, G.; Chen, M.; Xu, S.; Wu, Y. Anti-allergic and anti-inflammatory
properties of a potent histamine H1 receptor antagonist, desloratadine citrate disodium injection, and its
anti-inflammatory mechanism on EA.hy926 endothelial cells. Eur. J. Pharmacol. 2015,754, 1–10. [CrossRef]
29.
Thangam, E.B.; Jemima, E.A.; Singh, H.; Baig, M.S.; Khan, M.; Mathias, C.B.; Church, M.K.; Saluja, R. The
Role of Histamine and Histamine Receptors in Mast Cell-Mediated Allergy and Inflammation: The Hunt for
New Therapeutic Targets. Front. Immunol. 2018,9, 1873. [CrossRef]
30.
Shim, Y.K.; Kim, N. The Eect of H2 Receptor Antagonist in Acid Inhibition and Its Clinical Ecacy. Korean
J. Gastroenterol. Taehan Sohwagi Hakhoe Chi 2017,70, 4–12. [CrossRef]
31.
van Wering, H.M.; Benninga, M.A. Histamine-2 Receptor Antagonist in the Treatment of Gastroesophageal Reflux
Disease; Springer: Berlin/Heidelberg, Germany, 2017; pp. 987–994. [CrossRef]
32.
Mohsen, A.; Yoshikawa, T.; Miura, Y.; Nakamura, T.; Naganuma, F.; Shibuya, K.; Iida, T.; Harada, R.;
Okamura, N.; Watanabe, T.; et al. Mechanism of the histamine H(3) receptor-mediated increase in exploratory
locomotor activity and anxiety-like behaviours in mice. Neuropharmacology
2014
,81, 188–194. [CrossRef]
[PubMed]
33.
Jarskog, L.F.; Lowy, M.T.; Grove, R.A.; Keefe, R.S.; Horrigan, J.P.; Ball, M.P.; Breier, A.; Buchanan, R.W.;
Carter, C.S.; Csernansky, J.G.; et al. A Phase II study of a histamine H (3) receptor antagonist GSK239512 for
cognitive impairment in stable schizophrenia subjects on antipsychotic therapy. Schizophr. Res.
2015
,164,
136–142. [CrossRef] [PubMed]
34.
Sadek, B.; Saad, A.; Latacz, G.; Kuder, K.; Olejarz, A.; Karcz, T.; Stark, H.; Kiec-Kononowicz, K.
Non-imidazole-based histamine H3 receptor antagonists with anticnvulsant activity in dierent seizure
models in male adult rats. Drug Des. Dev. Ther. 2016,10, 3879–3898. [CrossRef] [PubMed]
35.
Sadek, B.; Saad, A.; Subramanian, D.; Shafiullah, M.; Lazewska, D.; Kiec-Kononowiczc, K. Anticonvulsant
and procognitive properties of the non-imidazole histamine H3 receptor antagonist DL77 in male adult rats.
Neuropharmacology 2016,106, 46–55. [CrossRef]
36.
Sadek, B.; Schwed, J.S.; Subramanian, D.; Weizel, L.; Walter, M.; Adem, A.; Stark, H. Non-imidazole histamine
H3 receptor ligands incorporating antiepileptic moieties. Eur. J. Med. Chem. 2014,77, 269–279. [CrossRef]
37.
Eissa, N.; Khan, N.; Ojha, S.K.; Lazewska, D.; Kiec-Kononowicz, K.; Sadek, B. The Histamine H3 Receptor
Antagonist DL77 Ameliorates MK801-Induced Memory Deficits in Rats. Front. Neurosci.
2018
,12, 42.
[CrossRef]
38.
Liu, W.L. Histamine H4 receptor antagonists for the treatment of inflammatory disorders. Drug Discov. Today
2014,19, 1222–1225. [CrossRef]
Life 2020,10, 50 14 of 17
39.
Corr
ê
a, M.F.; Varela, M.T.; Balbino, A.M.; Torrecilhas, A.C.; Landgraf, R.G.; Troncone, L.R.P.; Fernandes, J.P.d.S.
1-[(2,3-Dihydro-1-benzofuran-2-yl) methyl] piperazines as novel anti-inflammatory compounds: Synthesis
and evaluation on H3R/H4R. Chem. Biol. Drug Des. 2017,90, 317–322. [CrossRef]
40.
Sterle, H.A.; Nicoud, M.B.; Massari, N.A.; T
á
quez Delgado, M.A.; Herrero Ducloux, M.V.; Cremaschi, G.A.;
Medina, V.A. Immunomodulatory role of histamine H4 receptor in breast cancer. Br. J. Cancer
2019
,120,
128–138. [CrossRef]
41.
Lazewska, D.; Mogilski, S.; Hagenow, S.; Kuder, K.; Gluch-Lutwin, M.; Siwek, A.; Wiecek, M.; Kaleta, M.;
Seibel, U.; Buschauer, A.; et al. Alkyl derivatives of 1,3,5-triazine as histamine H4 receptor ligands. Bioorg.
Med. Chem. 2019,27, 1254–1262. [CrossRef]
42.
Stasiak, A.; Gola, J.; Kraszewska, K.; Mussur, M.; Kobos, J.; Mazurek, U.; Stark, H.; Fogel, W.A. Experimental
autoimmune myocarditis in rats and therapeutic histamine H1–H4 receptor inhibition. J. Physiol. Pharmacol.
2018,69, 889–900. [CrossRef]
43.
Alexander, S.P.; Christopoulos, A.; Davenport, A.P.; Kelly, E.; Marrion, N.V.; Peters, J.A.; Faccenda, E.;
Harding, S.D.; Pawson, A.J.; Sharman, J.L. The Concise Guide to PHARMACOLOGY 2017/18: G
protein-coupled receptors. Br. J. Pharmacol. 2017,174, S17–S129. [CrossRef] [PubMed]
44.
Tiligada, E.; Ennis, M. Histamine pharmacology: From Sir Henry Dale to the 21st century. Br. J. Pharmacol.
2020,177, 469–489. [CrossRef] [PubMed]
45.
Wifling, D.; Bernhardt, G.; Dove, S.; Buschauer, A. The Extracellular Loop 2 (ECL2) of the Human Histamine
H4 Receptor Substantially Contributes to Ligand Binding and Constitutive Activity. PLoS ONE
2015
,10,
e0117185. [CrossRef]
46.
Wifling, D.; Löel, K.; Nordemann, U.; Strasser, A.; Bernhardt, G.; Dove, S.; Seifert, R.; Buschauer, A.
Molecular determinants for the high constitutive activity of the human histamine H4 receptor: Functional
studies on orthologues and mutants. Br. J. Pharmacol. 2015,172, 785–798. [CrossRef]
47. UniProt Database. Available online: https://www.uniprot.org/(accessed on 23 April 2020).
48. Clustal Omega Service. Available online: https://www.ebi.ac.uk/Tools/msa/clustalo/(accessed on 23 April 2020).
49.
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. [CrossRef]
50.
Thurmond, R.L.; Venable, J.; Savall, B.; La, D.; Snook, S.; Dunford, P.J.; Edwards, J.P. Clinical Development of
Histamine H4 Receptor Antagonists. In Histamine and Histamine Receptors in Health and Disease; Hattori, Y.,
Seifert, R., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 301–320.
51.
Thurmond, R.L. The histamine H4 receptor: From orphan to the clinic. Front. Pharmacol.
2015
,6, 65.
[CrossRef]
52.
Deiteren, A.; De Man, J.G.; Pelckmans, P.A.; De Winter, B.Y. Histamine H(4) receptors in the gastrointestinal
tract. Br. J. Pharmacol. 2015,172, 1165–1178. [CrossRef]
53.
Ehling, S.; Rossbach, K.; Dunston, S.M.; Stark, H.; Baumer, W. Allergic inflammation is augmented via
histamine H4 receptor activation: The role of natural killer cells
in vitro
and
in vivo
.J. Dermatol. Sci.
2016
,
83, 106–115. [CrossRef]
54.
Grosicki, M.; Kie´c-Kononowicz, K. Human eosinophils potential pharmacological model applied in human
histamine H4 receptor research. Curr. Med. Chem. 2015,22, 2087–2099. [CrossRef]
55.
De Benedetto, A.; Yoshida, T.; Fridy, S.; Park, J.E.; Kuo, I.H.; Beck, L.A. Histamine and Skin Barrier: Are
Histamine Antagonists Useful for the Prevention or Treatment of Atopic Dermatitis? J. Clin. Med.
2015
,4,
741–755. [CrossRef] [PubMed]
56.
Levoin, N.; Labeeuw, O.; Billot, X.; Calmels, T.; Danvy, D.; Krief, S.; Berrebi-Bertrand, I.; Lecomte, J.M.;
Schwartz, J.C.; Capet, M. Discovery of nanomolar ligands with novel scaolds for the histamine H4 receptor
by virtual screening. Eur. J. Med. Chem. 2017,125, 565–572. [CrossRef]
57.
Labeeuw, O.; Levoin, N.; Billot, X.; Danvy, D.; Calmels, T.; Krief, S.; Ligneau, X.; Berrebi-Bertrand, I.; Robert, P.;
Lecomte, J.M.; et al. Synthesis and evaluation of a 2-benzothiazolylphenylmethyl ether class of histamine H4
receptor antagonists. Bioorg. Med. Chem. Lett. 2016,26, 5263–5266. [CrossRef] [PubMed]
58.
Ohsawa, Y.; Hirasawa, N. The Role of Histamine H1 and H4 Receptors in Atopic Dermatitis: From Basic
Research to Clinical Study. Allergol. Int. 2014,63, 533–542. [CrossRef]
59.
Savall, B.M.; Chavez, F.; Tays, K.; Dunford, P.J.; Cowden, J.M.; Hack, M.D.; Wolin, R.L.; Thurmond, R.L.;
Edwards, J.P. Discovery and SAR of 6-alkyl-2,4-diaminopyrimidines as histamine H(4) receptor antagonists.
J. Med. Chem. 2014,57, 2429–2439. [CrossRef]
Life 2020,10, 50 15 of 17
60.
Chigbu, D.I.; Coyne, A.M. Update and clinical utility of alcaftadine ophthalmic solution 0.25% in the
treatment of allergic conjunctivitis. Clin. Ophthalmol. 2015,9, 1215–1225. [CrossRef] [PubMed]
61.
McLaurin, E.B.; Marsico, N.P.; Ackerman, S.L.; Ciolino, J.B.; Williams, J.M.; Villanueva, L.; Hollander, D.A.
Ocular itch relief with alcaftadine 0.25% versus olopatadine 0.2% in allergic conjunctivitis: Pooled analysis
of two multicenter randomized clinical trials. Adv. Ther. 2014,31, 1059–1071. [CrossRef] [PubMed]
62.
Grzybowska-Kowalczyk, A.; Maslinska, D.; Wojciechowska, M.; Szukiewicz, D.; Wojtecka-Lukasik, E.;
Paradowska, A.; Maldyk, P.; Maslinski, S. Expression of histamine H4 receptor in human osteoarthritic
synovial tissue. Inflamm. Res. 2008,57 (Suppl. 1), S63–S64. [CrossRef] [PubMed]
63.
Grzybowska-Kowalczyk, A.; Wojtecka-Lukasik, E.; Maslinska, D.; Gujski, M.; Maslinski, S. Human and
clinical aspects of histamine. Inflamm. Res. 2007,56 (Suppl. 1), S59–S60. [CrossRef]
64.
Rzodkiewicz, P.; Wojtecka-Łukasik, E.; Ma´sli ´nski, S. Role of histamine in rheumatoid diseases. Reumatol.
Rheumatol. 2010,48, 49–53.
65.
Yu, B.; Shao, Y.; Li, P.; Zhang, J.; Zhong, Q.; Yang, H.; Hu, X.; Chen, B.; Peng, X.; Wu, Q.; et al. Copy number
variations of the human histamine H4 receptor gene are associated with systemic lupus erythematosus. Br. J.
Dermatol. 2010,163, 935–940. [CrossRef] [PubMed]
66.
Deiteren, A.; De Man, J.G.; Ruyssers, N.E.; Moreels, T.G.; Pelckmans, P.A.; De Winter, B.Y. Histamine H4 and
H1 receptors contribute to postinflammatory visceral hypersensitivity. Gut 2014,63, 1873–1882. [CrossRef]
[PubMed]
67.
Andersson, C.; Tufvesson, E.; Diamant, Z.; Bjermer, L. Revisiting the role of the mast cell in asthma. Curr.
Opin. Pulm. Med. 2016,22, 10–17. [CrossRef] [PubMed]
68.
George, L.; Brightling, C.E. Eosinophilic airway inflammation: Role in asthma and chronic obstructive
pulmonary disease. Ther. Adv. Chronic Dis. 2016,7, 34–51. [CrossRef] [PubMed]
69.
Metcalfe, D.D.; Pawankar, R.; Ackerman, S.J.; Akin, C.; Clayton, F.; Falcone, F.H.; Gleich, G.J.; Irani, A.M.;
Johansson, M.W.; Klion, A.D.; et al. Biomarkers of the involvement of mast cells, basophils and eosinophils
in asthma and allergic diseases. World Allergy Organ. J. 2016,9, 7. [CrossRef] [PubMed]
70.
Grosicki, M.; W
ó
jcik, T.; Chlopicki, S.; Kie´c-Kononowicz, K.
In vitro
study of histamine and histamine
receptor ligands influence on the adhesion of purified human eosinophils to endothelium. Eur. J. Pharmacol.
2016,777, 49–59. [CrossRef]
71.
Rosa, A.C.; Pini, A.; Lucarini, L.; Lanzi, C.; Veglia, E.; Thurmond, R.L.; Stark, H.; Masini, E. Prevention
of bleomycin-induced lung inflammation and fibrosis in mice by naproxen and JNJ7777120 treatment. J.
Pharmacol. Exp. Ther. 2014,351, 308–316. [CrossRef]
72.
Thurmond, R.L.; Chen, B.; Dunford, P.J.; Greenspan, A.J.; Karlsson, L.; La, D.; Ward, P.; Xu, X.L. Clinical and
preclinical characterization of the histamine H(4) receptor antagonist JNJ-39758979. J. Pharmacol. Exp. Ther.
2014,349, 176–184. [CrossRef]
73.
Hartwig, C.; Munder, A.; Glage, S.; Wedekind, D.; Schenk, H.; Seifert, R.; Neumann, D. The histamine
H4-receptor (H
4
R) regulates eosinophilic inflammation in ovalbumin-induced experimental allergic asthma
in mice. Eur. J. Immunol. 2015,45, 1129–1140. [CrossRef]
74.
Mahapatra, S.; Albrecht, M.; Behrens, B.; Jirmo, A.; Behrens, G.; Hartwig, C.; Neumann, D.; Raap, U.;
Bahre, H.; Herrick, C.; et al. Delineating the role of histamine-1- and -4-receptors in a mouse model of
Th2-dependent antigen-specific skin inflammation. PLoS ONE 2014,9, e87296. [CrossRef]
75.
Veglia, E.; Grange, C.; Pini, A.; Moggio, A.; Lanzi, C.; Camussi, G.; Chazot, P.L.; Rosa, A.C. Histamine
receptor expression in human renal tubules: A comparative pharmacological evaluation. Inflamm. Res.
2015
,
64, 261–270. [CrossRef] [PubMed]
76.
Rosa, A.C.; Grange, C.; Pini, A.; Katebe, M.; Benetti, E.; Collino, M.; Miglio, G.; Bani, D.; Camussi, G.;
Chazot, P. Overexpression of histamine H4 receptors in the kidney of diabetic rat. Inflamm. Res.
2013
,62,
357–365. [CrossRef] [PubMed]
77.
Pini, A.; Grange, C.; Veglia, E.; Argenziano, M.; Cavalli, R.; Guasti, D.; Calosi, L.; Gh
è
, C.; Solarino, R.;
Thurmond, R.L.; et al. Histamine H4 receptor antagonism prevents the progression of diabetic nephropathy
in male DBA2/J mice. Pharmacol. Res. 2018,128, 18–28. [CrossRef] [PubMed]
78.
Zhou, P.; Homberg, J.R.; Fang, Q.; Wang, J.; Li, W.; Meng, X.; Shen, J.; Luan, Y.; Liao, P.; Swaab, D.F.;
et al. 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
,76, 61–73.
[CrossRef]
Life 2020,10, 50 16 of 17
79.
Nuutinen, S.; Panula, P. Histamine in neurotransmission and brain diseases. Adv. Exp. Med. Biol.
2010
,709,
95–107. [CrossRef]
80.
Medhurst, A.D.; Roberts, J.C.; Lee, J.; Chen, C.P.; Brown, S.H.; Roman, S.; Lai, M.K. Characterization of
histamine H3 receptors in Alzheimer’s Disease brain and amyloid over-expressing TASTPM mice. Br. J.
Pharmacol. 2009,157, 130–138. [CrossRef]
81.
Patnaik, R.; Sharma, A.; Skaper, S.D.; Muresanu, D.F.; Lafuente, J.V.; Castellani, R.J.; Nozari, A.; Sharma, H.S.
Histamine H3 Inverse Agonist BF 2649 or Antagonist with Partial H4 Agonist Activity Clobenpropit Reduces
Amyloid Beta Peptide-Induced Brain Pathology in Alzheimer’s Disease. Mol. Neurobiol.
2018
,55, 312–321.
[CrossRef]
82.
Shan, L.; Bao, A.M.; Swaab, D.F. The human histaminergic system in neuropsychiatric disorders. Trends
Neurosci. 2015,38, 167–177. [CrossRef]
83.
Kim, K.W.; Kim, B.M.; Lee, K.A.; Lee, S.H.; Firestein, G.S.; Kim, H.R. Histamine and Histamine H4 Receptor
Promotes Osteoclastogenesis in Rheumatoid Arthritis. Sci. Rep. 2017,7, 1197. [CrossRef]
84.
Abd-Allah, A.R.; Ahmad, S.F.; Alrashidi, I.; Abdel-Hamied, H.E.; Zoheir, K.M.; Ashour, A.E.; Bakheet, S.A.;
Attia, S.M. Involvement of histamine 4 receptor in the pathogenesis and progression of rheumatoid arthritis.
Int. Immunol. 2014,26, 325–340. [CrossRef]
85.
Massari, N.A.; Medina, V.A.; Lamas, D.J.M.; Cricco, G.P.; Croci, M.; Sambuco, L.; Bergoc, R.M.; Rivera, E.S.
Role of H4 receptor in histamine-mediated responses in human melanoma. Melanoma Res.
2011
,21, 395–404.
[CrossRef] [PubMed]
86.
Medina, V.A.; Brenzoni, P.G.; Lamas, D.J.; Massari, N.; Mondillo, C.; Nunez, M.A.; Pignataro, O.; Rivera, E.S.
Role of histamine H4 receptor in breast cancer cell proliferation. Front. Biosci.
2011
,3, 1042–1060. [CrossRef]
87.
Cricco, G.P.; Mohamad, N.A.; Sambuco, L.A.; Genre, F.; Croci, M.; Guti
é
rrez, A.S.; Medina, V.; Bergoc, R.;
Rivera, E.; Martin, G. Histamine regulates pancreatic carcinoma cell growth through H3 and H4 receptors.
Inflamm. Res. 2008,57, 23–24. [CrossRef] [PubMed]
88.
Boer, K.; Helinger, E.; Helinger, A.; Pocza, P.; Pos, Z.; Demeter, P.; Baranyai, Z.; Dede, K.; Darvas, Z.; Falus, A.
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. [CrossRef] [PubMed]
89.
Saligrama, N.; Noubade, R.; Case, L.K.; del Rio, R.; Teuscher, C. Combinatorial roles for histamine H1-H2
and H3-H4 receptors in autoimmune inflammatory disease of the central nervous system. Eur. J. Immunol.
2012,42, 1536–1546. [CrossRef] [PubMed]
90.
Werfel, T. Novel systemic drugs in treatment of atopic dermatitis: Results from phase II and phase III studies
published in 2017/2018. Curr. Opin. Allergy Clin. Immunol. 2018,18, 432–437. [CrossRef] [PubMed]
91.
Attali, P.; Gomeni, R.; Wersinger, E.; Poli, S.; Venail, F. The eects of SENS-111, a new H4R antagonist, on
vertigo induced by caloric test in healthy volunteers (HV) is related to plasma concentrations. Clin. Ther.
2016,38, e4. [CrossRef]
92.
A Synovial Biopsy Study of JNJ-38518168 in Participants with Active Rheumatoid Arthritis Despite
Methotrexate Therapy (TERMINATED), Database Entry NCT01862224. Available online: https://www.
clinicaltrials.gov/(accessed on 24 May 2013).
93.
A Dose Range Finding Study of JNJ-38518168 in Patients with Active Rheumatoid Arthritis in Spite of
Treatment with Methotrexate (TERMINATED), Database Entry NCT01679951. Available online: https:
//www.clinicaltrials.gov/(accessed on 6 September 2012).
94.
A Study of JNJ-38518168 in Symptomatic Adult Participants with Uncontrolled, Persistent Asthma
(COMPLETED), Database Entry NCT01823016. Available online: https://www.clinicaltrials.gov/(accessed
on 4 April 2013).
95.
Riddy, D.M.; Cook, A.E.; Diepenhorst, N.A.; Bosnyak, S.; Brady, R.; la Cour, C.M.; Mocaer, E.; Summers, R.J.;
Charman, W.N.; Sexton, P.M. Isoform-specific biased agonism of histamine H3 receptor agonists. Mol.
Pharmacol. 2017,91, 87–99. [CrossRef]
Life 2020,10, 50 17 of 17
96.
Kollmeier, A.; Greenspan, A.; Xu, X.; Silko, P.; Barnathan, E.; Loza, M.; Jiang, J.; Zhou, B.; Chen, B.;
Thurmond, R. Phase 2a, randomized, double-blind, placebo-controlled, multicentre, parallel-group study of
an H4R-antagonist (JNJ-39758979) in adults with uncontrolled asthma. Clin. Exp. Allergy 2018,48, 957–969.
[CrossRef]
97.
Kollmeier, A.; Francke, K.; Chen, B.; Dunford, P.J.; Greenspan, A.J.; Xia, Y.; Xu, X.L.; Zhou, B.; Thurmond, R.L.
The histamine H(4) receptor antagonist, JNJ 39758979, is eective in reducing histamine-induced pruritus in
a randomized clinical study in healthy subjects. J. Pharmacol. Exp. Ther. 2014,350, 181–187. [CrossRef]
98.
Kiss, R.; Keseru, G.M. Novel histamine H4 receptor ligands and their potential therapeutic applications: An
update. Expert Opin. Ther. Pat. 2014,24, 1185–1197. [CrossRef] [PubMed]
99.
A Dose Range Finding Study of JNJ-39758979 in Patients with Active Rheumatoid Arthritis Currently
Treated with Methotrexate (WITHDRAWN), Database Entry NCT01480388. Available online: https:
//www.clinicaltrials.gov/(accessed on 28 November 2011).
100.
Study of JNJ-39758979 in Symptomatic Adult Patients with Uncontrolled Asthma (WITHDRAWN), Database
Entry NCT01493882. Available online: https://www.clinicaltrials.gov/(accessed on 16 December 2011).
101.
Murata, Y.; Song, M.; Kikuchi, H.; Hisamichi, K.; Xu, X.L.; Greenspan, A.; Kato, M.; Chiou, C.F.; Kato, T.;
Guzzo, C. Phase 2a, randomized, double-blind, placebo-controlled, multicenter, parallel-group study of a
H4R-antagonist (JNJ-39758979) in Japanese adults with moderate atopic dermatitis. J. Dermatol.
2015
,42,
129–139. [CrossRef] [PubMed]
102.
A Study to Determine the Ecacy of ZPL-3893787 in Subjects with Atopic Dermatitis (COMPLETED),
Database Entry NCT02424253. Available online: https://www.clinicaltrials.gov/(accessed on 23 April 2015).
103.
A Study to Determine the Ecacy of ZPL-3893787 in Subjects with Plaque Psoriasis (COMPLETED), Database
Entry NCT02618616. Available online: https://www.clinicaltrials.gov/(accessed on 1 December 2015).
104.
A Study to Assess the Safety and Ecacy of ZPL389 in Patients with Moderate to Severe Atopic Dermatitis
(RECRUITING), Database Entry NCT03517566. Available online: https://www.clinicaltrials.gov/(accessed
on 7 May 2018).
105.
A Study to Assess the Safety and Ecacy of ZPL389 with TCS/TCI in Atopic Dermatitis Patients (ZESTExt)
(RECRUITING), Database Entry NCT03948334. Available online: https://www.clinicaltrials.gov/(accessed
on 13 May 2019).
106.
Ecacy of SENS-111 in Patients Suering From Acute Unilateral Vestibulopathy (RECRUITING), Database
Entry NCT03110458. Available online: https://www.clinicaltrials.gov/(accessed on 12 April 2017).
107.
Rosethorne, E.M.; Charlton, S.J. Agonist-biased signaling at the histamine H4 receptor: JNJ7777120 recruits
β-arrestin without activating G proteins. Mol. Pharmacol. 2011,79, 749–757. [CrossRef] [PubMed]
108.
Strasser, A.; Wittmann, H.J.; Buschauer, A.; Schneider, E.H.; Seifert, R. Species-dependent activities of
G-protein-coupled receptor ligands: Lessons from histamine receptor orthologs. Trends Pharmacol. Sci.
2013
,
34, 13–32. [CrossRef]
109.
Seifert, R.; Strasser, A.; Schneider, E.H.; Neumann, D.; Dove, S.; Buschauer, A. Molecular and cellular analysis
of human histamine receptor subtypes. Trends Pharmacol. Sci. 2013,34, 33–58. [CrossRef]
110.
Monczor, F.; Fernandez, N. Current knowledge and perspectives on histamine H1 and H2 receptor
pharmacology: Functional selectivity, receptor crosstalk, and repositioning of classic histaminergic ligands.
Mol. Pharmacol. 2016,90, 640–648. [CrossRef]
111. Kenakin, T. Signaling bias in drug discovery. Expert. Opin. Drug Discov. 2017,12, 321–333. [CrossRef]
112.
Tiligada, E.; Ishii, M.; Riccardi, C.; Spedding, M.; Simon, H.U.; Teixeira, M.M.; Landys Chovel Cuervo, M.;
Holgate, S.T.; Levi-Schaer, F. The expanding role of immunopharmacology: IUPHAR Review 16. Br. J.
Pharmacol. 2015,172, 4217–4227. [CrossRef]
113.
Riccardi, C.; Levi-Schaer, F.; Tiligada, E. Immunopharmacology and Inflammation; Springer: Berlin/Heidelberg,
Germany, 2018.
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... 228 H3 receptor is localized in the brain. 229 H3 receptor is an important therapeutic target for cognitive disorders. 230 The neurological function of the H4 receptor remains unclear. ...
... 230 The neurological function of the H4 receptor remains unclear. 229 H4 receptor can be detected in the non-neuronal cells of the brain. 229 H4 receptor activation is involved in the inflammatory responses regulated by mast cells, eosinophils, and T cells. ...
... 229 H4 receptor activation is involved in the inflammatory responses regulated by mast cells, eosinophils, and T cells. 229 Histamine acts on H1 and H3 receptors to control normal sleep/wake behavior. 231 Alterations in histamine signaling are found in both neurodegenerative and psychiatric disorders. ...
Article
Full-text available
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
... HRH4 encodes the GPCR protein HRH4 [30], which is linked to several inflammatory processes [31]. In allergic asthma, histamine signaling via HRH4 is implicated in the immune-inflammatory response [32,33]. ...
... Nonetheless, we observed that HRH4 expression highly correlated with other gene expressions in all patient groups (Figure 1), suggesting a transversal role in the molecular mechanisms underlying T2-asthma. That is a likely hypothesis, given the several lines of evidence linking the receptor to this disease [31][32][33]. ...
Article
Full-text available
Type 2 (T2)-asthma is often associated with chronic rhinosinusitis with nasal polyposis (CRSwNP). Additionally, non-steroidal anti-inflammatory drug intolerance leads to NSAID-exacerbated respiratory disease (N-ERD). Previous transcriptomic data in non-CRSwNP T2-asthma patients showed differentially expressed genes (DEGs). Of them, we focused on ALOX15 , CLC , CYSLTR2 , HRH4 , and SMPD3 to investigate their role in T2-asthma patients. The study included 100 healthy controls (HCs) and 103T2-asthma patients, divided into asthmatics (54), asthmatics with CRSwNP (30), and N-ERD (19). Quantitative PCR analysis was performed on blood-derived RNA samples first to validate the five DEGs. The data were further analyzed to find potential associations and biomarkers. Patients, regardless of stratification, exhibited significantly higher gene expressions than HCs. The patterns of association revealed that ALOX15 was exclusively present in the non-comorbidity group, SMPD3 and CLC in the comorbidity groups, and HRH4 in all patient groups. ALOX15, CYSLTR2 , and SMPD3 expression showed potential as biomarkers to confirm the diagnosis of T2-asthma using peripheral blood eosinophils (PBE) as the initial criterion. PBE combined with gene expressions, especially SMPD3 , may improve the diagnosis. CLC and CYSLTR2 expressions play a specific role in discriminating N-ERD. We validated the transcriptomic data of five DEGs in T2-asthma. Different patterns of association were identified in patient stratification, suggesting different molecular mechanisms underlying the spectrum of T2-asthma. Potential biomarkers were also found and used to design an algorithm with practical diagnostic utility for T2-asthma, including risk stratification for N-ERD.
... While the affinity of H 1 R and H 2 R for histamine is in the μM range, H 3 R and H 4 R are known to have a high affinity in nM range 1,34 . Histamine receptor subtypes therefore require different concentrations of histamine for activation; however, the molecular basis for the selectivity between these subtypes remains unknown given the lack of structural information on histamine receptors. ...
Article
Full-text available
Histamine is a biogenic amine that participates in allergic and inflammatory processes by stimulating histamine receptors. The histamine H4 receptor (H4R) is a potential therapeutic target for chronic inflammatory diseases such as asthma and atopic dermatitis. Here, we show the cryo-electron microscopy structures of the H4R-Gq complex bound with an endogenous agonist histamine or the selective agonist imetit bound in the orthosteric binding pocket. The structures demonstrate binding mode of histamine agonists and that the subtype-selective agonist binding causes conformational changes in Phe3447.39, which, in turn, form the “aromatic slot”. The results provide insights into the molecular underpinnings of the agonism of H4R and subtype selectivity of histamine receptors, and show that the H4R structures may be valuable in rational drug design of drugs targeting the H4R.
... On the other hand, inhibiting HR-3 has a slight initial effect on the mentioned disorder. However, it is important to note that the HR-3 antagonist, thioperamide, partially blocks HR-4, as it shares about 40% homology with HR3 [43,44]. ...
Article
Full-text available
In recent years, there has been a noteworthy revival of interest in the function of mast cells (MCs) in the human body. It is now acknowledged that MCs impact a wide array of processes beyond just allergies, leading to a shift in research direction. Unfortunately, some earlier conclusions were drawn from animal models with flawed designs, particularly centered around the receptor tyrosine kinase (Kit) pathway. Consequently, several subsequent findings may have been unreliable. Thus, what is now required is a re-examination of these earlier findings. Nevertheless, the remaining data are fascinating and hold promise for a better comprehension of numerous diseases and the development of more effective therapies. As the field continues to progress, many intriguing issues warrant further investigation and analysis. For instance, exploring the bidirectional action of MCs in rheumatoid arthritis, understanding the extent of MCs’ impact on symptoms associated with Ehlers–Danlos syndrome, and unraveling the exact role of the myofibroblast–mast cell–neuropeptides axis in the joint capsule during post-traumatic contractures are all captivating areas for exploration. Hence, in this review, we summarize current knowledge regarding the influence of MCs on the pathogenesis of selected musculoskeletal diseases, including rheumatoid arthritis, spondyloarthritis, psoriatic arthritis, gout, muscle and joint injuries, tendinopathy, heterotopic ossification, and Ehlers–Danlos syndrome. We believe that this review will provide in-depth information that can guide and inspire further research in this area.
... Numerous studies demonstrated the anti-pruritic effects of H1R antihistamines in urticaria, and their efficiency in AD is still limited. H4 receptors (H4Rs), the most recently discovered subtype, are expressed on keratinocytes (Figure 3a), neurons (Figure 4a), and various immune cell populations like peripheral mononuclear leukocytes and exert immuno-regulatory effects through the upregulation of IL-31, a T-cellderived cytokine strongly linked with the development of skin barrier dysfunction, local inflammation, and pruritus [105]. Emerging clinical studies specifically targeted the H4R subtype to control the pruritus. ...
Article
Full-text available
Atopic dermatitis represents a complex and multidimensional interaction that represents potential fields of preventive and therapeutic management. In addition to the treatment armamentarium available for atopic dermatitis, novel drugs targeting significant molecular pathways in atopic dermatitis biologics and small molecules are also being developed given the condition’s complex pathophysiology. While most of the patients are expecting better efficacy and long-term control, the response to these drugs would still depend on numerous factors such as complex genotype, diverse environmental triggers and microbiome-derived signals, and, most importantly, dynamic immune responses. This review article highlights the challenges and the recently developed pharmacological agents in atopic dermatitis based on the molecular pathogenesis of this condition, creating a specific therapeutic approach toward a more personalized medicine.
... Histamine receptor 3, HR H3 , is expressed in the central nervous system and participates in neuronal histamine turnover as well as the modulation of the release of other neurotransmitters, such as dopamine and serotonin [6]. Histamine receptor 4, HR H4 , is expressed in peripheral immune cells and is involved in immunomodulation [7]. ...
Article
Full-text available
Histamine receptor 2 (HRH2) blockers are used to treat peptic ulcers and gastric reflux. Chlorquinaldol and chloroxine, which contain an 8-hydroxyquinoline (8HQ) core, have recently been identified as blocking HRH2. To gain insight into the mode of action of 8HQ-based blockers, here, we leverage an HRH2-based sensor in yeast to evaluate the role of key residues in the HRH2 active site on histamine and 8HQ-based blocker binding. We find that the HRH2 mutations D98A, F254A, Y182A, and Y250A render the receptor inactive in the presence of histamine, while HRH2:D186A and HRH2:T190A retain residual activity. Based on molecular docking studies, this outcome correlates with the ability of the pharmacologically relevant histamine tautomers to interact with D98 via the charged amine. Docking studies also suggest that, unlike established HRH2 blockers that interact with both ends of the HRH2 binding site, 8HQ-based blockers interact with only one end, either the end framed by D98/Y250 or T190/D186. Experimentally, we find that chlorquinaldol and chloroxine still inactivate HRH2:D186A by shifting their engagement from D98 to Y250 in the case of chlorquinaldol and D186 to Y182 in the case of chloroxine. Importantly, the tyrosine interactions are supported by the intramolecular hydrogen bonding of the 8HQ-based blockers. The insight gained in this work will aid in the development of improved HRH2 therapeutics. More generally, this work demonstrates that Gprotein-coupled receptor (GPCR)-based sensors in yeast can help elucidate the mode of action of novel ligands for GPCRs, a family of receptors that bind 30% of FDA therapeutics.
... This suggests that mast cells have a vital effect on OA progression (which may be related to structural damage). Mehta et al found that the activation of the histamine H4 receptor participates in cytokines activation and mediates eosinophil chemotaxis, which may also occur in OA [40]. Previous studies support our results. ...
Preprint
Full-text available
Purpose We analyze the immune infiltration model of osteoarthritis to determine the relevant diagnostic biomarkers (OA), and to provide some help for the treatment and diagnosis of OA. Methods From the Gene Expression Omnibus (GEO) database, we downloaded GSE168505 and GSE114007 gene expression datasets, including 24 patients and 21 healthy controls. The R software Limma package and SVA package were used to analyze the batch effect. We selected differentially expressed genes (DEGs), and we then analyzed the DEGs’ functional enrichment. We performed differential analysis to pick out the differentially expressed immune-related genes (DEIRGs) in the merged data set. We first selected the candidate genes by the least absolute shrinkage and selection operator (LASSO) method, and then further screened the diagnostic markers by support vector machine-recursive feature elimination algorithm (SVM-RFE). In dataset GSE129147, the diagnostic value was determined by drawing the receiver operating characteristic (ROC) curve. In addition, we used the CIBERSORT program to assess the 22 kinds immune cells of infiltration models. Finally, an in vitro cell model of OA was established by interleukin-1β(IL-1β) to verify the bioinformatics results. Results Through differential analysis, 454 differential genes were identified, mainly involved ossification, extracellular matrix organization, collagen − containing extracellular matrix, metalloendopeptidase activity, PI3K − Akt signaling pathway, regulation of cell population proliferation, and other biological processes. We screened BIRC5 and TNFSF11 as candidate biomarkers by machine learning. In the data set GSE129147, BIRC5 and TNFSF11 were verified as diagnostic markers of OA by the ROC curve. The following correlation analysis found that BIRC5 and TNFSF11 were correlated with Mast cells resting, NK cells resting, Monocytes, Plasma cells, Eosinophil, Macrophages M0, and Macrophages M2. The expression of BIRC5 and TNFSF11 was up-regulated in the OA model in vitro. Conclusion We conclude that BIRC5 and TNFSF11 can be biomarkers for diagnosing OA. This discovery provides a direction for the occurrence of OA and the exploration of new treatment methods from the perspective of immunology.
... The histaminergic system, along with other biological systems, may have larger unexplored roles in various eye diseases, such as glaucoma, dry eye disease, ocular tumorigenesis, ocular vascular disease [15,24]. ...
... Among the four types of HRs, H4Rs have been associated mostly with immunomodulation [57], and H4Rs have been detected mainly upon cells involved in the immune system or the hematopoietic system in humans as well as in mice (including T cells, MCs, neutrophils, macrophages, and eosinophils) [48]. Although MCs and/or basophils have been considered to be the cells delivering histamine to H4Rs by their release of it [58], various other cells are also supposed to produce histamine via induction of HDC [48]. ...
Article
Full-text available
Objective and methods Nitrogen-containing bisphosphonates (NBPs, anti-bone-resorptive agents) have inflammatory side-effects. Alendronate (Ale, an NBP) intradermally injected into mouse ear-pinnae together with LPS (bacterial cell-wall component) induces augmented ear-swelling that depends on IL-1 and neutrophils. Using this model, we examined histamine’s involvement in Ale + LPS-induced inflammation. Results Ale increased histamine in ear-pinnae by inducing histidine decarboxylase (HDC). This induction was augmented by LPS. In HDC-deficient mice, such augmented ear-swelling was not induced. At peak-swelling, 74.5% of HDC-expressing cells were neutrophils and only 0.2% were mast cells (MCs). The augmented swelling was markedly reduced by a histamine H4-receptor (H4R) antagonist, but not by an H1R antagonist. In MC-deficient mice, unexpectedly, Ale + LPS induced prolonged ear-swelling that was augmented and more persistent than in normal mice. MCs highly expressed H4Rs and produced MCP-1(inflammatory cytokine that recruits macrophages) and IL-10 (anti-inflammatory cytokine) in response to an H4R agonist. Conclusion Histamine produced by HDC-induction mainly in infiltrated neutrophils stimulates H4Rs, leading to augmented Ale + LPS-induced ear-swelling via MCP-1 production by MCs. Since MCP-1 is produced by other cells, too, the contribution of MCs and their H4Rs to augmented ear-swelling is partial. In the later phase of the swelling, MCs may be anti-inflammatory via IL-10 production.
Article
Full-text available
Coronaviruses (CoVs) are by far the largest group of known positive‐sense RNA viruses having an extensive range of natural hosts. In the past few decades, newly evolved Coronaviruses have posed a global threat to public health. Immune response is essential to control and eliminate CoV infections, however, maladjusted immune responses may result in immunopathology and impaired pulmonary gas exchange. Gaining a deeper understanding of the interaction between Coronaviruses and the innate immune systems of the hosts may shed light on the development and persistence of inflammation in the lungs and hopefully can reduce the risk of lung inflammation caused by CoVs. In this review, we provide an update on CoV infections and relevant diseases, particularly the host defense against CoV‐induced inflammation of lung tissue, as well as the role of the innate immune system in pathogenesis and clinical treatment. This article is protected by copyright. All rights reserved.
Article
Full-text available
Class A G protein-coupled receptors (GPCRs) influence virtually every aspect of human physiology. Understanding receptor activation mechanism is critical for discovering novel therapeutics since about one-third of all marketed drugs target members of this family. GPCR activation is an allosteric process that couples agonist binding to G protein recruitment, with the hallmark outward movement of transmembrane helix 6 (TM6). However, what leads to TM6 movement and the key residue level changes of this movement remain less well understood. Here, we report a framework to quantify conformational changes. By analyzing the conformational changes in 234 structures from 45 class A GPCRs, we discovered a common GPCR activation pathway comprising of 34 residue pairs and 35 residues. The pathway unifies previous findings into a common activation mechanism and strings together the scattered key motifs such as CWxP, DRY, Na+ pocket, NPxxY and PIF, thereby directly linking the bottom of ligand-binding pocket with G protein coupling region. Site-directed mutagenesis experiments support this proposition and reveal that rational mutations of residues in this pathway can be used to obtain receptors that are constitutively active or inactive. The common activation pathway provides the mechanistic interpretation of constitutively activating, inactivating and disease mutations. As a module responsible for activation, the common pathway allows for decoupling of the evolution of the ligand binding site and G protein binding region. Such an architecture might have facilitated GPCRs to emerge as a highly successful family of proteins for signal transduction in nature.
Article
Full-text available
Pulmonary fibrosis is the most frequent form of interstitial lung disease. Effective therapies are not yet available; novel therapeutic approaches are needed for counteracting fibrosis. Poly(ADP-ribose) polymerases are enzymes, involved in DNA repair and cell apoptosis. PARP-1 deficient mice exhibited reduced lung fibrosis in response to bleomycin treatment compared to wild-type controls. Histamine H4 receptors (H4Rs) have been recognized as a new target for inflammatory and immune diseases, and H4R ligands reduced inflammation and oxidative stress in lung tissue. The aim of the study was to evaluate the cross-talk between PARP-1 and H4R in a model of bleomycin-induced lung fibrosis in PARP-1−/− and WT mice. Animals were treated with bleomycin or saline by intra-tracheal injection. JNJ7777120, an H4R antagonist, or VUF8430, an H4R agonist, were administered i.p for 21 days. Airway resistance to inflation was evaluated, and lung tissues were processed for PARylated protein content, oxidative stress evaluation, and histology of small bronchi. The levels of pro-inflammatory (IL-1β and TNF-α), regulatory (IL-10), and pro-fibrotic (TGF-β) cytokines were evaluated. The deposition of αSMA was determined by immunofluorescence analysis. The results indicate that JNJ7777120 reduces PARylated protein production, decreases oxidative stress damage, and MPO, a marker for leukocyte tissue infiltration, in PARP-1−/− mice. A significant decrease in the production of both IL-1β and TNF-α and a significant increase in IL-10 levels are observed in mice treated with H4R antagonist, suggesting a crucial anti-inflammatory activity of JNJ7777120. The smooth muscle layer thickness, the goblet cell relative number, and collagen deposition decreased following JNJ7777120 administration. The H4R antagonist treatment also reduces TGF-β production and αSMA deposition, suggesting an important role of JNJ7777120 in airway remodeling. Our results show that PARylation is essential for the pathogenesis of pulmonary fibrosis and propose that PARP-1 and H4Rs are both involved in inflammatory and fibrotic responses. JNJ7777120 treatment, in a condition of PARP-1 inhibition, exerts anti-inflammatory and anti-fibrotic effects, reducing airway remodeling and bronchoconstriction. Therefore, selective inhibition of H4Rs together with non-toxic doses of selective PARP-1 inhibitors could have clinical relevance for the treatment of idiopathic pulmonary fibrosis.
Article
Full-text available
Histamine, acting via distinct histamine H1, H2, H3, and H4 receptors, regulates various physiological and pathological processes, including pain. In the last two decades, there has been a particular increase in evidence to support the involvement of H3 receptor and H4 receptor in the modulation of neuropathic pain, which remains challenging in terms of management. However, recent data show contrasting effects on neuropathic pain due to multiple factors that determine the pharmacological responses of histamine receptors and their underlying signal transduction properties (e.g., localization on either the presynaptic or postsynaptic neuronal membranes). This review summarizes the most recent findings on the role of histamine and the effects mediated by the four histamine receptors in response to the various stimuli associated with and promoting neuropathic pain. We particularly focus on mechanisms underlying histamine‐mediated analgesia, as we aim to clarify the analgesic potential of histamine receptor ligands in neuropathic pain. Linked Articles This article is part of a themed section on New Uses for 21st Century. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc
Article
Full-text available
Delirium is a very common, but refractory clinical state, notably present in intensive care and in the growing aging community. It is characterized by fluctuating disturbances in a number of key behavioral features, namely cognition, mood, attention, arousal, and self-awareness. Histamine is arguably the most pleotropic neurotransmitter in the human brain, and this review provides a rationale, and proposes that this neuroactive amine plays a role in modulating the characteristic features of delirium. While centrally permeable H1 and H2 histamine receptor antagonists have pro-delirium potential, we propose that centrally permeable H3 histamine receptor antagonists may provide an exciting new strategy to combat delirium. The Histamine H4 receptor may also have an indirect inflammatory neuroglial role which requires further exploration.
Article
Full-text available
Objective To review the existing literature on histamine and migraine with a focus on the molecule, its receptors, its use in inducing migraine, and antihistamines in the treatment of migraine. Background Histamine has been known to cause a vascular type headache for almost a hundred years. Research has focused on antihistamines as a possible treatment and histamine as a migraine provoking agent but there has been little interest in this field for the last 25 years. In recent years two additional histamine (H3 and H4) receptors have been discovered and a series of non-sedating antihistamines have been developed. It is therefore timely to review the field again. Methods For this review the PubMed/MEDLINE database was searched for eligible studies. We searched carefully for all articles on histamine, antihistamines and histamine receptors in relation to migraine and the nervous system. The following search terms were used: histamine, migraine disorders, migraine, headache, antihistamines, histamine antagonists, clinical trials, induced headache, histamine H3 receptor, histamine H4 receptor and pharmacology. Four hundred thirty-six titles were read, 135 abstracts were read, 112 articles were read in full and 53 articles were used in this review. Review process resulted in 12 articles added to a total of 65. Findings Early studies of H1 and H2 antihistamines lack scientific strength and show conflicting results. Most of the antihistaminic drugs used in these trials bind also to other receptors which makes it difficult to conclude on the antihistaminic effect. Histamine is an efficient inducer of migraine attacks in migraine patients by an H1 mechanism most likely extracerebrally. These findings merit further investigation of antihistamines in clinical drug trials. The H3 and H4 receptors are found in primarily in CNS and immune tissues, respectively. H3 is likely to be involved in antinociception and has been linked with cognitive, neurodegenerative and sleep disorders. The only marketed H3 agent, pitolisant, is a brain penetrant H3 antagonist/inverse agonist which increases central histamine and causes headache. The experimental H3 agonist Nα-methylhistamine has shown promising results as a migraine preventative in studies of uncertain quality. With the current limited knowledge of the H4 receptor it is questionable whether or not the receptor is involved in migraine. Conclusion There is insufficient support for first generation antihistamines (both H1 and H2) as preventive migraine medications and sedation and weight gain are unacceptable side effects. Non-sedating H1 antihistamines need to be appropriately tested. Central H3 receptors seem to have a role in migraine that merit further investigation. The histaminergic system may be a goal for novel migraine drugs.
Article
Molecular switches in GPCRs enable passing the signal from the agonist binding site, usually located close to the extracellular surface, to the intracellular part of the receptor. The switches are usually associated with conserved structural motifs on transmembrane helices (TMs), and they are accompanied by adjacent residues which provide the signal to the central residue in the toggle switch. In case of locks being the molecular switches, they are breaking (permanently or temporarily) upon agonist binding. Cascade action of switches is correlated with influx of water molecules to form a pathway linking both sides of the receptor. The switches remove the hydrophobic barriers and facilitate water movement while water molecules help to rearrange the hydrogen bond network inside the receptor.
Article
Myocarditis, a life threatening disease, is still not adequately treated. Histamine plays an important role in physiology and pathophysiology of cardiovascular system. All four histamine receptors (H1R - H4R), are present in the heart. Experimental autoimmune myocarditis (EAM) was used to investigate which histamine receptor had a greater impact on the disease's progression. EAM was evoked in Lewis rats by porcine myosin immunization. Mepyramine, ranitidine and ciproxifan were used to inhibit H1R, H2R and H3R receptors, respectively, and 2,4-diaminopyrimidines: ST994, ST1012, ST1006 were ligands of H4R. Quinapril, an ACE inhibitor, served as a reference drug. Drugs were administered daily, either from 0 - 2 weeks or from 2 to 4 weeks post EAM induction. Cardiac dysfunction developed with significant decreases in left ventricular ejection fraction and fractional shortening due to dilatation and wall thickening. EAM rats treated with mepyramine and ST994 in weeks 0 - 2 had the lowest decreases. These treated with ST994, ST1012 or quinapril performed much better the following 2 weeks without therapy than did the other groups. On autopsy their hearts were smaller, less fibrotic, histopathological changes in them of a lower grade. When the treatment started with 2 weeks' delay, the ST994-treated EAM rats showed the highest median survival. H4 receptor antagonism inhibits heart remodelling, preserves heart contractility, improves survival and may be of potent therapeutic relevance in human clinics. The blockade of H1 receptor inhibits heart dilatation but does not prolong the life.
Article
This study focuses on the design, synthesis, molecular modeling and biological evaluation of a novel group of alkyl-1,3,5-triazinyl-methylpiperazines. New compounds were synthesized and their affinities for human histamine H4 receptor (hH4R) were evaluated. Among them, 4-(cyclohexylmethyl)-6-(4-methylpiperazin-1-yl)-1,3,5-triazin-2-amine (14) exhibited hH4R affinity with a Ki of 160 nM and behaved as antagonist in functional assays: the cellular aequorin-based assay (IC50 = 32 nM) and [35S]GTPγS binding assay (pKb = 6.67). In addition, antinociceptive activity of 14in vivo was observed in Formalin test (in mice) and in Carrageenan-induced acute inflammation test (in rats).
Article
The activation of microglial cells is presumed to play a key role in the pathogenesis of Parkinson's disease (PD). The activity of microglia is regulated by the histamine-4 receptor (H4R), thus providing a novel target that may prevent the progression of PD. However, this putative mechanism has so far not been validated. In our previous study, we found that mRNA expression of H4R was upregulated in PD patients. In the present study, we validated this possible mechanism using the rotenone-induced PD rat model, in which mRNA expression levels of H4R-, and microglial markers were significantly increased in the ventral midbrain. Inhibition of H4R in rotenone-induced PD rat model by infusion of the specific H4R antagonist JNJ7777120 into the lateral ventricle resulted in blockade of microglial activation. In addition, pharmacological targeting of H4R in rotenone-lesioned rats resulted in reduced apomorphine-induced rotational behaviour, prevention of dopaminergic neuron degeneration and associated decreases in striatal dopamine levels. These changes were accompanied by a reduction of Lewy body-like neuropathology. Our results provide first proof of the efficacy of an H4R antagonist in a commonly used PD rat model, and proposes the H4R as a promising target to clinically tackle microglial activation and thereby the progression of PD.