Integrative genomic analyses of the histamine H1 receptor and its role in cancer prediction

  • Authors:
    • Minghai Wang
    • Xiaolong Wei
    • Lianghui Shi
    • Bin Chen
    • Guohai Zhao
    • Haiwei Yang
  • View Affiliations

  • Published online on: February 10, 2014     https://doi.org/10.3892/ijmm.2014.1649
  • Pages: 1019-1026
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The human histamine receptor H1 (HRH1) gene is located on chromosome 3p25 and encodes for a 487 amino acid G protein-coupled receptor (GPCR) with a long third intracellular loop (IL3). The HRH1 predominantly couples to Gαq/11 proteins, leading to the activation of phospholipase C (PLC) and subsequent release of the second messengers inositol trisphosphate (IP3) and diacylglycerol (DAG) followed by the activation of PKC and the release of [Ca2+]i. In the present study, we identified HRH1 genes from 14 vertebrate genomes and found that HRH1 exists in all types of vertebrates including fish, amphibians, birds and mammals. We identified 88 SNPs including 4 available alleles disrupting an existing exonic splicing enhancer and 84 SNPs causing missense mutation, which may impact the effect of histamine on the HRH1 protein. We found that the human HRH1 gene was expressed in many tissues or organs, and predominant expression of HRH1 was shown in the bone marrow, whole blood, lymph node, thymus, brain, cerebellum, retina, spinal cord, heart, smooth muscle, skeletal muscle, small intestine, colon, adipocytes, kidney, liver, lung, pancreas, thyroid salivary gland, skin, ovary, uterus, placenta, prostate and testis. When searched in the PrognoScan database, human HRH1 was also found to be expressed in bladder cancer, blood cancer, brain cancer, breast cancer, colorectal cancer, eye cancer, head and neck cancer, lung cancer, ovarian cancer, skin cancer and soft tissue cancer tissues. The relationship between the expression of HRH1 and prognosis was found to vary in different types of cancers, even in the same cancer from different databases. This implies that the function of HRH1 in these tumors may be multidimensional. GR, STAT5A and c-Myb regulatory transcription factor binding sites were identified in the HRH1 gene upstream (promoter) region, which may be involved in the effect of HRH1 in tumors.

Introduction

Histamine is a ubiquitous messenger molecule released from mast cells, enterochromaffin-like cells and neurons. Its various actions are mediated by histamine receptors H1, H2, H3 and H4 (1). The bovine histamine H1 receptor (HRH1) was the first HRH1 gene to be cloned, soon to be followed by other species, including human HRH1. The human HRH1 gene is located on chromosome 3p25 and encodes for a 487 amino acid G protein-coupled receptor (GPCR) with a long third intracellular loop (IL3) (1). The HRH1 predominantly couples to Gαq/11 proteins, leading to the activation of phospholipase C (PLC) and subsequent release of the second messengers inositol trisphosphate (IP3) and diacylglycerol (DAG) followed by the activation of protein kinase C (PKC) and the release of [Ca2+]i (1). HRH1 is expressed in a wide variety of tissues including the gastrointestinal tract, the central nervous system, the immunological system, and the cardiovascular and genitourinary systems (25).

HRH1 is involved in a wide array of processes including thermal regulation, memory and learning, and control of the sleep-wake cycle, food intake, and emotional and aggressive behaviors. HRH1 is also involved in the pathological process of allergy, including allergic rhinitis, atopic dermatitis, anaphylaxis and asthma, and also has a role in autoimmune diseases and malignancy (27). Histamine is involved in cell proliferation, and key events in tumor development and progression have been extensively investigated (8,9). It has been found that H1R stimulates proliferation of breast carcinoma, melanoma and astrocytoma tumor cells (1012). In this context, the investigation of H1R and its antagonists support a clear rationale for future supportive anticancer therapies. However, a comprehensive investigation of whether HRH1 is involved in the formation of various tumors has not yet been carried out.

In the present study, we identified HRH1 genes from the human, chimpanzee, macaque, orangutan, dog, cow, horse, mouse, rat, opossum, chicken, Xenopus tropicalis, zebrafish and fugu by comparative genomic analyses. A search for conserved transcription factor-binding sites within the promoter regions of the human HRH1 gene was then carried out. The expression data, functional relevant single nucleotide polymorphisms (SNPs) and comparative proteomic analyses were conducted. Furthermore, meta-analysis of the prognostic value of HRH1 genes in various types of cancers was also performed.

Materials and methods

Identification of novel HRH1 genes in vertebrate genomes and integrative genomic analyses

A search for HRH1 genes was carried out in the genome sequences of the human (Homo sapiens), chimpanzee (Pan troglodytes), macaque (Macaca mulatta), orangutan (Pongo pygmaeus), dog (Canis familiaris), cow (Bos taurus), horse (Equus caballus), mouse (Mus musculus), rat (Rattus norvegicus), opossum (Monodelphis domestica), chicken (Gallus gallus), Xenopus tropicalis, zebrafish (Danio rerio) and fugu (Takifugu rubripes) by the method described before using the human HRH1 gene (CAA84380) as a query. The assemblies used were human NCBI36, chimpanzee CHIMP2.1, macaque MMUL 1.0, orangutan PPYG2, dog CanFam 2.0, cow Btau_4.0, horse Equ Cab 2, mouse NCBI m37, rat RGSC 3.4, opossum monDom5, chicken WASHUC2, X. tropicalis JGI 4.1, zebrafish Zv8 and fugu FUGU 4.0. The identified putative HRH1 genes were BLASTed against the nr database of GenBank to confirm that the best hits were HRH1 genes (1315). Conserved transcription factor-binding sites within the promoter region of the human HRH1 gene were obtained from SABiosciences’ proprietary database which combines Text Mining Application and data from the UCSC Genome Browser.

Comparative proteomic analyses of HRH1 proteins

The amino acid sequences of HRH1 were deduced from the identified HRH1 genes and aligned using Clustal X 1.8 software (16). The phylogenetic tree of HRH1 was obtained by using ML (maximum likelihood) (PHYML v2.4.4) (17) and NJ (neighbor-joining) (MEGA 3.0) (18) methods, and the reliability of the tree was evaluated by the bootstrap method with 1,000 replications. The program Codeml implemented in the PAML 3.14 b software package was used to investigate whether HRH1 proteins are under positive selection (19). Six models of codon substitution, M0 (one-ratio), M1a (NearlyNeutral), M2a (PositiveSelection), M3 (discrete), M7 (β), and M8 (β and ω) were used in the analysis (20).

Functional relevant SNP evaluation of the human HRH1 gene

Functional relevant SNPs (single nucleotide polymorphisms) of the human HRH1 gene were identified as previously described (1315,21). The SNPs were extracted from Ensembl (http://www.ensembl.org) and NCBI’s SNPdb (http://www.ncbi.nlm.nih.gov). The SNPs that could disrupt ESE/ESS (exonic splicing enhancer/exonic splicing silencer) motifs and cause missence mutation were also identified.

In silico expression analyses of the human HRH1 gene

Expressed sequence tags (ESTs) derived from the human HRH1 gene were searched for using the BLAST programs as previously described (2225). Human HRH1 gene (NM_003118) was used as query sequences for the BLAST programs. The expression profiles for normal human tissues were obtained from GeneAnnot (26) and ArrayExpress (27). Northern analysis of NCBI’s uniGene dataset was also extracted (21). Moreover, protein expression of HRH1 was obtained from SPIRE (28) and MOPED (29).

Meta-analysis of the prognostic value of the HRH1 gene in cancer

A database named ‘PrognoScan’ has been developed (30). This is i) a large collection of publicly available cancer microarray datasets with clinical annotation, as well as ii) a tool for assessing the biological relationship between gene expression and prognosis. PrognoScan employs the minimum P-value approach for grouping patients for survival analysis. PrognoScan provides a powerful platform for evaluating potential tumor markers and therapeutic targets and is publicly accessible at http://gibk21.bio.kyutech.ac.jp/PrognoScan/index.html. The human HRH1 gene was inputted as a query and the data were collected for analysis.

Results

Comparative proteomics of HRH1 proteins identified in vertebrate genomes

HRH1 genes were identified in the genome sequences of the human, chimpanzee, macaque, orangutan, dog, cow, horse, mouse, rat, opossum, chicken, Xenopus tropicalis, zebrafish and fugu. Refined phylogentic trees using the identified HRH1 protein amino acid sequences by maximum likelihood (ML) and neighbor-joining (NJ) methods were almost identical (Fig. 1). We were unable to identify any site under positive selection with any of the six models in HRH1 proteins. Instead, the HRH1 proteins were under purifying selection (data not shown).

Expression profile of the human HRH1 gene

By EST sequence searching, the human HRH1 gene was found to be expressed in stomach, liver, heart, brain, placenta, embryonic tissue, kidney, vascular, intestine, esophagus, lung, adrenal gland, mouth, connective tissue, cervix, ovary, trachea, testis, uterus and skin. The investigation of available microarray experiments and ‘virtual northern blot’ showed a predominant expression of HRH1 in the bone marrow, whole blood, lymph node, thymus, brain, cerebellum, retina, spinal cord, heart, smooth muscle, skeletal muscle, small intestine, colon, adipocyte, kidney, liver, lung, pancreas, thyroid salivary gland, skin, ovary, uterus, placenta, prostate and testis. When searched in PrognoScan database, human HRH1 was also found to be expressed in bladder cancer, blood cancer, brain cancer, breast cancer, colorectal cancer, eye cancer, head and neck cancer, lung cancer, ovarian cancer, skin cancer and soft tissue cancer tissues. Among the protein expression databases SPIRE and MOPED, HRH1 protein was found to be only highly expressed in blood plasma.

Comparative genomics on the human HRH1 gene

Glucocorticoid receptor (GR), germ cell nuclear factor (GCNF), signal transducer and activator of transcription 5A (STAT5A), mini Zinc finger 1 (MIF-1), heat shock transcription factor 2 (HSF2) and c-Myb regulatory transcription factor binding sites were identified in the HRH1 gene upstream (promoter) region.

Functional relevant SNP evaluation of the human HRH1 gene

Available SNPs (2,455) were identified in the human HRH1 gene. Among these, 88 SNPs were functionally relevant; including 4 available alleles disrupting an existing exonic splicing enhancer and 84 SNPs causing missense mutation (Table I).

Table I

Functional relevant SNP evaluation of the human HRH1 gene.

Table I

Functional relevant SNP evaluation of the human HRH1 gene.

SNP IDChr 3 positionSequenceTypeAA changeSNP IDChr 3 positionSequenceTypeAA change
rs7931445011300765(+)AAGATG/ATGTGAMissense/I/Mrs19952299411301463(+)GGGGGA/TTGCCAMissenseD V
rs206746611300780(+)AACAAG/CACCACMissense/N/Krs19952845211301826(+)CACAGA/GGACAGMissenseE G
rs6173899011300881(+)CGTACG/AGAGTGMissense/Q/Rrs20078773511301096(+)TTGATA/CGCTACMissenseS R
rs18937605111300922(+)ACATCA/GTCAGCMissenseI Vrs19972056411301138(+)AGTATC/TGTACCMissenseR C
rs20057106711300958(+)GTGCCA/GTCGTCMissenseI Vrs13866940411300787(+)CCACTA/GTGGCCMissenseMV
rs13966445111301013(+)GGGCCG/ATCCTCMissense/H/Rrs11163289411301870(+)ACACAG/TGCCTGMissenseGC
rs14644134711301075(+)TCAGTG/ATCTTCMissense/I/Vrs14851113811300908(+)TGTGGG/AGAACCMissense/E/G
rs18178301811301151(+)GACCCA/GAGCCTMissenseQ Rrs20033590711301073(+)TTTCAA/GTGTCTMissenseN S
rs20194742411301246(+)CGGTGC/TGCCGAMissenseR Crs20141729811301732(+)AGCAGA/GGCCTGMissenseS G
rs13812099811301309(+)CTGCCA/GTCATCMissenseI Vrs20148030311301124(+)CCTCAG/TGTACCMissenseR M
rs20157563311301362(+)AAGATC/GTACAAMissenseI Mrs19984342111300782(+)CAAGAC/GCACTAMissenseT S
rs14593809811301394(+)GCACCG/AGGAGCMissense/Q/Rrs20146932611301462(+)AGGGGA/GATGCCMissenseND
rs765162011301532(+)TGGTGG/AATCTGMissense/E/Grs20193958811301699(+)TCAACC/TGGAGCMissenseRW
rs20121054711301661(+)GGCTGC/TGGCAGMissenseA Vrs20055383011301854(+)TTGAGC/GAGTGGMissenseS R
rs14968392711301678(+)GTAGCA/GGGGACMissenseR Grs19972056411301138(+)AGTATC/TGTACCMissenseR C
rs14765273111301693(+)TAGCCG/ATCAACMissense/I/Vrs13866940411300787(+)CCACTA/GTGGCCMissenseM V
rs7484080011301700(+)CAACCG/AGAGCCMissense/Q/Rrs11163289411301870(+)ACACAG/TGCCTGMissenseG C
rs18928777611301768(+)CAGAGC/GATCAGMissenseH Drs20069558411300808(+)TGATGC/TCCCTGMissenseP S
rs206746711301770(+)GAGGAT/GCAGATMissense/E/Drs14851113811300908(+)TGTGGG/AGAACCMissense/E/G
rs14191496611301810(+)CGGACT/CCAGATMissense/P/Srs20141729811301732(+)AGCAGA/GGCCTGMissenseS G
rs18083199711301858(+)GGAGTA/GGGTCTMissenseR Grs20148030311301124(+)CCTCAG/TGTACCMissenseR M
rs19987575111301988(+)CATGGC/TAGCCTMissenseA Vrs19984342111300782(+)CAAGAC/GCACTAMissenseT S
rs206747011302069(+)ACATTT/CGCACAMissense/S/Lrs20146932611301462(+)AGGGGA/GATGCCMissenseN D
rs7668910211302092(+)GGCTGG/TGCTACMissenseG Crs20193958811301699(+)TCAACC/TGGAGCMissenseR W
rs20202505511301685(+)GGACTA/GTGTAGMissenseY Crs20055383011301854(+)TTGAGC/GAGTGGMissenseS R
rs14958237811301513(+)GGAAGC/GCAAAAMissenseP Ars20060652411300890(+)TGAGCA/GGAAGCMissenseQ R
rs20105286911301024(+)TCTGCC/GTCTTTMissenseL Vrs19995461811301561(+)AAACCC/GCCAAGMissenseP A
rs20117584111301184(+)GTTTCC/TCTCTTMissenseP Lrs20206466911302032(+)TCATGG/TTCATTMissenseV F
rs20178574911301523(+)AGATGA/CTGGTGMissenseD Ars20090322011301886(+)CATCAA/GGTTTAMissenseK R
rs20121778211300932(+)CCTCTC/TGGTGGMissenseS Lrs20210054911302102(+)CATCAA/GCTCCAMissenseN S
rs20008690611301876(+)GCCTGA/GATTACMissenseN Drs20089276511302020(+)ATTTCA/GTCTTCMissenseI V
rs19963808211301796(+)ATCCTC/TCTCTCMissenseS Frs20145041611301389(+)TGCCAG/TCACCGMissenseQ H
rs20105300111301951(+)ACCGCA/GAAAGGMissenseK Ers20110119111301348(+)TCTGGC/TTCTATMissenseL F
rs20158666711301627(+)TCTACA/TGCTTTMissenseS Crs20172086311300901(+)TCCACA/GCTGTGMissenseT A
rs19982253211300889(+)GTGAGC/TGGAAGMissenseR Wrs13850131011301910(+)CCGCTC/TGCATTMissenseS L
rs20138665811301550(+)GTCACC/TATCCCMissenseP Lrs19964563111300863(+)CCTGCG/TGGTGCMissenseR L
rs14388299511300977(+)GAACAT/CCCTCTMissense/T/Irs20006663811301247(+)GGTGCA/GCCGAGMissenseH R
rs20081790911301139(+)GTATCA/GTACCAMissenseH Rrs20166127811301150(+)AGACCC/GGAGCCMissenseR G
rs20190312311301907(+)GCTCCA/GCTCGCMissenseH Rrs11352678611300934(+)TCTCGG/ATGGCGMissense/M/V
rs1785503411301646(+)TATTGA/TGCACAMissenseE Vrs20052611511302035(+)TGGTCA/GTTGCCMissenseI V
rs19982415411301171(+)TTCTGC/GGGGCCMissenseR Grs20089787011301538(+)ATCTGC/TCTTGAMissenseA V
rs20054776811301776(+)CAGATG/TTTAGGMissenseM I

[i] Available SNPs (2,455) were identified in the human HRH1 gene. Here, we show the 84 SNPs causing a missense mutation. AA change, amino acid change (slashes ‘/’ indicate uncoded amino acids).

Meta-analysis of the prognostic value of the human HRH1 gene in cancer

When given the gene, PrognoScan displays a summary in table format of tests for the gene with columns for dataset, cancer type, subtype, endpoint, cohort, contributor, array type, probe ID, number of patients, optimal cut-point, Pmin and Pcor. Among the databases which detected the expression of the HRH1 gene, 23 out of 153 tests showed an association between microarray expression in the HRH1 gene and cancer prognosis (bladder cancer 2/4, blood cancer 2/18, brain cancer 1/9, breast cancer 6/50, colorectal cancer 1/18, eye cancer 1/2, head and neck cancer 0/2, lung cancer 6/29, ovarian cancer 2/18, prostate cancer 0/1, skin cancer 0/2 and soft tissue cancer 2/2) with a 5% level of significance (Table II). Among the 6 breast cancer cases, low expression of the HRH1 gene was related to poor survival in 3 cases (GSE12276, GSE9893 and GSE1456-GPL96). However, high expression of the HRH1 gene was related to poor survival in another 3 cases of breast cancer (GSE6532-GPL570 and GSE3494-GPL96). As for the lung cancer cases, we found that high expression of the HRH1 gene was associated with poor survival in all 6 lung cancer cases including 5 adenocarcinoma cases and 1 non-small cell lung cancer case (GSE8894). Among the 2 blood cancer cases, high expression of the HRH1 gene was related to poor survival in the B-cell lymphoma case (GSE4475). However, in 1 multiple myeloma case (GSE2658), low expression of the HRH1 gene was related to poor survival. We found that high expression of the HRH1 gene was associated with poor survival in 2 cases of soft tissue cancer and in 1 case of brain cancer and colorectal cancer. In the other cancer cases, low expression of the HRH1 gene was associated with poor survival in 2 cases of bladder cancer and ovarian cancer and 1 case of eye cancer.

Table II

Dataset content from PrognoScan indicated an association between microarray expression of HRH1 and cancer prognosis.

Table II

Dataset content from PrognoScan indicated an association between microarray expression of HRH1 and cancer prognosis.

DatabaseCase typeSubtypeNo. of patientsEndpointCutpointP-valuePrognosisRefs.
GSE13507Bladder cancer165Overall survival0.250.0121(41,42)
GSE13507Bladder cancerTransitional cell carcinoma165Disease-specific survival0.280.0091(41,42)
GSE4475Blood cancerB-cell lymphoma158Overall survival0.760.0272(43)
GSE2658Blood cancerMultiple myeloma559Disease-specific survival0.670.0021(44)
GSE4271-GPL96Brain cancerAstrocytoma77Overall survival0.390.042(45)
GSE12276Breast cancer204Relapse-free survival0.880.0471(46)
GSE6532-GPL570Breast cancer87Distant metastasis-free survival0.860.012(47)
GSE6532-GPL570Breast cancer87Relapse-free survival0.860.012(47)
GSE9893Breast cancer155Overall survival0.290.0121(48)
GSE1456-GPL96Breast cancer159Relapse-free survival0.110.0071(49)
GSE3494-GPL96Breast cancer249Relapse-free survival0.80.00142(50)
GSE17536Colorectal cancer145Relapse-free survival0.90.0432(51)
GSE22138Eye cancerUveal melanoma63Relapse-free survival0.220.0171(52)
jacob-00182-CANDFLung cancerAdenocarcinoma82Relapse-free survival0.770.0472 (53)
GSE13213Lung cancerAdenocarcinoma117Relapse-free survival0.550.0462(54)
GSE31210Lung cancerAdenocarcinoma204Relapse-free survival0.680.032(55)
GSE31210Lung cancerAdenocarcinoma204Relapse-free survival0.850.0182(55)
jacob-00182-UMLung cancerAdenocarcinoma178Relapse-free survival0.680.022 (53)
GSE8894Lung cancerNon-small cell lung cancer138Relapse-free survival0.780.0362 (56)
GSE17260Ovarian cancer110Relapse-free survival0.880.0481(57)
GSE14764Ovarian cancer80Relapse-free survival0.120.011(57)
GSE30929Soft tissue cancerLiposarcoma140Relapse-free survival0.860.0012(58)
GSE30929Soft tissue cancerLiposarcoma140Relapse-free survival0.880.032(58)

[i] Out of 153 tests, 23 showed an association between microarray expression of HRH1 gene and cancer prognosis (bladder cancer 2/4, blood cancer 2/18, brain cancer 1/9, breast cancer 6/50, colorectal cancer 1/18, eye cancer 1/2, head and neck cancer 0/2, lung cancer 6/29, ovarian cancer 2/18, prostate cancer 0/1, skin cancer 0/2 and soft tissue cancer 2/2) with a 5% significance level.

Discussion

A large number of molecules involved in cell proliferation and key events in tumor development and progression have been extensively investigated including histamine (3133). Histamine exerts its functions through binding to G protein-associated histamine H1, H2, H3, and H4 receptors (HRH1, HRH2, HRH3 and HRH4), resulting in the activation of different signal transduction pathways (3133). In addition to the human HRH1, in the present study, we identified other HRH1 genes from other 13 vertebrate genomes and found that HRH1 exists in all types of vertebrates including the fish, amphibians, birds and mammals. All the identified HRH1 genes were encoding for a GPCR protein with a long third intracellular loop (IL3). The phylogenetic tree showed that HRH1 is separated for the order: fish, amphibians, birds and mammals. Primate HRH1s are almost the same and are clustered together. From the alignment and phylogenetic tree, mammalian HRH1s are conserved among the vertebrate genomes, suggesting that the function of HRH1 may have played an important physiological role for all vertebrates in the long process of evolution. Moreover, this process was under purifying selection, in accordance with the fact that multiple biological functions have been ascribed to this protein, including its function in central and peripheral tissues.

Histamine is an endogenous biogenic amine widely distributed throughout the organism and is long known to be a pleiotropic mediator in different (patho)-physiological conditions (1). Accordingly, by EST sequence search, one of its receptors, the HRH1 gene is also expressed in various tissues and cells including stomach, liver, heart, brain, placenta, embryonic tissue, kidney, vascular, intestine, esophagus, lung, adrenal gland, mouth, connective tissue, cervix, ovary, trachea, testis, uterus and skin. The same results were confirmed by the investigation of available microarray experiments and ‘virtual northern blot’, which showed a predominant expression of HRH1 in the bone marrow, whole blood, lymph node, thymus, brain, cerebellum, retina, spinal cord, heart, smooth muscle, skeletal muscle, small intestine, colon, adipocyte, kidney, liver, lung, pancreas, thyroid salivary gland, skin, ovary, uterus, placenta, prostate and testis. Available SNPs (2,455) were identified in the human HRH1 gene. We identified 88 SNPs including 4 available alleles disrupting an existing exonic splicing enhancer and 84 SNPs causing a missense mutation, which may impact the effect of histamine on the HRH1 protein. However, the effects of these SNPs on the physiological and pathological functions of HRH1 warrant further investigation.

Notably, most malignant cell lines and experimental tumors contain a high concentration of endogenous histamine, which can be released to the extracellular media via a paracrine or autocrine pathway. Moreover, histamine itself may be a crucial mediator in cancer development and progression by regulating diverse biological responses related to tumor growth including angiogenesis, cell invasion, migration, differentiation, apoptosis and modulation of the immune response (3133). As one receptor of histamine, HRH1 expression in normal tissues vs. cancer samples has not been well studied. Following a search in the PrognoScan database, human HRH1 was also found to be expressed in bladder cancer, blood cancer, brain cancer, breast cancer, colorectal cancer, eye cancer, head and neck cancer, lung cancer, ovarian cancer, skin cancer and soft tissue cancer tissues. The expression of HRH1 in these tumors may explain the effects of histamine on cancer behaviors. Out of 153, 23 tests showed an association between microarray expression in the HRH1 gene and cancer prognosis (bladder cancer 2/4, blood cancer 2/18, brain cancer 1/9, breast cancer 6/50, colorectal cancer 1/18, eye cancer 1/2, head and neck cancer 0/2, lung cancer 6/29, ovarian cancer 2/18, prostate cancer 0/1, skin cancer 0/2 and soft tissue cancer 2/2) with a 5% level of significance. This suggests that the expression of HRH1 is related to the prognosis of many types of cancers including hematological and solid cancers. The mechanism of HRH1 involved in the process of these tumors warrants further investigation. It is important to note that the relationship between the expression of HRH1 and prognosis varied in different cancers, even in the same cancer from different databases. This implies that the function of HRH1 in these tumors may be multidimensional, not just as a tumor suppressor or oncogene.

The GR, GCNF, STAT5A, MIF-1, HSF2 and c-Myb regulatory transcription factor binding sites were identified in the HRH1 gene upstream (promoter) region. GRs are nuclear hormone receptors of the NR3C class, which also includes mineralocorticoid, progesterone and androgen receptors. GRs are ubiquitously expressed and mediate a vast array of physiological functions. Glucocorticoids are widely used for the treatment of lymphoid malignancy due to their marked effects on cell cycle progression and apoptosis (34). Glucocorticoids are able to alter signaling in key survival pathways and this can result in reversible growth arrest or cell death in certain cell types (3436). STAT5A is a member of the STAT family of transcription factors. It is activated by, and mediates the responses of many cell ligands, such as IL2, IL3, IL7, GM-CSF, erythropoietin, thrombopoietin and different growth hormones. Activation of this protein in many types of cancers has been shown to be essential for tumorigenesis (37,38). C-Myb encodes a transcription factor that regulates the expression of numerous genes during cell cycle progression. c-Myb is a cellular proto-oncogene, which has the ability to regulate the expression of cell cycle genes and is involved in cell proliferation and carcinogenesis (39,40). These tumor-related transcriptional factors may be involved in the effect of HRH1 in tumors.

Acknowledgements

This study was sponsored by the Chinese National Natural Science Foundation (81372828, 81302331, 810001329), The Talent Foundation of The First Affiliated Yijishan Hospital of Wannan Medical College (YR201305), the Natural Science Foundation of Guangdong Province (no. 10151503102000045), the National Major Scientific and Technological Special Project for ‘Significant New Drug Development’ (2011ZX09302-003-02), the Jiangsu Province Major Scientific and Technological Special Project (BM2011017), and a Project Funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions.

References

1 

Seifert R, Strasser A, Schneider EH, Neumann D, Dove S and Buschauer A: Molecular and cellular analysis of human histamine receptor subtypes. Trends Pharmacol Sci. 34:33–58. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Yoshizawa M, Tashiro M, Fukudo S, et al: Increased brain histamine H1 receptor binding in patients with anorexia nervosa. Biol Psychiatry. 65:329–335. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Booth RG, Fang L, Wilczynski A, et al: Molecular determinants of ligand-directed signaling for the histamine H1 receptor. Inflamm Res. 57(Suppl 1): S43–S44. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Swan C, Richards SA, Duroudier NP, Sayers I and Hall IP: Alternative promoter use and splice variation in the human histamine H1 receptor gene. Am J Respir Cell Mol Biol. 35:118–226. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Oda T, Morikawa N, Saito Y, Masuho Y and Matsumoto S: Molecular cloning and characterization of a novel type of histamine receptor preferentially expressed in leukocytes. J Biol Chem. 275:36781–36788. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Breunig E, Michel K, Zeller F, Seidl S, Weyhern CW and Schemann M: Histamine excites neurones in the human submucous plexus through activation of H1, H2, H3 and H4 receptors. J Physiol. 583:731–742. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Brew OB and Sullivan MH: Localisation of mRNAs for diamine oxidase and histamine receptors H1 and H2, at the feto-maternal interface of human pregnancy. Inflamm Res. 50:449–452. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Arisawa T, Tahara T, Ozaki K, et al: Association between common genetic variant of HRH2 and gastric cancer risk. Int J Oncol. 41:497–503. 2012.

9 

Lefranc F, Yeaton P, Brotchi J and Kiss R: Cimetidine, an unexpected anti-tumor agent, and its potential for the treatment of glioblastoma. Int J Oncol. 28:1021–1030. 2006.PubMed/NCBI

10 

Davio C, Baldi A, Mladovan A, Cricco G, Fitzsimons C and Bergoc RR: Expression of histamine receptors in different cell lines derived from mammary gland and human breast carcinomas. Inflamm Res. 44(Suppl 1): S70–S71. 1995. View Article : Google Scholar : PubMed/NCBI

11 

Jiang CG, Liu FR, Yu M, Li JB and Xu HM: Cimetidine induces apoptosis in gastric cancer cells in vitro and inhibits tumor growth in vivo. Oncol Rep. 23:693–700. 2010.PubMed/NCBI

12 

Fukuda M, Tanaka S, Suzuki S, Kusama K, Kaneko T and Sakashita H: Cimetidine induces apoptosis of human salivary gland tumor cells. Oncol Rep. 17:673–678. 2007.PubMed/NCBI

13 

Yang L, Luo Y and Wei J: Integrative genomic analyses on Ikaros and its expression related to solid cancer prognosis. Oncol Rep. 24:571–577. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Yang L, Luo Y, Wei J and He S: Integrative genomic analyses on IL28RA, the common receptor of interferon-λ1, -λ2 and -λ3. Int J Mol Med. 25:807–812. 2010.PubMed/NCBI

15 

Yang L, Wei J and He S: Integrative genomic analyses on interferon-λs and their roles in cancer prediction. Int J Mol Med. 25:299–304. 2010.

16 

Thompson JD, Gibson TJ, Plewniak F, Jeanmougin F and Higgins DG: The CLUSTAL-X windows interface: flexible strategies for multiple sequence alignment aided by quality analysis tools. Nucleic Acids Res. 15:4876–4882. 1997. View Article : Google Scholar : PubMed/NCBI

17 

Guindon S, Lethiec F, Duroux P and Gascuel O: PHYML Online - a web server for fast maximum likelihood-based phylogenetic inference. Nucleic Acids Res. 33:W557–W559. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Kumar S, Tamura K and Nei M: MEGA3: Integrated software for molecular evolutionary genetics analysis and sequence alignment. Brief Bioinform. 5:150–163. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Yang Z: PAML: a program package for phylogenetic analysis by maximum likelihood. Comput Appl Biosci. 13:555–556. 1997.PubMed/NCBI

20 

Yang Z, Nielsen R, Goldman N and Pedersen AM: Codon-substitution models for heterogeneous selection pressure at amino acid sites. Genetics. 155:431–449. 2000.PubMed/NCBI

21 

Yu H, Yuan J, Xiao C and Qin Y: Integrative genomic analyses of recepteur d’origine nantais and its prognostic value in cancer. Int J Mol Med. 31:1248–1254. 2013.

22 

Katoh Y and Katoh M: Integrative genomic analyses on GLI1: positive regulation of GLI1 by Hedgehog-GLI, TGFβ-Smads, and RTK-PI3K-AKT signals, and negative regulation of GLI1 by Notch-CSL-HES/HEY, and GPCR-Gs-PKA signals. Int J Oncol. 35:187–192. 2009.

23 

Katoh M and Katoh M: Integrative genomic analyses of WNT11: transcriptional mechanisms based on canonical WNT signals and GATA transcription factors signaling. Int J Mol Med. 24:247–251. 2009.

24 

Katoh M and Katoh M: Transcriptional mechanisms of WNT5A based on NF-κB, Hedgehog, TGFβ, and Notch signaling cascades. Int J Mol Med. 23:763–769. 2009.

25 

Katoh M and Katoh M: Integrative genomic analyses of ZEB2: Transcriptional regulation of ZEB2 based on SMADs, ETS1, HIF1α, POU/OCT, and NF-κB. Int J Oncol. 34:1737–1742. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Chalifa-Caspi V, Yanai I, Ophir R, et al: GeneAnnot: comprehensive two-way linking between oligonucleotide array probesets and GeneCards genes. Bioinformatics. 20:1457–1458. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Parkinson H, Sarkans U, Shojatalab M, et al: ArrayExpress - a public repository for microarray gene expression data at the EBI. Nucleic Acids Res. 33:D553–D555. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Kolker E, Higdon R, Morgan P, et al: SPIRE: Systematic protein investigative research environment. J Proteomics. 75:122–126. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Kolker E, Higdon R, Haynes W, et al: MOPED: Model Organism Protein Expression Database. Nucleic Acids Res. 40:D1093–D1099. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Mizuno H, Kitada K, Nakai K and Sarai A: PrognoScan: a new database for meta-analysis of the prognostic value of genes. BMC Med Genomics. 2:182009. View Article : Google Scholar : PubMed/NCBI

31 

Xu TR, Yang Y, Ward R, Gao L and Liu Y: Orexin receptors: multi-functional therapeutic targets for sleeping disorders, eating disorders, drug addiction, cancers and other physiological disorders. Cell Signal. 25:2413–2423. 2013. View Article : Google Scholar

32 

Medina VA and Rivera ES: Histamine receptors and cancer pharmacology. Br J Pharmacol. 61:755–767. 2010. View Article : Google Scholar

33 

Blaya B, Nicolau-Galmés F, Jangi SM, et al: Histamine and histamine receptor antagonists in cancer biology. Inflamm Allergy Drug Targets. 9:146–157. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Schlossmacher G, Stevens A and White A: Glucocorticoid receptor-mediated apoptosis: mechanisms of resistance in cancer cells. J Endocrinol. 211:17–25. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Feng Y, Bai X, Yang Q, Wu H and Wang D: Downregulation of 15-lipoxygenase 2 by glucocorticoid receptor in prostate cancer cells. Int J Oncol. 36:1541–1549. 2010.PubMed/NCBI

36 

Jang JH, Woo SM, Um HJ, et al: RU486, a glucocorticoid receptor antagonist, induces apoptosis in U937 human lymphoma cells through reduction in mitochondrial membrane potential and activation of p38 MAPK. Oncol Rep. 30:506–512. 2013.

37 

Koptyra M, Gupta S, Talati P and Nevalainen MT: Signal transducer and activator of transcription 5a/b: biomarker and therapeutic target in prostate and breast cancer. Int J Biochem Cell Biol. 43:1417–1421. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Hou L, Xu B, Mohankumar KM, Goffin V, Perry JK, Lobie PE and Liu DX: The prolactin receptor mediates HOXA1-stimulated oncogenicity in mammary carcinoma cells. Int J Oncol. 41:2285–2295. 2012.PubMed/NCBI

39 

Haeri M, Li Y, Li Y, Li Q, Spaner DE and Ben-David Y: Insertional activation of myb by F-MuLV in SCID mice induces myeloid leukemia. Int J Oncol. 43:169–176. 2013.PubMed/NCBI

40 

Zhang J, Luo N, Luo Y, Peng Z, Zhang T and Li S: microRNA-150 inhibits human CD133-positive liver cancer stem cells through negative regulation of the transcription factor c-Myb. Int J Oncol. 40:747–756. 2012.PubMed/NCBI

41 

Kim WJ, Kim EJ, Kim SK, et al: Predictive value of progression-related gene classifier in primary non-muscle invasive bladder cancer. Mol Cancer. 9:32010. View Article : Google Scholar : PubMed/NCBI

42 

Lee JS, Leem SH, Lee SY, et al: Expression signature of E2F1 and its associated genes predict superficial to invasive progression of bladder tumors. J Clin Oncol. 28:2660–2667. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Hummel M, Bentink S, Berger H, et al: Molecular Mechanisms in Malignant Lymphomas Network Project of the Deutsche Krebshilfe. A biologic definition of Burkitt’s lymphoma from transcriptional and genomic profiling. N Engl J Med. 354:2419–2430. 2006.PubMed/NCBI

44 

Zhan F, Huang Y, Colla S, et al: The molecular classification of multiple myeloma. Blood. 108:2020–2028. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Phillips HS, Kharbanda S, Chen R, et al: Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 9:157–173. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Bos PD, Zhang XH, Nadal C, et al: Genes that mediate breast cancer metastasis to the brain. Nature. 459:1005–1009. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Loi S, Haibe-Kains B, Majjaj S, et al: PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor-positive breast cancer. Proc Natl Acad Sci USA. 107:10208–10213. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Chanrion M, Negre V, Fontaine H, et al: A gene expression signature that can predict the recurrence of tamoxifen-treated primary breast cancer. Clin Cancer Res. 14:1744–1752. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Pawitan Y, Bjöhle J, Amler L, et al: Gene expression profiling spares early breast cancer patients from adjuvant therapy: derived and validated in two population-based cohorts. Breast Cancer Res. 7:R953–R964. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Miller LD, Smeds J, George J, et al: An expression signature for p53 status in human breast cancer predicts mutation status, transcriptional effects, and patient survival. Proc Natl Acad Sci USA. 102:13550–13555. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Smith JJ, Deane NG, Wu F, et al: Experimentally derived metastasis gene expression profile predicts recurrence and death in patients with colon cancer. Gastroenterology. 138:958–968. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Laurent C, Valet F, Planque N, et al: High PTP4A3 phosphatase expression correlates with metastatic risk in uveal melanoma patients. Cancer Res. 71:666–674. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Director’s Challenge Consortium for the Molecular Classification of Lung Adenocarcinoma. Shedden K, Taylor JM, et al: Gene expression-based survival prediction in lung adenocarcinoma: a multi-site, blinded validation study. Nat Med. 14:822–827. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Tomida S, Takeuchi T, Shimada Y, et al: Relapse-related molecular signature in lung adenocarcinomas identifies patients with dismal prognosis. J Clin Oncol. 27:2793–2799. 2009. View Article : Google Scholar : PubMed/NCBI

55 

Okayama H, Kohno T, Ishii Y, et al: Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 72:100–111. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Lee ES, Son DS, Kim SH, et al: Prediction of recurrence-free survival in postoperative non-small cell lung cancer patients by using an integrated model of clinical information and gene expression. Clin Cancer Res. 14:7397–7404. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Yoshihara K, Tajima A, Yahata T, et al: Gene expression profile for predicting survival in advanced-stage serous ovarian cancer across two independent datasets. PLoS One. 5:e96152010. View Article : Google Scholar : PubMed/NCBI

58 

Gobble RM, Qin LX, Brill ER, et al: Expression profiling of liposarcoma yields a multigene predictor of patient outcome and identifies genes that contribute to liposarcomagenesis. Cancer Res. 71:2697–2705. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

2014-April
Volume 33 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang M, Wei X, Shi L, Chen B, Zhao G and Yang H: Integrative genomic analyses of the histamine H1 receptor and its role in cancer prediction. Int J Mol Med 33: 1019-1026, 2014
APA
Wang, M., Wei, X., Shi, L., Chen, B., Zhao, G., & Yang, H. (2014). Integrative genomic analyses of the histamine H1 receptor and its role in cancer prediction. International Journal of Molecular Medicine, 33, 1019-1026. https://doi.org/10.3892/ijmm.2014.1649
MLA
Wang, M., Wei, X., Shi, L., Chen, B., Zhao, G., Yang, H."Integrative genomic analyses of the histamine H1 receptor and its role in cancer prediction". International Journal of Molecular Medicine 33.4 (2014): 1019-1026.
Chicago
Wang, M., Wei, X., Shi, L., Chen, B., Zhao, G., Yang, H."Integrative genomic analyses of the histamine H1 receptor and its role in cancer prediction". International Journal of Molecular Medicine 33, no. 4 (2014): 1019-1026. https://doi.org/10.3892/ijmm.2014.1649