Open Access

Association between HO‑1 gene promoter polymorphisms and diseases (Review)

  • Authors:
    • Lin-Lin Ma
    • Lei Sun
    • Yu-Xi Wang
    • Bai-He Sun
    • Yan-Fei Li
    • Yue-Ling Jin
  • View Affiliations

  • Published online on: November 25, 2021     https://doi.org/10.3892/mmr.2021.12545
  • Article Number: 29
  • Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Heme oxygenase‑1 (HO‑1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. These degradation products serve an important role in the regulation of inflammation, oxidative stress and apoptosis. While the expression level of HO‑1 is typically low in most cells, it may be highly expressed when induced by a variety of stimulating factors, a process that contributes to the regulation of cell homeostasis. In the 5'‑non‑coding region of the HO‑1 gene, there are two polymorphic sites, namely the (GT)n dinucleotide and T(‑413)A single nucleotide polymorphism sites, which regulate the transcriptional activity of HO‑1. These polymorphisms have been shown to be closely associated with the occurrence and progression of numerous diseases, including cardiovascular, pulmonary, liver and kidney, various types of cancer and viral diseases. The present article reviews the progress that has been made in research on the association between the two types of polymorphisms and these diseases, which is expected to provide novel strategies for the diagnosis, treatment and prognosis of various diseases.

Introduction

Heme oxygenase-1 (HO-1), also known as heat shock protein 32, is the rate-limiting enzyme in the heme degradation pathway that degrades hemoglobin into biliverdin, free iron and carbon monoxide (CO) (1). Biliverdin is subsequently reduced to bilirubin via biliverdin reductase. Both biliverdin and bilirubin are bile pigments with antioxidant properties (2). Endogenous CO can act as a second messenger, thereby affecting a variety of physiological and pathological processes, including cell proliferation, inflammation, apoptosis and injury (3). As a cytoprotective enzyme, HO-1 serves an important role in maintaining cell homeostasis.

To date, three HO polymorphic isoenzymes have been identified, namely HO-1, HO-2 and HO-3, of which HO-1 is inducible. The HO-1-encoding gene is located on the long arm of chromosome 22q12 in the human body, with a total length of 6.8 kb (4). HO-1 is expressed in a variety of tissues, including the liver, spleen, kidney, heart, lung, brain, blood vessels, smooth muscle cells and endothelial cells, among which the highest expression is observed in the liver, spleen and kidney (5,6). Under normal physiological conditions, the expression of HO-1 is relatively low in most tissues. However, under various pathophysiological stress or stimulation conditions, such as hypoxia, ultraviolet light, inflammatory mediators, heme, ischemia and other harmful stimuli, HO-1 expression is induced to protect cells against oxidative and inflammatory damage (7,8). In the presence of the aforementioned stimulatory factors, the induced expression of HO-1 is primarily affected by redox-sensitive transcription factors, including nuclear factor erythroid 2-related factor 2 (Nrf2), activator protein 1 (AP1), hypoxia-inducible factor (HIF) and BTB and CNC homology 1 (Bach1) (9). In addition, a large number of epidemiological genetic studies have shown that the expression of HO-1 is regulated by genetic polymorphisms in the promoter of its gene, which might be associated with a variety of diseases, such as coronary atherosclerosis, rheumatoid arthritis and viral diseases (1013).

The present review focuses on the clinical evidence that HO-1 gene polymorphisms are associated with the incidence and outcome of different diseases. To further understand this association, a brief discussion of the regulation of HO-1-induced expression mediated by transcription factors is included.

Regulation of HO-1 expression

As a stress-induced cytoprotective enzyme, the investigation of the molecular mechanism underlying HO-1 regulation began with studies on how various stress stimulation conditions, including stress, hypoxia and small-molecule inducers, could influence the activity of the enzyme. Under these stress conditions, the expression of HO-1 is primarily regulated by redox-sensitive transcription factors, including Nrf2, AP1, HIF and Bach1. The present review discusses the mechanisms through which each of these factors regulate HO-1.

Nrf2 is considered to be the primary transcription factor that regulates the expression of HO-1 (14). Under basic conditions, Nrf2 can form a protein complex with Kelch-like ECH-associated protein 1 (KEAP1) in the cytoplasm, which inhibits the activity of Nrf2 via proteasome-mediated degradation of Nrf2 (15,16). When cells are exposed to chemical or physical stimuli, Nrf2 separates from KEAP1 and transfers to the nucleus, where it binds to MAF bZIP transcription factor (Maf). The Nrf2/Maf heterodimers bind the antioxidant response element (ARE), leading to the transcriptional activation of antioxidant genes, including the HO-1 gene (17,18). Bach1, a heme sensor, acts as a major transcriptional repressor of HO-1 expression. Bach1 impairs the DNA-binding activity of Nrf2 by competing with Nrf2 for binding to the AREs of numerous antioxidant genes under basal conditions. High levels of heme in cells can cause Bach1 to dissociate from AREs, leading to nuclear export and subsequent proteasome-mediated degradation of Bach1. Under these circumstances, transcriptional expression of HO-1 occurs (19,20). In general, in response to various stress stimuli, the feedback loop formed by Nrf2, KEAP1 and Bach1 has a pivotal role in regulating the expression of HO-1.

Other transcriptional factors have been shown to be involved in the regulation of HO-1 expression, including HIF-1, AP1 and NF-κB (21,22). Similarly to Nrf2, these factors can combine with the cis-regulatory element of the HO-1 gene, resulting in an increase in HO-1 expression (21). HIF-1, a redox-responsive transcription factor, induces HO-1 expression in different cell types under hypoxic conditions (2326). However, a specific mutant of HIF-1 lost its DNA-binding activity, which resulted in inhibition of HO-1 expression (27). Mechanistic studies have shown that HIF-1α/HIF-1β heterodimers are necessary for hypoxia-induced HO-1 expression, since hypoxia induces the expression of HIF-1α, which binds to specific DNA sequences dependent on HIF-1β (27). AP1 transcriptional factor is generated by dimers of the Jun and Fos family proteins. AP1 is able to bind to tetradecanoylphorbol-13-acetate-responsive elements of the HO-1 gene promoter (28,29), subsequently leading to the upregulation of HO-1 expression, which has been demonstrated by functional studies and sequence analysis of the promoter (21,3032). Similarly to AP1, the NF-κB family of transcription factors are dimerization complex factors formed by REL proto-oncogene, NF-κB subunit family proteins. NF-κB homodimers comprising p52 and p50 are transcription repressors, whereas NF-κB heterodimers formed by p65 and p50 act primarily as transcription activators (21,33). AP1 and NF-κB have been shown to regulate HO-1 expression primarily in immune cells, thereby fulfilling an important role in protecting against inflammation-mediated damage, including that mediated by reactive oxygen species (34).

In addition to the direct regulation mediated by a range of transcription activators, a large number of HO-1 inducers are also able to indirectly activate the expression of the protein through acting on the protein kinase signaling pathway. Numerous plant-derived polyphenols have been found to induce HO-1 gene expression by activating the MAPK, PI3K/AKT and AMP-activated protein kinase (AMPK) signaling pathways (3537). For example, salvianolic acid A, a plant-derived polyphenol that has been widely studied, provides significant protection in several cell types following exposure to different stimuli by activating the PI3K/Akt/mTOR complex 1 or MAPK ERK/p38 signaling pathways (3840). Quercetin, another polyphenol antioxidant, activates the MAPK signaling pathway, thereby inducing the expression of HO-1, which provides protection against apoptosis and inflammation (41,42). Additionally, plant-derived polyphenols have been reported to show antioxidative stress and anti-inflammatory properties via AMPK-dependent induction of HO-1 (43,44). The precise mechanism by which the protein kinase signaling pathway induces the expression of HO-1 involves the cross-talk of transcription factors, including Nrf2, AP1 and HIF-1, and has been previously reviewed in detail (21,34). In general, the induced expression of HO-1 is an adaptive strategy that enables cells to achieve self-protection under stress conditions. However, a variety of genetic epidemiological studies have shown that there are two important genetic polymorphisms in the promoter region of the HO-1 gene (4547). These polymorphisms determine the expression level of HO-1 and affect several diseases, such as cardiovascular, pulmonary, liver, renal, cancer and viral diseases.

HO-1 promoter polymorphism

HO-1 gene polymorphisms are primarily based on two sites: The (GT)n double nucleotide polymorphism (also called microsatellite polymorphism) and the T(−413)A single nucleotide polymorphism (SNP), both of which can regulate HO-1 gene transcription under specific conditions (45). These two polymorphic sites on the HO-1 gene promoter are shown in Fig. 1.

(GT)n double nucleotide polymorphism

Of the two polymorphism sites found in the HO-1 promoter region, the (GT)n dinucleotide repeat polymorphism (rs3074372) occurs more commonly. The number of (GT)n repeats ranges from 12–45 repeats (48), and the distribution of (GT)n repeats is bimodal, with alleles of ~23 and 30 repeat lengths (5,12,4852). According to this genetic distribution in humans and the effect of GTn repeats on the transcriptional activity of HO-1, short (S) alleles have been defined as having <25 GTn repeats, whereas those with ≥25 repeats were defined as long (L) alleles (49,53). Alternatively, other studies have used a different classification method: S alleles have been denoted by <27 GT, middle alleles (M) as 27–32 GT and L alleles as ≥33 GT (11,54). The biological function generated through the use of either classification system should be consistent, taking into account the (GT)n distribution frequency (5).

Through the use of the luciferase reporter gene system in vitro, previous studies have confirmed that S alleles are associated with a higher transcriptional activity of HO-1 compared with L alleles (51,52). Consistent with this, genetic epidemiological studies have shown that subjects with S alleles have higher levels of HO-1 mRNA compared with subjects with L alleles (12,13). Moreover, when lymphocytes isolated from humans were exposed to H2O2 stimulation in vitro, the HO-1 expression level in cells with S alleles was found to be higher compared with that of cells with L alleles. Functionally, lymphocytes with S alleles exhibit a stronger antiapoptotic ability (11). On the other hand, L (GT)n repeats have been demonstrated to be associated with a higher susceptibility to the development of numerous diseases, such as hypertension, atherosclerosis, acute kidney injury, breast cancer and pancreatitis. Clinical studies have shown that the (GT)n repeat length of the HO-1 promoter is correlated with the progression of a variety of diseases, such as hypertension, atherosclerosis, and acute kidney injury (5557).

T(−413)A SNP

T(−413)A SNP (rs2071746), another HO-1 promoter polymorphism, was detected by reverse transcription-quantitative PCR. Yamada et al (52) transfected plasmids containing different HO-1 gene promoters into bovine aortic endothelial cells to detect functional differences caused by SNPs. The results obtained showed that cells containing the A(−413)-(GT)30 allele were able to significantly enhance the biological activity of HO-1. Several studies have subsequently confirmed that the A allele is also associated with a high transcription level of HO-1, which correlates closely with the occurrence and progression of certain human diseases, including cardiovascular, and liver diseases (5860). A previous study reported that the AA genotype of T(−413)A could significantly reduce the occurrence of myocardial infarction and angina pectoris (61). In addition, the survival rate of transplanted livers was shown to be significantly higher following liver transplantation from donors who carried the A allele (62).

Population characteristics of HO-1 gene promoter polymorphisms

According to a study of HO-1 gene promoter polymorphism in Caucasian and East Asian populations, the distribution of (GT)n alleles showed a bimodal pattern with (GT)23 and (GT)30 being the two most common alleles in the two groups (50,51). However, the distribution of (GT)n alleles showed a trimodal pattern with peaks at (GT)23, (GT)30 and (GT)39 in African-American populations (50). Compared with Caucasian populations, meta-analysis showed that East Asian populations had more S alleles and fewer L alleles (58). (GT)n polymorphism includes three genotypes, namely SS, SL and LL. East Asian populations showed higher SS frequencies, ranging from 15 to 25%, whereas Caucasian populations displayed SS frequencies ranging from 6 to 17% (58). The SS genotype was associated with a lower incidence of chronic diseases, such as coronary heart disease and rheumatoid arthritis (12,58). The incidence of rheumatoid arthritis in East Asian populations is lower than that in Caucasian populations, which might be related to the higher SS genotype in East Asians. In addition, a recent study showed that Caucasian American populations have a higher SS allele frequency compared with African American populations, which might be the cause of higher neurocognitive impairment caused by human immunodeficiency virus (HIV) infection in African American populations (50). Similarly, the T(−413)A SNP polymorphism genotype also contains three genotypes, namely AA, AT and TT. These genotypes also differs between Caucasian and East Asian populations. Meta-analysis showed that East Asian populations had a lower AA genotype (~22%) compared with Caucasian populations (~35%). People with the AA genotype displayed a lower risk of coronary heart disease (58).

Mechanism of promoter polymorphisms regulating HO-1 expression

To date, the mechanism through which the T(−413)A and (GT)n gene polymorphisms regulate the expression of HO-1 has not been elucidated. A potential explanation may be that (GT)n microsatellite repeats tend to form a Z-DNA conformation, such as a left-handed double-helix structure (63). The Z-DNA conformation only occurs rarely in the genome, although its incidence does increase under certain conditions, such as the presence of a purine/pyrimidine-alternating sequence, DNA supercoiling and a high concentration of salt and certain cations (5,64). During the transcription process, the local Z-DNA conformation reduces the transcription activity of DNA. Therefore, the L (GT)n microsatellite repeats in the promoter tend to form a Z-DNA conformation, which consequently reduces the transcriptional activity of HO-1. In addition, a previous study showed that the T(−413)A and (GT)n polymorphisms may be involved in the splicing and translation of the HO-1 transcript (65). An extra exon (Exon1a) was identified for the first time in the 5′-untranslated region (5′-UTR) of HO-1. Moreover, the T(−413)A and (GT)n loci are located between Exon1a and Exon1. T(−413)A and (GT)n are located in the primary transcripts of HO-1 as alternative intronic or exonic elements. Rapid amplification of cDNA ends and luciferase reporter assays were subsequently used to confirm that T(−413)A and (GT)n are involved in 5′-UTR alternative splicing of the HO-1 primary transcript, which may result in translational control of HO-1 through regulating mRNA stability and translational efficiency (65). Moreover, expression of Exon 1a induced by hemin in hepatoma HepG2 cells is shown to decrease mature HO-1 mRNA isoform translation, which might explain the previous finding that human HO-1 mRNA and protein levels fail to correlate (66).

Association between HO-1 gene polymorphism and clinical diseases

As a cytoprotective enzyme, the intracellular expression level of HO-1 is influenced by the (GT)n dinucleotide polymorphism and T(−413)A SNP in its promoter region. Several studies have shown that these two polymorphisms affect the occurrence and progression of a variety of diseases, including liver and kidney, lung, cardiovascular, cancer and viral diseases (48,52,5558,67). The association between two HO-1 promoter polymorphisms and diseases are summarized in Table I.

Table I.

Association between two HO-1 promoter polymorphisms and a variety of diseases.

Table I.

Association between two HO-1 promoter polymorphisms and a variety of diseases.

A, (GT)n dinucleotide polymorphism

DiseasePolymorphismP-value(Refs.)
Cardiovascular disease
  Coronary heart diseaseS (GT)n repeats reduce the risk of coronary heart disease or restenosisP<0.05(58,68)
  HypertensionCarriers with the L (GT)n allele have an increased risk of hypertension, whereas the S (GT)n allele may play a protective roleP<0.05(55,70)
  AtherosclerosisSubjects with (GT)n repeats ≥32 display an increased risk of cardiovascular disease and enhanced atherosclerosis progressionP<0.01(56)
Pulmonary disease
  Chronic obstructive pulmonary diseaseL (GT)n allele is a high risk factor for chronic obstructive pulmonary diseaseP<0.01(52,78,79)
Liver disease
  Liver transplantationS (GT)n sequence (n<25) is beneficial to the functional improvement and survival of allograftsP<0.05(82)
Renal disease
  Acute kidney injuryCarrying the L (GT)n allele increases the risk of acute kidney injury after heart surgery, whereas carrying S (GT)n allele increases the risk of acute kidney injury in patients with sepsisP<0.05(57,87)
Cancer
  Pancreatic cancer, melanomaThe S (GT)n allele sequence is associated with an increased risk and lesion of pancreatic cancer and melanomaP<0.001 (pancreatic cancer) or P<0.05 (melanoma)(88,89)
  Acute lymphoblastic leukemiaPatients with S (GT)n repeats display chemotherapy resistance and chemo-induced neutropeniaP<0.05(92)
  Breast cancer, esophageal squamous cell carcinoma and laryngeal squamous cell carcinomaL (GT)n repeats are associated with a higher risk of these type of cancerP<0.05(67,9395)
  Gastric cancer and lung adenocarcinomaThe effect of HO-1 promoter polymorphism on cancer may depend on a variety of factors, including tumor type, subject ethnicity and other factorsN/A(9698)
Viral disease
  HIVS (GT)n allele inhibits neuroimmune activation, HIV-associated neuroinflammation and neurocognitive deficitsP<0.05(10,48,50)
  HCVS (GT)n allele decreases the infection titer of HCVP>0.05(101)
Other disease
  Neonatal hyperbilirubinS (GT)n repeat gene promoter results in a higher risk of neonatal hyperbilirubinemiaP<0.05(103)
  Rheumatoid arthritisS (GT)n allele reduces the risk of rheumatoid arthritis and prevents the development of joint injuryP<0.05(12,104)
  PancreatitisHO-1 containing LL allele doubles the risk of pancreatic necrosis in patients with acute pancreatitisP<0.05(105)

B, T(−413)A single nucleotide polymorphism

Disease PolymorphismP-value(Refs.)

Cardiovascular disease
  Coronary heart diseaseThe A allele reduces the risk of coronary heart diseaseP<0.05(58)
  HypertensionAA genotype increases the incidence of hypertension in a female populationP<0.01(59)
Liver disease
  Liver transplantationPatients carrying the T allele are more likely to develop esophageal varices compared with those carrying the A alleleP<0.001(60)
Other disease
  Type II diabetesPatients with the TT genotype are more prone to developing proteinuriaP<0.05(106)
  Ischemic heart diseaseThe AA genotype of HO-1 reduces the incidence of ischemic heart diseaseP<0.05(61)

[i] HO-1, heme oxygenase-1; L, long; S, short; HIV, human immunodeficiency virus; HCV, hepatitis C virus.

Cardiovascular disease

HO-1 is a protective factor for cardiovascular diseases (68). Previous studies have shown that the level of serum bilirubin, a downstream product of HO-1, is negatively correlated with the occurrence of coronary heart disease (CHD), and lower levels of serum bilirubin increase the risk of CHD. Through a correlation analysis, Zhang et al (58) demonstrated that the HO-1 (GT)n repeat length polymorphism and T(−413)A SNP were both associated with a risk of CHD following percutaneous coronary intervention. Patients with the S allele of HO-1 (GT)n had a reduced risk of restenosis compared with those with the L allele of HO-1 (GT)n. Similarly, the HO-1 A allele of T(−413)A was associated with a reduced risk of CHD. Consistent with the aforementioned results, Liang et al (69) showed that the HO-1 promoter (GT)n repeat sequence in patients with CHD with a reduced ejection fraction was longer compared with that in patients with CHD with moderate ejection fraction.

Ono et al (59) demonstrated that the HO-1 AA genotype is associated with an increased incidence of hypertension in a female population. Wenzel et al (55) reported that the individuals carrying the HO-1 L allele had an increased risk for the prevalence of arterial hypertension and higher overall mortality. In line with this, another study demonstrated that the HO-1 S allele may serve a protective role against environmental stressors in blood pressure regulation and cardiovascular mortality risk in hypertensive individuals (70). Therefore, the (GT)n repeat or SNP polymorphisms may be used as molecular markers for screening individuals who are at risk of hypertension.

Atherosclerosis is an inflammatory disease (71). As an anti-inflammatory protein, HO-1 has been confirmed to have a protective effect on blood vessels and is able to inhibit the formation of atherosclerotic lesions (7274) and thrombosis (75). Liang et al (76) also found that the HO-1 promoter (GT)n repeat sequence was longer in patients with microvascular angina compared with that in healthy controls. Similarly, Pechlaner et al (56) demonstrated that there is a strong correlation between the HO-1(GT)n repeat polymorphism and cardiovascular disease risk in subjects for whom the number of (GT)n repeats was ≥32. Taken together, these studies provided some indicators for how atherosclerosis may be accelerated and antioxidant defenses decreased in high-risk vascular groups.

In conclusion, individuals carrying the L allele of HO-1 (GT)n were shown to have an increased risk of CHD, hypertension and atherosclerosis, whereas the A allele of T(−413)A in the HO-1 gene promoter is associated with a lower risk of CHD, but an increased risk of hypertension. However, these controversial conclusions might have resulted from the limited number of subjects and different populations included in the CHD studies, thus these limitations also need be taken into consideration.

Pulmonary disease

The induction of HO-1 expression is considered to be an important protective mechanism involved in acute and chronic lung diseases, and HO-1 is an important enzymes that exerts antioxidant protective effects in the lung (77). HO-1 gene polymorphism is closely associated with chronic obstructive pulmonary disease (COPD) (52,78,79).

A previous study confirmed that insufficient or inhibited levels of HO-1 expression are an important internal factor for inducing the occurrence of COPD (80). Zhang et al (81) found that HO-1 exerts a protective function on the lungs, and the number of GT repeats was shown to influence the effects of oral N-acetylcysteine treatment. Patients with L (GT)n alleles were shown to have a much higher rate of deterioration compared with patients that lacked the L alleles. From this study, it may be concluded that HO-1 gene polymorphism has an important role in COPD. Yamada et al (52), Du et al (78) and Zhou et al (79) also demonstrated that the L (GT)n allele frequency in patients with COPD was significantly higher compared with that in the control group.

Liver disease

HO-1 has also been shown to protect allografts against ischemia/reperfusion and immune rejection. By studying 60 patients with allogeneic orthotopic liver transplantation and their accompanying donors, Zhang et al (82) found that donor-derived HO-1 gene promoter polymorphism exerted a protective effect on allograft function following orthotopic transplantation. A short (GT)n sequence (n<25 repeats) leads to increased HO-1 transcriptional activity, and a series of transplantation models have demonstrated that HO-1 upregulation is beneficial to the functional improvement and survival of the allograft (8285). In addition, studies have shown that patients with liver cirrhosis who carry the HO-1 T allele are more likely to develop esophageal varices than those with liver cirrhosis who carry the A allele. Therefore, the T allele of the HO-1 gene SNP is a risk factor for esophageal varices in patients with cirrhosis (60).

Renal disease

Acute kidney injury (AKI) refers to a multifactorial clinical syndrome caused by a variety of diseases, such as hypovolemia, diabetes, heart or lung disease (86). Leaf et al (57) demonstrated that repeated HO-1 polymorphism is associated with the development of AKI after cardiac surgery in adults, and carrying the HO-1 L (GT)n sequence (n>27 repeats) increases the risk of AKI after cardiac surgery. Hemolysis may be the primary cause of AKI, but the factors associated with cardiac surgery are complex and varied. The most plausible mechanism that has been proposed to date is that the expression of HO-1 is reduced in patients with L (GT)n repeats, resulting in a decrease in the decomposition of heme. Therefore, patients with reduced HO-1 expression caused by L (GT)n repeats may have increased susceptibility to heme-related AKI during cardiac surgery.

In a different study, Vilander et al (87) classified the HO-1 (GT)n repeat sequence in 300 patients with severe AKI sepsis and 353 patients without AKI sepsis. The alleles were classified according to the number of replicates. The short sequence S was defined as <27 repeats, the long sequence L was defined as ≥27 repeats and the very long sequence (L2) was defined as ≥34 repeats. Patients with at least one L2 sequence had lower plasma HO-1 concentrations compared with those without the L2 sequence. Patients with the S allele were found to be at an increased risk for AKI, and the adjusted odds ratio for each S allele was 1.3 in the additive genetic model. The conclusions of the aforementioned two studies are different as the susceptibility factors of the study populations were different. In the study conducted by Leaf et al, patients underwent cardiopulmonary bypass surgery, whereas all patients were critically ill with sepsis in the study conducted by Vilander et al. In addition, urination criteria were not included in the study by Leaf et al, but patients with mild AKI were excluded in the study by Vilander et al. In conclusion, it is necessary to distinguish the different phenotypes of AKI clinical syndromes when conducting pathophysiological studies on patients with AKI.

Cancer

Increasing evidence on the association between HO-1 gene promoter polymorphism and cancer has been reported. Previous studies have shown that S (GT)n repeats in the HO-1 promoter lead to higher transcriptional activity, which leads to an increased risk of pancreatic cancer and melanoma (88,89). In pancreatic cancer studies, patients with HO-1 S (GT)n repeats developed cancer more rapidly and were more likely to experience recurrence. This finding was consistent with the fact that HO-1 expression in pancreatic tumors was higher compared with that in healthy tissues, and that HO-1 overexpression has been shown to accelerate the growth, angiogenesis and metastasis of pancreatic tumors (90,91). In pediatric patients with acute lymphoblastic leukemia, patients with HO-1 S (GT)n repeats were associated with chemotherapy resistance and chemo-induced neutropenia (92). In other studies, L (GT)n repeats in the promoter of the HO-1 gene have been reported to be associated with a higher risk of breast cancer, esophageal squamous cell carcinoma and laryngeal squamous cell carcinoma (67,9395).

However, the effects of HO-1 promoter polymorphism in certain types of cancer have given rise to inconsistent results. For example, Lo et al (96) studied 183 patients with gastric cancer and 250 control patients in the Taiwan area. The results demonstrated that the L (GT)n repeat of the HO-1 gene promoter was associated with a high frequency of gastric cancer, and that the M (GT)n repeat may have a protective effect on gastric adenocarcinoma with a low frequency of lymphatic vascular invasion. However, Sawa et al (97) found that, in the female Japanese population, HO-1 S (GT)n repeats were associated with a risk of gastric cancer. This difference in the reported results may have been due to environmental factors and living habits. In addition, for lung adenocarcinoma, male smokers with the L allele had a significantly increased risk of lung adenocarcinoma compared with those with the non-L allele. In female non-smokers, L allele carriers did not differ between patients and controls (98). For sporadic colorectal cancer, studies have shown no association between HO-1 promoter (GT)n repeat polymorphism/T(−413)A SNP and the risk of disease (99). Taken together, these findings suggested that the effect of HO-1 gene promoter polymorphism on cancer may depend on a variety of factors, including tumor type and subject ethnicity (100).

Viral diseases

Studies have shown that gene polymorphisms in the HO-1 promoter region are associated with the occurrence and progression of viral infectious diseases. Gill et al (48) demonstrated that individual HO-1 (GT)n allele repeat length polymorphisms have a unique modifying effect on the risk of HIV-induced central nervous system neuroinflammation and associated neuropathy. The HO-1 S (GT)n allele increases HO-1 promoter activity and exerts a neuroprotective function through inhibiting neuroimmune activation. Similarly, previous studies have shown that the HO-1 S (GT)n allele sequence in African American populations reduces HIV-associated neuroinflammation and limits HIV-induced neurocognitive deficits (10,50). Therefore, therapeutic strategies that induce HO-1 expression should be able to reduce HIV-associated central nervous system inflammation and reduce the risk of HIV neurological disease development.

Kah et al (101) showed that inoculating mice with human hepatocytes containing the HO-1 S (GT)n allele led to a decrease in the infection titer of hepatitis C virus (HCV) compared with mice inoculated with the HO-1 L (GT)n allele. Therefore, HO-1-mediated HCV inhibition is (GT)n polymorphism-dependent. In addition, it has been suggested that HO-1 promoter polymorphism may influence coronavirus COVID-19 disease severity, such as acute lung injury and thromboembolism. Therefore, the repeat length of the HO-1 (GT)n allele in patients with severe COVID-19 should be investigated (102).

Other diseases

HO-1 gene promoter polymorphism has also been reported to be associated with neonatal hyperbilirubinemia (103), rheumatoid arthritis (12,104), pancreatitis (105), type II diabetes mellitus (106), heart disease (107) and other diseases to varying degrees. HO-1 carrying an S (GT)n repeat gene promoter was found to be associated with a higher risk of neonatal hyperbilirubinemia (103). By contrast, carrying the S (GT)n microsatellite allele was found to reduce the risk of rheumatoid arthritis and prevent the development of joint injury, whereas carrying the L (GT)n microsatellite allele led to an increase in the susceptibility to rheumatoid arthritis and promoted the development of joint injury (12,104). For pancreatitis, the results of logistic regression analysis revealed that HO-1 containing the L (GT)n allele doubled the risk of pancreatic necrosis in patients with acute pancreatitis (105). However, in patients undergoing heart transplantation, (GT)n polymorphism was not found to be associated with heart failure or patient survival. The researchers assumed that lower HO-1 activity in patients with L (GT)n alleles might be compensated by other protective genes or enzymes (107). In addition, in a retrospective analysis of patients with type II diabetes, HO-1 T(−413)A SNP was significantly associated with the prevalence of low proteinuria, and patients with the TT genotype were more prone to develop proteinuria. Furthermore, in vitro experiments revealed that the promoter activity of HO-1 containing the A allele was higher than that of T allele (106). The AA genotype of HO-1 for T(−413)A SNP polymorphism was also found to reduce the incidence of ischemic heart disease, which might be associated with the high expression level of HO-1 (61).

Conclusions and prospects

As a stress-induced cytoprotective enzyme, HO-1 expression is regulated by a complex network, including transcription factors, cytoplasmic signaling pathways, post-translational modifications and genetic polymorphisms. The research of HO-1 induced expression primarily focuses on transcription factors. To treat a variety of diseases related to inflammation and immune regulation, screening small molecule HO-1 inducers that target transcription factors is the most commonly used method (108,109). However, this attempt at translational research was not successful and encountered great challenges. In addition to toxicity and bioavailability issues, HO-1 gene polymorphism in the population may also be an important reason for the difficulty of this translational study. Therefore, if the HO-1 gene polymorphism is added as a biomarker in clinical trials, it may improve the success rate of translational research. The current challenge is that the clinical results of the correlation between HO-1 gene polymorphism and disease are still controversial, especially due to the small sample sizes and inconsistent definition standards. On the other hand, the molecular mechanism underlying HO-1 polymorphism-mediated effects on HO-1 gene expression under disease conditions has not been fully revealed, which limits our understanding of the correlation between HO-1 gene polymorphisms and different diseases. Through the method of in vitro transient transfection, the relationship between (GT)n repeats length and HO-1 expression level has been established at the cellular level; however, the research of (GT)n repeat length effects on HO-1 expression at the animal level has not yet been reported. With the emergence of CRISPR technology, it is possible to construct HO-1 gene polymorphism transgenic mice. Future research should focus on exploring the molecular mechanisms by which HO-1 gene polymorphism and transcription factors coregulate HO-1 expression and affect disease using transgenic mouse systems. These investigations will provide further support for the development of targeted HO-1 reagents in different diseases.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81902052).

Availability of data and materials

Not applicable.

Authors' contributions

LLM and LS wrote and prepared the final version of the manuscript. YXW and BHS contributed to revising the manuscript. YFL and YLJ conceptualized the idea of the review. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HO-1

heme oxygenase-1

CO

carbon monoxide

CHD

coronary heart disease

S

short

L

long

COPD

chronic obstructive pulmonary disease

AKI

acute kidney injury

References

1 

Gozzelino R, Jeney V and Soares MP: Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol Toxicol. 50:323–354. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Wagener FA, Volk HD, Willis D, Abraham NG, Soares MP, Adema GJ and Figdor CG: Different faces of the heme-heme oxygenase system in inflammation. Pharmacol Rev. 55:551–571. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Kim HP, Ryter SW and Choi AM: CO as a cellular signaling molecule. Annu Rev Pharmacol Toxicol. 46:411–449. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Calabrese V, Butterfield DA, Scapagnini G, Stella AG and Maines MD: Redox regulation of heat shock protein expression by signaling involving nitric oxide and carbon monoxide: Relevance to brain aging, neurodegenerative disorders, and longevity. Antioxid Redox Signal. 8:444–477. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Exner M, Minar E, Wagner O and Schillinger M: The role of heme oxygenase-1 promoter polymorphisms in human disease. Free Radic Biol Med. 37:1097–1104. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Linnenbaum M, Busker M, Kraehling JR and Behrends S: Heme oxygenase isoforms differ in their subcellular trafficking during hypoxia and are differentially modulated by cytochrome P450 reductase. PLoS One. 7:e354832012. View Article : Google Scholar : PubMed/NCBI

7 

Shibahara S: The heme oxygenase dilemma in cellular homeostasis: New insights for the feedback regulation of heme catabolism. Tohoku J Exp Med. 200:167–186. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Zhou J, Terluk MR, Basso L, Mishra UR, Orchard PJ, Cloyd JC, Schröder H and Kartha RV: N-acetylcysteine provides cytoprotection in murine oligodendrocytes through heme oxygenase-1 activity. Biomedicines. 8:2402020. View Article : Google Scholar : PubMed/NCBI

9 

Jazwa A and Cuadrado A: Targeting heme oxygenase-1 for neuroprotection and neuroinflammation in neurodegenerative diseases. Curr Drug Targets. 11:1517–1531. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Seu L, Burt TD, Witte JS, Martin JN, Deeks SG and McCune JM: Variations in the heme oxygenase-1 microsatellite polymorphism are associated with plasma CD14 and viral load in HIV-infected African-Americans. Genes Immun. 13:258–267. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Hirai H, Kubo H, Yamaya M, Nakayama K, Numasaki M, Kobayashi S, Suzuki S, Shibahara S and Sasaki H: Microsatellite polymorphism in heme oxygenase-1 gene promoter is associated with susceptibility to oxidant-induced apoptosis in lymphoblastoid cell lines. Blood. 102:1619–1621. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Rueda B, Oliver J, Robledo G, López-Nevot MA, Balsa A, Pascual-Salcedo D, González-Gay MA, González-Escribano MF and Martín J: HO-1 promoter polymorphism associated with rheumatoid arthritis. Arthritis Rheum. 56:3953–3958. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Brydun A, Watari Y, Yamamoto Y, Okuhara K, Teragawa H, Kono F, Chayama K, Oshima T and Ozono R: Reduced expression of heme oxygenase-1 in patients with coronary atherosclerosis. Hypertens Res. 30:341–348. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Loboda A, Damulewicz M, Pyza E, Jozkowicz A and Dulak J: Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell Mol Life Sci. 73:3221–3247. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Zhang DD: Mechanistic studies of the Nrf2-Keap1 signaling pathway. Drug Metab Rev. 38:769–789. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Jaramillo MC and Zhang DD: The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev. 27:2179–2191. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Motohashi H, Katsuoka F, Engel JD and Yamamoto M: Small Maf proteins serve as transcriptional cofactors for keratinocyte differentiation in the Keap1-Nrf2 regulatory pathway. Proc Natl Acad Sci USA. 101:6379–6384. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, et al: An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 236:313–322. 1997. View Article : Google Scholar : PubMed/NCBI

19 

Dhakshinamoorthy S, Jain AK, Bloom DA and Jaiswal AK: Bach1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H: Quinone oxidoreductase 1 gene expression and induction in response to antioxidants. J Biol Chem. 280:16891–16900. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Reichard JF, Motz GT and Puga A: Heme oxygenase-1 induction by NRF2 requires inactivation of the transcriptional repressor BACH1. Nucleic Acids Res. 35:7074–7086. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Medina MV, Sapochnik D, Garcia Solá M and Coso O: Regulation of the expression of heme oxygenase-1: Signal transduction, gene promoter activation, and beyond. Antioxid Redox Signal. 32:1033–1044. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Alam J and Cook JL: How many transcription factors does it take to turn on the heme oxygenase-1 gene? Am J Respir Cell Mol Biol. 36:166–174. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Dawn B and Bolli R: HO-1 induction by HIF-1: A new mechanism for delayed cardioprotection? Am J Physiol Heart Circ Physiol. 289:H522–H524. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Li X, Yu J, Gong L, Zhang Y, Dong S, Shi J, Li C, Li Y, Zhang Y and Li H: Heme oxygenase-1(HO-1) regulates Golgi stress and attenuates endotoxin-induced acute lung injury through hypoxia inducible factor-1α (HIF-1α)/HO-1 signaling pathway. Free Radic Biol Med. 165:243–253. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Tong Y, Kai J, Wang S, Yu Y, Xie S, Zheng H, Wang Y, Liu Y, Zhu K, Guan X, et al: VHL regulates the sensitivity of clear cell renal cell carcinoma to SIRT4-mediated metabolic stress via HIF-1α/HO-1 pathway. Cell Death Dis. 12:6212021. View Article : Google Scholar : PubMed/NCBI

26 

He C, Zhang W, Li S, Ruan W, Xu J and Xiao F: Edaravone improves septic cardiac function by inducing an HIF-1α/HO-1 pathway. Oxid Med Cell Longev. 2018:52163832018. View Article : Google Scholar : PubMed/NCBI

27 

Lee PJ, Jiang B-H, Chin BY, Iyer NV, Alam J, Semenza GL and Choi AK: Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in response to hypoxia. J Biol Chem. 272:5375–5381. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Tanos T, Marinissen MJ, Leskow FC, Hochbaum D, Martinetto H, Gutkind JS and Coso OA: Phosphorylation of c-Fos by members of the p38 MAPK family. Role in the AP-1 response to UV light. J Biol Chem. 280:18842–18852. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Monje P, Marinissen MJ and Gutkind JS: Phosphorylation of the carboxyl-terminal transactivation domain of c-Fos by extracellular signal-regulated kinase mediates the transcriptional activation of AP-1 and cellular transformation induced by platelet-derived growth factor. Mol Cell Biol. 23:7030–7043. 2003. View Article : Google Scholar : PubMed/NCBI

30 

McBride K and Nemer M: The C-terminal domain of c-fos is required for activation of an AP-1 site specific for jun-fos heterodimers. Mol Cell Biol. 18:5073–5081. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Yeh CH, Chen TP, Wang YC, Lin YM and Lin PJ: HO-1 activation can attenuate cardiomyocytic apoptosis via inhibition of NF-kappaB and AP-1 translocation following cardiac global ischemia and reperfusion. J Surg Res. 155:147–156. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Lee PJ, Camhi SL, Chin BY, Alam J and Choi AM: AP-1 and STAT mediate hyperoxia-induced gene transcription of heme oxygenase-1. Am J Physiol Lung Cell Mol Physiol. 279:L175–L182. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Paine A, Eiz-Vesper B, Blasczyk R and Immenschuh S: Signaling to heme oxygenase-1 and its anti-inflammatory therapeutic potential. Biochem Pharmacol. 80:1895–1903. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Campbell NK, Fitzgerald HK and Dunne A: Regulation of inflammation by the antioxidant haem oxygenase 1. Nat Rev Immunol. 21:411–425. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Merecz-Sadowska A, Sitarek P, Kucharska E, Kowalczyk T, Zajdel K, Cegliński T and Zajdel R: Antioxidant properties of plant-derived phenolic compounds and their effect on skin fibroblast cells. Antioxidants (Basel). 10:7262021. View Article : Google Scholar : PubMed/NCBI

36 

Yarmohammadi F, Hayes AW and Karimi G: Natural compounds against cytotoxic drug-induced cardiotoxicity: A review on the involvement of PI3K/Akt signaling pathway. J Biochem Mol Toxicol. 35:e226832021. View Article : Google Scholar : PubMed/NCBI

37 

Zhou Y, Jiang Z, Lu H, Xu Z, Tong R, Shi J and Jia G: Recent advances of natural polyphenols activators for Keap1-Nrf2 signaling pathway. Chem Biodivers. 16:e19004002019. View Article : Google Scholar : PubMed/NCBI

38 

Zhang H, Liu YY, Jiang Q, Li KR, Zhao YX, Cao C and Yao J: Salvianolic acid A protects RPE cells against oxidative stress through activation of Nrf2/HO-1 signaling. Free Radic Biol Med. 69:219–228. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Zhang HF, Wang JH, Wang YL, Gao C, Gu YT, Huang J, Wang JH and Zhang Z: Salvianolic acid A protects the kidney against oxidative stress by activating the Akt/GSK-3β/Nrf2 signaling pathway and inhibiting the NF-κB signaling pathway in 5/6 nephrectomized rats. Oxid Med Cell Longev. 2019:28535342019.PubMed/NCBI

40 

Gu X, Zheng C, Zheng Q, Chen S, Li W, Shang Z and Zhang H: Salvianolic acid A attenuates early brain injury after subarachnoid hemorrhage in rats by regulating ERK/P38/Nrf2 signaling. Am J Transl Res. 9:5643–5652. 2017.PubMed/NCBI

41 

Sun GY, Chen Z, Jasmer KJ, Chuang DY, Gu Z, Hannink M and Simonyi A: Quercetin attenuates inflammatory responses in BV-2 microglial cells: Role of MAPKs on the Nrf2 pathway and induction of heme oxygenase-1. PLoS One. 10:e01415092015. View Article : Google Scholar : PubMed/NCBI

42 

Yao P, Nussler A, Liu L, Hao L, Song F, Schirmeier A and Nussler N: Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. J Hepatol. 47:253–261. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Campbell NK, Fitzgerald HK, Fletcher JM and Dunne A: Plant-derived polyphenols modulate human dendritic cell metabolism and immune function via AMPK-dependent induction of heme oxygenase-1. Front Immunol. 10:3452019. View Article : Google Scholar : PubMed/NCBI

44 

Kosuru R, Kandula V, Rai U, Prakash S, Xia Z and Singh S: Pterostilbene decreases cardiac oxidative stress and inflammation via activation of AMPK/Nrf2/HO-1 pathway in fructose-fed diabetic rats. Cardiovasc Drugs Ther. 32:147–163. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Nakasone R, Ashina M, Abe S, Tanimura K, Van Rostenberghe H and Fujioka K: The role of heme oxygenase-1 promoter polymorphisms in perinatal disease. Int J Environ Res Public Health. 18:35202021. View Article : Google Scholar : PubMed/NCBI

46 

Öllinger R and Pratschke J: Role of heme oxygenase-1 in transplantation. Transpl Int. 23:1071–1081. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Schulz S, Chisholm KM, Zhao H, Kalish F, Yang Y, Wong RJ and Stevenson DK: Heme oxygenase-1 confers protection and alters T-cell populations in a mouse model of neonatal intestinal inflammation. Pediatr Res. 77:640–648. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Gill AJ, Garza R, Ambegaokar SS, Gelman BB and Kolson DL: Heme oxygenase-1 promoter region (GT)n polymorphism associates with increased neuroimmune activation and risk for encephalitis in HIV infection. J Neuroinflammation. 15:702018. View Article : Google Scholar : PubMed/NCBI

49 

Kaplan M, Wong RJ and Stevenson DK: Heme oxygenase-1 promoter polymorphisms: Do they modulate neonatal hyperbilirubinemia? J Perinatol. 37:901–905. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Garza R, Gill AJ, Bastien BL, Garcia-Mesa Y, Gruenewald AL, Gelman BB, Tsima B, Gross R, Letendre SL and Kolson DL: Heme oxygenase-1 promoter (GT)n polymorphism associates with HIV neurocognitive impairment. Neurol Neuroimmunol Neuroinflamm. 7:e7102020. View Article : Google Scholar : PubMed/NCBI

51 

Chen YH, Lin SJ, Lin MW, Tsai HL, Kuo SS, Chen JW, Charng MJ, Wu TC, Chen LC, Ding YA, et al: Microsatellite polymorphism in promoter of heme oxygenase-1 gene is associated with susceptibility to coronary artery disease in type 2 diabetic patients. Hum Genet. 111:1–8. 2002. View Article : Google Scholar : PubMed/NCBI

52 

Yamada N, Yamaya M, Okinaga S, Nakayama K, Sekizawa K, Shibahara S and Sasaki H: Microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with susceptibility to emphysema. Am J Hum Genet. 66:187–195. 2000. View Article : Google Scholar : PubMed/NCBI

53 

Espinoza JA, González PA and Kalergis AM: Modulation of antiviral immunity by heme oxygenase-1. Am J Pathol. 187:487–493. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Zhou Y, Wang SN, Li H, Zha W, Peng Q, Li S, Chen Y and Jin L: Quantitative trait analysis of polymorphisms in two bilirubin metabolism enzymes to physiologic bilirubin levels in Chinese newborns. J Pediatr. 165:1154–1160.e1. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Wenzel P, Rossmann H, Müller C, Kossmann S, Oelze M, Schulz A, Arnold N, Simsek C, Lagrange J, Klemz R, et al: Heme oxygenase-1 suppresses a pro-inflammatory phenotype in monocytes and determines endothelial function and arterial hypertension in mice and humans. Eur Heart J. 36:3437–3446. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Pechlaner R, Willeit P, Summerer M, Santer P, Egger G, Kronenberg F, Demetz E, Weiss G, Tsimikas S, Witztum JL, et al: Heme oxygenase-1 gene promoter microsatellite polymorphism is associated with progressive atherosclerosis and incident cardiovascular disease. Arterioscler Thromb Vasc Biol. 35:229–236. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Leaf DE, Body SC, Muehlschlegel JD, McMahon GM, Lichtner P, Collard CD, Shernan SK, Fox AA and Waikar SS: Length polymorphisms in heme oxygenase-1 and AKI after cardiac surgery. J Am Soc Nephrol. 27:3291–3297. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Zhang MM, Zheng YY, Gao Y, Zhang JZ, Liu F, Yang YN, Li XM, Ma YT and Xie X: Heme oxygenase-1 gene promoter polymorphisms are associated with coronary heart disease and restenosis after percutaneous coronary intervention: A meta-analysis. Oncotarget. 7:83437–83450. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Ono K, Mannami T and Iwai N: Association of a promoter variant of the haeme oxygenase-1 gene with hypertension in women. J Hypertens. 21:1497–1503. 2003. View Article : Google Scholar : PubMed/NCBI

60 

Ellakany WI, Mahmoud MoheyEldin K, Invernizzi P, Mahmoud ElKady A, Eldin Fathy Abou Elkheir H, Abdel Haleem Abo Elwafa R and Ellakany A: Study of the influence of heme oxygenase 1 gene single nucleotide polymorphism (rs2071746) on esophageal varices among patients with cirrhosis. Eur J Gastroenterol Hepatol. 30:888–892. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Ono K, Goto Y, Takagi S, Baba S, Tago N, Nonogi H and Iwai N: A promoter variant of the heme oxygenase-1 gene may reduce the incidence of ischemic heart disease in Japanese. Atherosclerosis. 173:313–319. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Buis CI, van der Steege G, Visser DS, Nolte IM, Hepkema BG, Nijsten M, Slooff MJ and Porte RJ: Heme oxygenase-1 genotype of the donor is associated with graft survival after liver transplantation. Am J Transplant. 8:377–385. 2008. View Article : Google Scholar : PubMed/NCBI

63 

Rich A, Nordheim A and Wang AH: The chemistry and biology of left-handed Z-DNA. Annu Rev Biochem. 53:791–846. 1984. View Article : Google Scholar : PubMed/NCBI

64 

Naylor LH and Clark EM: d(TG)n.d(CA)n sequences upstream of the rat prolactin gene form Z-DNA and inhibit gene transcription. Nucleic Acids Res. 18:1595–1601. 1990. View Article : Google Scholar : PubMed/NCBI

65 

Kramer M, Sponholz C, Slaba M, Wissuwa B, Claus RA, Menzel U, Huse K, Platzer M and Bauer M: Alternative 5′ untranslated regions are involved in expression regulation of human heme oxygenase-1. PLoS One. 8:e772242013. View Article : Google Scholar : PubMed/NCBI

66 

Doberer D, Haschemi A, Andreas M, Zapf TC, Clive B, Jeitler M, Heinzl H, Wagner O, Wolzt M and Bilban M: Haem arginate infusion stimulates haem oxygenase-1 expression in healthy subjects. Br J Pharmacol. 161:1751–1762. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Hong CC, Ambrosone CB, Ahn J, Choi JY, McCullough ML, Stevens VL, Rodriguez C, Thun MJ and Calle EE: Genetic variability in iron-related oxidative stress pathways (Nrf2, NQ01, NOS3, and HO-1), iron intake, and risk of postmenopausal breast cancer. Cancer Epidemiol Biomarkers Prev. 16:1784–1794. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Abraham NG and Kappas A: Pharmacological and clinical aspects of heme oxygenase. Pharmacol Rev. 60:79–127. 2008. View Article : Google Scholar : PubMed/NCBI

69 

Liang KW, Lee WJ, Lee WL, Wu JP, Lee IT, Wang JS and Sheu WH: Subjects with coronary artery disease and reduced ejection fraction have longer (GT)n repeats in the heme-oxygenase 1 gene promoter. Heart Vessels. 36:615–620. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Wu MM, Chiou HY, Chen CL, Hsu LI, Lien LM, Wang CH, Hsieh YC, Wang YH, Hsueh YM, Lee TC, et al: Association of heme oxygenase-1 GT-repeat polymorphism with blood pressure phenotypes and its relevance to future cardiovascular mortality risk: An observation based on arsenic-exposed individuals. Atherosclerosis. 219:704–708. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Ross R: Atherosclerosis-an inflammatory disease. N Engl J Med. 340:115–126. 1999. View Article : Google Scholar : PubMed/NCBI

72 

Duckers HJ, Boehm M, True AL, Yet SF, San H, Park JL, Clinton Webb R, Lee ME, Nabel GJ and Nabel EG: Heme oxygenase-1 protects against vascular constriction and proliferation. Nat Med. 7:693–698. 2001. View Article : Google Scholar : PubMed/NCBI

73 

Ishikawa K, Sugawara D, Wang XP, Suzuki K, Itabe H, Maruyama Y and Lusis AJ: Heme oxygenase-1 inhibits atherosclerotic lesion formation in ldl-receptor knockout mice. Circ Res. 88:506–512. 2001. View Article : Google Scholar : PubMed/NCBI

74 

Tulis DA, Durante W, Peyton KJ, Evans AJ and Schafer AI: Heme oxygenase-1 attenuates vascular remodeling following balloon injury in rat carotid arteries. Atherosclerosis. 155:113–122. 2001. View Article : Google Scholar : PubMed/NCBI

75 

Lindenblatt N, Bordel R, Schareck W, Menger MD and Vollmar B: Vascular heme oxygenase-1 induction suppresses microvascular thrombus formation in vivo. Arterioscler Thromb Vasc Biol. 24:601–606. 2004. View Article : Google Scholar : PubMed/NCBI

76 

Liang KW, Lee WJ, Lee IT, Lee WL, Wang JS, Wu JP and Sheu WHH: Subjects with microvascular angina have longer GT repeats polymorphism in the haem oxygenase-1 gene promoter. Biomarkers. 25:144–148. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Raval CM and Lee PJ: Heme oxygenase-1 in lung disease. Curr Drug Targets. 11:1532–1540. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Du Y, Zhang H, Xu Y, Ding Y, Chen X, Mei Z, Ding H and Jie Z: Association among genetic polymorphisms of GSTP1, HO-1, and SOD-3 and chronic obstructive pulmonary disease susceptibility. Int J Chron Obstruct Pulmon Dis. 14:2081–2088. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Zhou H, Ying X, Liu Y, Ye S, Yan J and Li Y: Genetic polymorphism of heme oxygenase 1 promoter in the occurrence and severity of chronic obstructive pulmonary disease: A meta-analysis. J Cell Mol Med. 21:894–903. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Fischer BM, Pavlisko E and Voynow JA: Pathogenic triad in COPD: Oxidative stress, protease-antiprotease imbalance, and inflammation. Int J Chron Obstruct Pulmon Dis. 6:413–421. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Zhang JQ, Zhang JQ, Fang LZ, Liu L, Fu WP and Dai LM: Effect of oral N-acetylcysteine on COPD patients with microsatellite polymorphism in the heme oxygenase-1 gene promoter. Drug Des Devel Ther. 9:6379–6387. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Zhang ZY, Guan J, Li H, Zhou ZQ and Zhou GW: Heme oxygenase-1 promoter polymorphism protects liver allograft. Indian J Surg. 78:14–19. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Soares M, Lin Y, Anrather J, Csizmadia E, Takigami K, Sato K, Grey ST, Colvin RB, Choi AM, Poss KD and Bach FH: Expression of heme oxygenase-1 can determine cardiac xenograft survival. Nat Med. 4:1073–1077. 1998. View Article : Google Scholar : PubMed/NCBI

84 

Katori M, Buelow R, Ke B, Ma J, Coito AJ, Iyer S, Southard D, Busuttil RW and Kupiec-Weglinski JW: Heme oxygenase-1 overexpression protects rat hearts from cold ischemia/reperfusion injury via an antiapoptotic pathway. Transplantation. 73:287–292. 2002. View Article : Google Scholar : PubMed/NCBI

85 

Ozaki KS, Marques GM, Nogueira E, Feitoza RQ, Cenedeze MA, Franco MF, Mazzali M, Soares MP, Pacheco-Silva A and Câmara NO: Improved renal function after kidney transplantation is associated with heme oxygenase-1 polymorphism. Clin Transplant. 22:609–616. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Nisula S, Kaukonen KM, Vaara ST, Korhonen AM, Poukkanen M, Karlsson S, Haapio M, Inkinen O, Parviainen I, Suojaranta-Ylinen R, et al: Incidence, risk factors and 90-day mortality of patients with acute kidney injury in Finnish intensive care units: The FINNAKI study. Intensive Care Med. 39:420–428. 2013. View Article : Google Scholar : PubMed/NCBI

87 

Vilander LM, Vaara ST, Donner KM, Lakkisto P, Kaunisto MA and Pettilä V; FINNAKI Study Group, : Heme oxygenase-1 repeat polymorphism in septic acute kidney injury. PLoS One. 14:e02172912019. View Article : Google Scholar : PubMed/NCBI

88 

Vashist YK, Uzungolu G, Kutup A, Gebauer F, Koenig A, Deutsch L, Zehler O, Busch P, Kalinin V, Izbicki JR and Yekebas EF: Heme oxygenase-1 germ line GTn promoter polymorphism is an independent prognosticator of tumor recurrence and survival in pancreatic cancer. J Surg Oncol. 104:305–311. 2011. View Article : Google Scholar : PubMed/NCBI

89 

Okamoto I, Krögler J, Endler G, Kaufmann S, Mustafa S, Exner M, Mannhalter C, Wagner O and Pehamberger H: A microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with risk for melanoma. Int J Cancer. 119:1312–1315. 2006. View Article : Google Scholar : PubMed/NCBI

90 

Berberat PO, Dambrauskas Z, Gulbinas A, Giese T, Giese N, Künzli B, Autschbach F, Meuer S, Büchler MW and Friess H: Inhibition of heme oxygenase-1 increases responsiveness of pancreatic cancer cells to anticancer treatment. Clin Cancer Res. 11:3790–3798. 2005. View Article : Google Scholar : PubMed/NCBI

91 

Sunamura M, Duda DG, Ghattas MH, Lozonschi L, Motoi F, Yamauchi J, Matsuno S, Shibahara S and Abraham NG: Heme oxygenase-1 accelerates tumor angiogenesis of human pancreatic cancer. Angiogenesis. 6:15–24. 2003. View Article : Google Scholar : PubMed/NCBI

92 

Bukowska-Strakova K, Włodek J, Pitera E, Kozakowska M, Konturek-Cieśla A, Cieśla M, Gońka M, Nowak W, Wieczorek A, Pawińska-Wąsikowska K, et al: Role of HMOX1 promoter genetic variants in chemoresistance and chemotherapy induced neutropenia in children with acute lymphoblastic leukemia. Int J Mol Sci. 22:9882021. View Article : Google Scholar : PubMed/NCBI

93 

Tang D, Tang WJ, Shi XL, Li WP, Zhou H, Lu LM and Tao L: Association of the microsatellite (GT)n repeat polymorphisms of the HO-1 gene promoter and corresponding serum levels with the risk of laryngeal squamous cell carcinoma. Acta Otolaryngol. 136:806–811. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Zhang L, Song FF, Huang YB, Zheng H, Song FJ and Chen KX: Association between the (GT)n polymorphism of the HO-1 gene promoter region and cancer risk: A meta-analysis. Asian Pac J Cancer Prev. 15:4617–4622. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Hu JL, Li ZY, Liu W, Zhang RG, Li GL, Wang T, Ren JH and Wu G: Polymorphism in heme oxygenase-1 (HO-1) promoter and alcohol are related to the risk of esophageal squamous cell carcinoma on Chinese males. Neoplasma. 57:86–92. 2010. View Article : Google Scholar : PubMed/NCBI

96 

Lo SS, Lin SC, Wu CW, Chen JH, Yeh WI, Chung MY and Lui WY: Heme oxygenase-1 gene promoter polymorphism is associated with risk of gastric adenocarcinoma and lymphovascular tumor invasion. Ann Surg Oncol. 14:2250–2256. 2007. View Article : Google Scholar : PubMed/NCBI

97 

Sawa T, Mounawar M, Tatemichi M, Gilibert I, Katoh T and Ohshima H: Increased risk of gastric cancer in Japanese subjects is associated with microsatellite polymorphisms in the heme oxygenase-1 and the inducible nitric oxide synthase gene promoters. Cancer Lett. 269:78–84. 2008. View Article : Google Scholar : PubMed/NCBI

98 

Kikuchi A, Yamaya M, Suzuki S, Yasuda H, Kubo H, Nakayama K, Handa M, Sasaki T, Shibahara S, Sekizawa K and Sasaki H: Association of susceptibility to the development of lung adenocarcinoma with the heme oxygenase-1 gene promoter polymorphism. Hum Genet. 116:354–360. 2005. View Article : Google Scholar : PubMed/NCBI

99 

Jirásková A, Novotný J, Novotný L, Vodicka P, Pardini B, Naccarati A, Schwertner HA, Hubácek JA, Puncochárová L, Šmerhovský Z and Vítek L: Association of serum bilirubin and promoter variations in HMOX1 and UGT1A1 genes with sporadic colorectal cancer. Int J Cancer. 131:1549–1555. 2012. View Article : Google Scholar : PubMed/NCBI

100 

Podkalicka P, Mucha O, Józkowicz A, Dulak J and Łoboda A: Heme oxygenase inhibition in cancers: Possible tools and targets. Contemp Oncol (Pozn). 22:32–23. 2018.PubMed/NCBI

101 

Kah J, Volz T, Lütgehetmann M, Groth A, Lohse AW, Tiegs G, Sass G and Dandri M: Haem oxygenase-1 polymorphisms can affect HCV replication and treatment responses with different efficacy in humanized mice. Liver Int. 37:1128–1137. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Singh D, Wasan H and Reeta KH: Heme oxygenase-1 modulation: A potential therapeutic target for COVID-19 and associated complications. Free Radic Biol Med. 161:263–271. 2020. View Article : Google Scholar : PubMed/NCBI

103 

Zhou JF, Luo JY, Zhu WB, Yang CY, Zeng YL and Qiu XL: Association between genetic polymorphism of heme oxygenase 1 promoter and neonatal hyperbilirubinemia: A meta-analysis. J Matern Fetal Neonatal Med. 34:12–23. 2021. View Article : Google Scholar : PubMed/NCBI

104 

Wagener FA, Toonen EJ, Wigman L, Fransen J, Creemers MC, Radstake TR, Coenen MJ, Barrera P, van Riel PL and Russel FG: HMOX1 promoter polymorphism modulates the relationship between disease activity and joint damage in rheumatoid arthritis. Arthritis Rheum. 58:3388–3393. 2008. View Article : Google Scholar : PubMed/NCBI

105 

Gulla A, Gulbinas A, Dambrauskas Z and Strupas K: Heme oxygenase-1 polymorphism is associated with the development of necrotic acute pancreatitis via vascular cell adhesion molecule-1 and the E-selectin expression regulation pathway. Pancreas. 48:787–791. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Lee EY, Lee YH, Kim SH, Chung KS, Kwon O, Kim BS, Nam CM, Park CS, Lee BW, Kang ES, et al: Association between heme oxygenase-1 promoter polymorphisms and the development of albuminuria in type 2 diabetes: A case-control study. Medicine (Baltimore). 94:e18252015. View Article : Google Scholar : PubMed/NCBI

107 

Holweg CT, Balk AH, Uitterlinden AG, Niesters HG, Maat LP, Weimar W and Baan CC: Functional heme oxygenase-1 promoter polymorphism in relation to heart failure and cardiac transplantation. J Heart Lung Transplant. 24:493–497. 2005. View Article : Google Scholar : PubMed/NCBI

108 

Foresti R, Bains SK, Pitchumony TS, de Castro Brás LE, Drago F, Dubois-Randé JL, Bucolo C and Motterlini R: Small molecule activators of the Nrf2-HO-1 antioxidant axis modulate heme metabolism and inflammation in BV2 microglia cells. Pharmacol Res. 76:132–148. 2013. View Article : Google Scholar : PubMed/NCBI

109 

Funes SC, Rios M, Fernández-Fierro A, Covián C, Bueno SM, Riedel CA, Mackern-Oberti JP and Kalergis AM: Naturally derived heme-oxygenase 1 inducers and their therapeutic application to immune-mediated diseases. Front Immunol. 11:14672020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2022
Volume 25 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma L, Sun L, Wang Y, Sun B, Li Y and Jin Y: Association between HO‑1 gene promoter polymorphisms and diseases (Review). Mol Med Rep 25: 29, 2022
APA
Ma, L., Sun, L., Wang, Y., Sun, B., Li, Y., & Jin, Y. (2022). Association between HO‑1 gene promoter polymorphisms and diseases (Review). Molecular Medicine Reports, 25, 29. https://doi.org/10.3892/mmr.2021.12545
MLA
Ma, L., Sun, L., Wang, Y., Sun, B., Li, Y., Jin, Y."Association between HO‑1 gene promoter polymorphisms and diseases (Review)". Molecular Medicine Reports 25.1 (2022): 29.
Chicago
Ma, L., Sun, L., Wang, Y., Sun, B., Li, Y., Jin, Y."Association between HO‑1 gene promoter polymorphisms and diseases (Review)". Molecular Medicine Reports 25, no. 1 (2022): 29. https://doi.org/10.3892/mmr.2021.12545