1
|
Fares K, El-Deeb MK, Elsammak O, Ouf A, Saeed HMS, Baess A, Elsammak M, El-Attar E. SNP (A > G - rs13057211) but not GT(n) polymorphism in HMOX-1 promotor gene is associated with COVID-19 mortality. BMC Pulm Med 2023; 23:514. [PMID: 38129860 PMCID: PMC10734135 DOI: 10.1186/s12890-023-02785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
INTRODUCTION COVID-19 causes severe inflammatory respiratory distress syndrome. The global pandemic caused millions of cases of morbidity and mortality worldwide. Patients may present with variable symptoms including dyspnea, fever, and GIT manifestations. The HMOX-1 gene is located on the long (q) arm of chromosome 22 at position 12.3. HMOX-1 is expressed in all mammalian tissues at basal levels and is considered as a stress response enzyme. HMOX-1 has a specific polymorphic site with variable GT(n) repeats at the promotor region. Several authors evaluated the HMOX-1 GT(n) promoter polymorphism in different inflammatory conditions. We evaluated HMOX-1 promoter polymorphism in relation to serum Hemoxygenase level and inflammatory makers (CRP, Ferritin, PCT, IL-6 and D-dimer) in patients affected by SARS-COV-2 disease. SUBJECTS AND METHODS Ninety patients confirmed to be infected with COVID-19 were followed up till the study end point (recovery and discharge or death). HMOX-1 promotor GT(n) polymorphism was evaluated using Sanger sequencing. HMOX-1 enzyme serum level was measured by ELISA and the level of different inflammatory markers was assessed by available commercial kits. RESULTS A novel Single nucleotide polymorphism (SNP) (A > G) - rs13057211 in the GT(n) region of HMOX-1 promoter gene was found in 40 (61.5%) COVID-19 patients out of the studied 65 patients. This (A > G) SNP was associated with higher mortality rate in COVID-19 as it was detected in 27 patients (75% of the patients who succumbed to the disease) (p = 0.021, Odds ratio = 3.7; 95% CI:1.29-10.56). Serum IL-6 (Interleuken-6) was positively correlated the length of Hospital Stay (LOHS) and procalcitonin (PCT); (p = 0.014, r: 0.651 and p < 0.001, r:0.997) respectively while negatively correlated with levels of HMOX-1 enzyme serum level (p = 0.013, r: -0.61). CRP correlated positively with LOHS (p = 0.021, r = 0.4), PCT (p = 0.044, r = 0.425) and age (p < 0.001, r = 0.685). Higher levels of D-Dimer and PCT were observed in patients with the long repeat. There was no significant difference between patients who recovered and those who died from COVID-19 as regards HMOX-1 level and GT(n) polymorphism. CONCLUSION We report a novel SNP (A > G, rs13057211) in the GT(n) region of HMOX-1 promoter gene that was associated with mortality in COVID-19 patients, however no significant difference was found in HMOX-1 serum level or HMOX-1 (GT)n repeats within the studied groups.
Collapse
Affiliation(s)
- Kerolos Fares
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Mona K El-Deeb
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al Jouf University, Sakakah, Saudi Arabia
| | - Omar Elsammak
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amged Ouf
- Department of Biology and Biotechnology Graduate Program, School of Sciences and Engineering (SSE), The American University in Cairo (AUC), New Cairo, Egypt
| | - Hesham Mahmoud Sayd Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research Alexandria University, Alexandria, Egypt
| | - Ayman Baess
- Department of Chest Diseases, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed Elsammak
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| | - Eman El-Attar
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
2
|
Generation of human induced pluripotent stem cell lines with HMOX1 promoter polymorphism and CRISPR/Cas9-mediated deletion of exon 50 of DMD gene. Stem Cell Res 2023; 66:103004. [PMID: 36565681 DOI: 10.1016/j.scr.2022.103004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/03/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), originating from the lack of functional dystrophin, clinically manifests as devastating disease of skeletal muscles with progressive cardiac involvement. HMOX1 promoter polymorphism may reflect different activity of heme oxygenase-1 (HO-1) that may be critical for DMD progression. Here we generated human induced pluripotent stem cell (hiPSC) lines from healthy donors-derived peripheral blood mononuclear cells with different variants of HMOX1 promoter (GT repeats), and engineered by CRISPR/Cas9-mediated deletion of exon 50 of DMD gene. Such in vitro model could add to molecular understanding of DMD and verify the prognostic value of HMOX1 promoter polymorphism.
Collapse
|
3
|
Wu YH, Hsieh HL. Roles of Heme Oxygenase-1 in Neuroinflammation and Brain Disorders. Antioxidants (Basel) 2022; 11:antiox11050923. [PMID: 35624787 PMCID: PMC9137505 DOI: 10.3390/antiox11050923] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
The heme oxygenase (HO) system is believed to be a crucial mechanism for the nervous system under stress conditions. HO degrades heme to carbon monoxide, iron, and biliverdin. These heme degradation products are involved in modulating cellular redox homeostasis. The first identified isoform of the HO system, HO-1, is an inducible protein that is highly expressed in peripheral organs and barely detectable in the brain under normal conditions, whereas HO-2 is a constitutive protein that is highly expressed in the brain. Several lines of evidence indicate that HO-1 dysregulation is associated with brain inflammation and neurodegeneration, including Parkinson’s and Alzheimer’s diseases. In this review, we summarize the essential roles that the HO system plays in ensuring brain health and the molecular mechanism through which HO-1 dysfunction leads to neurodegenerative diseases and disruption of nervous system homeostasis. We also provide a summary of the herbal medicines involved in the regulation of HO-1 expression and explore the current situation regarding herbal remedies and brain disorders.
Collapse
Affiliation(s)
- Yi-Hsuan Wu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan;
| | - Hsi-Lung Hsieh
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan;
- Department of Nursing, Division of Basic Medical Sciences, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-3-211-8999 (ext. 5421)
| |
Collapse
|
4
|
Szade A, Szade K, Mahdi M, Józkowicz A. The role of heme oxygenase-1 in hematopoietic system and its microenvironment. Cell Mol Life Sci 2021; 78:4639-4651. [PMID: 33787980 PMCID: PMC8195762 DOI: 10.1007/s00018-021-03803-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/09/2021] [Accepted: 02/24/2021] [Indexed: 12/22/2022]
Abstract
Hematopoietic system transports all necessary nutrients to the whole organism and provides the immunological protection. Blood cells have high turnover, therefore, this system must be dynamically controlled and must have broad regeneration potential. In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. First, we discuss how HO-1 influences hematopoietic stem cells (HSC) self-renewal, aging and differentiation. We also describe a critical role of HO-1 in endothelial cells and mesenchymal stromal cells that constitute the specialized bone marrow niche of HSC. We further discuss the molecular and cellular mechanisms by which HO-1 modulates innate and adaptive immune responses. Finally, we highlight how modulation of HO-1 activity regulates the mobilization of bone marrow hematopoietic cells to peripheral blood. We critically discuss the issue of metalloporphyrins, commonly used pharmacological modulators of HO-1 activity, and raise the issue of their important HO-1-independent activities.
Collapse
Affiliation(s)
- Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Mahdi Mahdi
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| |
Collapse
|
5
|
Choi HI, Kim K, Lee J, Chang Y, Rhee HY, Park S, Lee WI, Choe W, Ryu CW, Jahng GH. Relationship between Brain Tissue Changes and Blood Biomarkers of Cyclophilin A, Heme Oxygenase-1, and Inositol-Requiring Enzyme 1 in Patients with Alzheimer's Disease. Diagnostics (Basel) 2021; 11:740. [PMID: 33919311 PMCID: PMC8143350 DOI: 10.3390/diagnostics11050740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/30/2022] Open
Abstract
Cyclophilin A (CypA), heme oxygenase-1 (HO-1), and inositol-requiring enzyme 1 (IRE1) are believed to be associated with Alzheimer's disease (AD). In this study, we investigated the association between gray matter volume (GMV) changes and blood levels of CypA, HO-1, and IRE1 in cognitively normal (CN) subjects and those with amnestic mild cognitive impairment (aMCI) and AD. Forty-five elderly CN, 34 aMCI, and 39 AD subjects were enrolled in this study. The results of voxel-based multiple regression analysis showed that blood levels of CypA, HO-1, and IRE1 were correlated with GMV on brain magnetic resonance imaging (MRI) in the entire population (p = 0.0005). The three serum protein levels were correlated with GMV of signature AD regions in the population as a whole. CypA values increased with increasing GMV in the occipital gyrus (r = 0.387, p < 0.0001) and posterior cingulate (r = 0.196, p = 0.034). HO-1 values increased with increasing GMV at the uncus (r = 0.307, p = 0.0008), lateral globus pallidus and putamen (r = 0.287, p = 0.002), and hippocampus (r = 0.197, p = 0.034). IRE1 values decreased with increasing GMV at the uncus (r = -0.239, p = 0.010) and lateral globus pallidus and putamen (r = -0.335, p = 0.0002). Associations between the three serum protein levels and regional GMV indicate that the blood levels of these biomarkers may reflect the pathological mechanism of AD in the brain.
Collapse
Affiliation(s)
- Hyon-Il Choi
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Korea; (H.-I.C.); (S.P.)
| | - Kiyoon Kim
- Department of Biochemistry and Molecular Biology, Graduate School, Kyung Hee University, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Jiyoon Lee
- Department of Biomedical Engineering, Undergraduate School, College of Electronics and Information, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Seoul 17104, Korea; (J.L.); (Y.C.)
| | - Yunjung Chang
- Department of Biomedical Engineering, Undergraduate School, College of Electronics and Information, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Seoul 17104, Korea; (J.L.); (Y.C.)
| | - Hak Young Rhee
- Department of Medicine, College of Medicine, Kyung Hee University, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (H.Y.R.); (W.-I.L.)
- Department of Neurology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-gu, Seoul 05278, Korea
| | - Soonchan Park
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Korea; (H.-I.C.); (S.P.)
- Department of Medicine, College of Medicine, Kyung Hee University, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (H.Y.R.); (W.-I.L.)
| | - Woo-In Lee
- Department of Medicine, College of Medicine, Kyung Hee University, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (H.Y.R.); (W.-I.L.)
- Department of Laboratory Medicine, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-gu, Seoul 05278, Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, College of Medicine, Kyung Hee University, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Chang-Woo Ryu
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Korea; (H.-I.C.); (S.P.)
- Department of Medicine, College of Medicine, Kyung Hee University, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (H.Y.R.); (W.-I.L.)
| | - Geon-Ho Jahng
- Department of Radiology, Kyung Hee University Hospital at Gangdong, 892 Dongnam-ro, Gangdong-Gu, Seoul 05278, Korea; (H.-I.C.); (S.P.)
- Department of Medicine, College of Medicine, Kyung Hee University, 26 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (H.Y.R.); (W.-I.L.)
| |
Collapse
|
6
|
GT-Repeat Polymorphism in the HO-1 Gene Promoter Is Associated with Risk of Liver Cancer: A Follow-Up Study from Arseniasis-Endemic Areas in Taiwan. J Clin Med 2021; 10:jcm10071489. [PMID: 33916685 PMCID: PMC8038349 DOI: 10.3390/jcm10071489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
The induction of heme oxygenase-1 (HO-1) has been shown to have therapeutic potential in experimental models of hepatitis and liver fibrosis, which are closely related to liver cancer. In humans, HO-1 induction is transcriptionally modulated by the length of a GT-repeat [(GT)n] in the promoter region. We aimed to investigate the effect of HO-1 (GT)n variants on liver cancer in a human population. We determined the HO-1 genotype in 1153 study subjects and examined their association with liver cancer risk during a 15.9-year follow-up. Allelic polymorphisms were classified as short [S, <27 (GT)n] or long [L, ≥27 (GT)n]. Newly developed cancer cases were identified through linkage to the National Cancer Registry of Taiwan. Multivariate Cox regression analysis was used to evaluate the effect of the HO-1 (GT)n variants. Alpha-fetoprotein (AFP) and cirrhosis history were also examined. The S/S genotype was found to be significantly associated with liver cancer risk, compared to the L/S and L/L genotypes. The S/S genotype group also had a higher percentage of subjects with abnormal AFP levels than other groups. There were significant percentages of cirrhosis among groups who carried S-alleles. Our findings indicate that short (GT)n variants in the HO-1 gene may confer susceptibility to rather than protection from liver cirrhosis/cancer.
Collapse
|
7
|
Endometriosis Is Associated with Functional Polymorphism in the Promoter of Heme Oxygenase 1 ( HMOX1) Gene. Cells 2021; 10:cells10030695. [PMID: 33800989 PMCID: PMC8003868 DOI: 10.3390/cells10030695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/15/2023] Open
Abstract
Endometriosis is a common gynecological disorder characterized by the ectopic growth of endometrial-like tissue outside the uterine cavity. Etiopathogenesis of endometriosis is poorly understood; it is plausible, however, that the disease may be associated with oxidative stress related to local heme and iron metabolism. Therefore, the aim of the study was to reveal a possible association of endometriosis with a stress-inducible heme oxygenase 1 (HMOX1). For this purpose, 228 patients with clinically confirmed endometriosis and 415 control parous women from general Polish population were examined for functional -413A>T (rs2071746) single-nucleotide polymorphism (SNP) and (GT)n dinucleotide repeat length polymorphism in the promoter of HMOX1 gene. In addition, -413A>T SNP was assessed by the specific TaqMan® SNP Genotyping Assay, and (GT)n polymorphism was determined by PCR product size analysis. We found that endometriosis is associated with an increased frequency of -413A(GT)31,32 haplotype (OR (95%CI) = 1.27 (1.01-1.60), p = 0.0381) and -413A(GT)31,32 homozygous genotype [OR (95%CI) = 1.51 (1.06-2.17), p = 0.0238]. These data suggest that endometriosis is associated with functional polymorphism of HMOX1 gene, and this gene may play a part in the pathogenesis of this disorder.
Collapse
|
8
|
Yamashita K, Kawasaki A, Matsushita T, Furukawa H, Kondo Y, Okiyama N, Nagaoka S, Shimada K, Sugii S, Katayama M, Hirohata S, Okamoto A, Chiba N, Suematsu E, Setoguchi K, Migita K, Sumida T, Tohma S, Hamaguchi Y, Hasegawa M, Sato S, Kawaguchi Y, Takehara K, Tsuchiya N. Association of functional (GA)n microsatellite polymorphism in the FLI1 gene with susceptibility to human systemic sclerosis. Rheumatology (Oxford) 2021; 59:3553-3562. [PMID: 32696043 DOI: 10.1093/rheumatology/keaa306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 04/08/2020] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Susceptibility genes that can account for characteristic features of SSc such as fibrosis, vasculopathy and autoimmunity remain to be determined. In mice, deficiency of Friend leukaemia integration 1 transcription factor (Fli1) causes SSc-like disease with these features. The human FLI1 gene contains (GA)n microsatellite, which has been shown to be associated with expression level. Because microsatellite polymorphisms are difficult to capture by genome-wide association studies, we directly genotyped FLI1 (GA)n microsatellite and examined its association with SSc. METHODS Genomic DNA from 639 Japanese SSc patients and 851 healthy controls was genotyped for (GA)n microsatellite using the fragment assay. The cut-off repeat number for susceptibility to SSc was determined by receiver operating characteristics (ROC) analysis. Association with susceptibility and clinical characteristics was examined using logistic regression analysis. FLI1 mRNA levels were determined using quantitative RT-PCR. RESULTS Based on the ROC analysis, (GA)n alleles with ≥22 repeats were collectively defined as L alleles and alleles with ≤21 repeats as S alleles. (GA)n L alleles were significantly associated with susceptibility to SSc (P = 5.0e-04, odds ratio 1.34, additive model). Significant association was observed both in diffuse cutaneous and limited cutaneous SSc. Among the SSc, (GA)n L alleles were significantly enriched in the patients with a modified Rodnan total skin thickness score ≥10 compared with those with a score <10. FLI1 mRNA levels were significantly decreased in healthy controls carrying (GA)n L alleles as compared with non-carriers. CONCLUSION Extended repeat alleles of FLI1 (GA)n microsatellite may be associated with lower FLI1 mRNA levels and susceptibility to human SSc.
Collapse
Affiliation(s)
- Keita Yamashita
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Aya Kawasaki
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | | | - Hiroshi Furukawa
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan.,Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
| | - Yuya Kondo
- Department of Internal Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoko Okiyama
- Department of Dermatology, University of Tsukuba, Tsukuba, Japan
| | - Shouhei Nagaoka
- Department of Rheumatology, Yokohama Minami Kyosai Hospital, Yokohama, Japan
| | - Kota Shimada
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan.,Department of Rheumatology, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Shoji Sugii
- Department of Rheumatology, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Masao Katayama
- Department of Internal Medicine, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Shunsei Hirohata
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Sagamihara, Japan
| | - Akira Okamoto
- Department of Rheumatology, National Hospital Organization Himeji Medical Center, Himeji, Japan
| | - Noriyuki Chiba
- Department of Rheumatology, National Hospital Organization Morioka Medical Center, Morioka, Japan
| | - Eiichi Suematsu
- Department of Internal Medicine and Rheumatology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Keigo Setoguchi
- Allergy and Immunological Diseases, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shigeto Tohma
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan.,Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
| | | | - Minoru Hasegawa
- Department of Dermatology, University of Fukui, Fukui, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo, Tokyo, Japan
| | - Yasushi Kawaguchi
- Department of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | | | - Naoyuki Tsuchiya
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
9
|
Puentes-Pardo JD, Moreno-SanJuan S, Carazo Á, León J. Heme Oxygenase-1 in Gastrointestinal Tract Health and Disease. Antioxidants (Basel) 2020; 9:antiox9121214. [PMID: 33276470 PMCID: PMC7760122 DOI: 10.3390/antiox9121214] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022] Open
Abstract
Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. HO-1, a stress inducible enzyme, is considered as an anti-oxidative and cytoprotective agent. As many studies suggest, HO-1 is highly expressed in the gastrointestinal tract where it is involved in the response to inflammatory processes, which may lead to several diseases such as pancreatitis, diabetes, fatty liver disease, inflammatory bowel disease, and cancer. In this review, we highlight the pivotal role of HO-1 and its downstream effectors in the development of disorders and their beneficial effects on the maintenance of the gastrointestinal tract health. We also examine clinical trials involving the therapeutic targets derived from HO-1 system for the most common diseases of the digestive system.
Collapse
Affiliation(s)
- Jose D. Puentes-Pardo
- Research Unit, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18011 Granada, Spain
- Correspondence: (J.D.P.-P.); (J.L.); Tel.: +34-958-023-706 (J.L.)
| | - Sara Moreno-SanJuan
- Cytometry and Microscopy Research Service, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain;
| | - Ángel Carazo
- Genomic Research Service, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain;
| | - Josefa León
- Research Unit, Instituto de Investigacion Biosanitaria de Granada, ibs.GRANADA, 18012 Granada, Spain
- Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, 18016 Granada, Spain
- Correspondence: (J.D.P.-P.); (J.L.); Tel.: +34-958-023-706 (J.L.)
| |
Collapse
|
10
|
Singh D, Wasan H, Reeta KH. Heme oxygenase-1 modulation: A potential therapeutic target for COVID-19 and associated complications. Free Radic Biol Med 2020; 161:263-271. [PMID: 33091573 PMCID: PMC7571447 DOI: 10.1016/j.freeradbiomed.2020.10.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to infect hundred thousands of people every day worldwide. Since it is a novel virus, research continues to update the possible therapeutic targets when new evidence regarding COVID-19 are gathered. This article presents an evidence-based hypothesis that activating the heme oxygenase-1 (HO-1) pathway is a potential target for COVID-19. Interferons (IFNs) have broad-spectrum antiviral activity including against SARS-CoV-2. Induction of HO-1 and increase in the heme catabolism end-product confer antiviral activity. IFN activation results in inhibition of viral replication in various viral infections. COVID-19 induced inflammation as well as acute respiratory distress syndrome (ARDS), and coagulopathies are now known major causes of mortality. A protective role of HO-1 induction in inflammation, inflammation-induced coagulation, and ARDS has been reported. Based on an association of HO-1 promoter polymorphisms and disease severity, we propose an evaluation of the status of these polymorphisms in COVID-19 patients who become severely ill. If an association is established, it might be helpful in identifying patients at high risk. Hence, we hypothesize that HO-1 pathway activation could be a therapeutic strategy against COVID-19 and associated complications.
Collapse
Affiliation(s)
- Devendra Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Himika Wasan
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
11
|
Si Z, Wang X. The Neuroprotective and Neurodegeneration Effects of Heme Oxygenase-1 in Alzheimer's Disease. J Alzheimers Dis 2020; 78:1259-1272. [PMID: 33016915 DOI: 10.3233/jad-200720] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by complex pathological and biological features. Notably, extracellular amyloid-β deposits as senile plaques and intracellular aggregation of hyperphosphorylated tau as neurofibrillary tangles remain the primary premortem criterion for the diagnosis of AD. Currently, there exist no disease-modifying therapies for AD, and many clinical trials have failed to show its benefits for patients. Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. Several studies highlight the crucial pathological roles of HO-1 in the molecular processes of AD. The beneficial roles of HO-1 overexpression in AD brains are widely accepted due to its ability to convert pro-oxidant heme to biliverdin and bilirubin (antioxidants), which promote restoration of a suitable tissue redox microenvironment. However, the intracellular oxidative stress might be amplified by metabolites of HO-1 and exacerbate the progression of AD under certain circumstances. Several lines of evidence have demonstrated that upregulated HO-1 is linked to tauopathies, neuronal damage, and synapse aberrations in AD. Here, we review the aspects of the molecular mechanisms by which HO-1 regulates AD and the latest information on the pathobiology of AD. We further highlight the neuroprotective and neurodystrophic actions of HO-1 and the feasibility of HO-1 as a therapeutic target for AD.
Collapse
Affiliation(s)
- Zizhen Si
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Xidi Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Szade K, Zukowska M, Szade A, Nowak W, Skulimowska I, Ciesla M, Bukowska‐Strakova K, Gulati GS, Kachamakova‐Trojanowska N, Kusienicka A, Einwallner E, Kijowski J, Czauderna S, Esterbauer H, Benes V, L Weissman I, Dulak J, Jozkowicz A. Heme oxygenase-1 deficiency triggers exhaustion of hematopoietic stem cells. EMBO Rep 2020; 21:e47895. [PMID: 31885181 PMCID: PMC7001511 DOI: 10.15252/embr.201947895] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 11/14/2019] [Accepted: 11/22/2019] [Indexed: 01/01/2023] Open
Abstract
While intrinsic changes in aging hematopoietic stem cells (HSCs) are well characterized, it remains unclear how extrinsic factors affect HSC aging. Here, we demonstrate that cells in the niche-endothelial cells (ECs) and CXCL12-abundant reticular cells (CARs)-highly express the heme-degrading enzyme, heme oxygenase 1 (HO-1), but then decrease its expression with age. HO-1-deficient animals (HO-1-/- ) have altered numbers of ECs and CARs that produce less hematopoietic factors. HSCs co-cultured in vitro with HO-1-/- mesenchymal stromal cells expand, but have altered kinetic of growth and differentiation of derived colonies. HSCs from young HO-1-/- animals have reduced quiescence and regenerative potential. Young HO-1-/- HSCs exhibit features of premature exhaustion on the transcriptional and functional level. HO-1+/+ HSCs transplanted into HO-1-/- recipients exhaust their regenerative potential early and do not reconstitute secondary recipients. In turn, transplantation of HO-1-/- HSCs to the HO-1+/+ recipients recovers the regenerative potential of HO-1-/- HSCs and reverses their transcriptional alterations. Thus, HSC-extrinsic activity of HO-1 prevents HSCs from premature exhaustion and may restore the function of aged HSCs.
Collapse
Affiliation(s)
- Krzysztof Szade
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
| | - Monika Zukowska
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| | - Agata Szade
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| | - Witold Nowak
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| | - Izabella Skulimowska
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| | - Maciej Ciesla
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| | - Karolina Bukowska‐Strakova
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
- Department of Clinical ImmunologyInstitute of PediatricsJagiellonian University Medical CollegeKrakowPoland
| | - Gunsagar Singh Gulati
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
| | - Neli Kachamakova‐Trojanowska
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
- Malopolska Centre of BiotechnologyJagiellonian UniversityKrakowPoland
| | - Anna Kusienicka
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| | - Elisa Einwallner
- Department of Laboratory MedicineCenter of Translational ResearchMedical University of ViennaViennaAustria
| | - Jacek Kijowski
- Department of TransplantationInstitute of PediatricsJagiellonian University Medical CollegeKrakowPoland
| | - Szymon Czauderna
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| | - Harald Esterbauer
- Department of Laboratory MedicineCenter of Translational ResearchMedical University of ViennaViennaAustria
| | | | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
| | - Jozef Dulak
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
- Malopolska Centre of BiotechnologyJagiellonian UniversityKrakowPoland
| | - Alicja Jozkowicz
- Department of Medical BiotechnologyFaculty of Biochemistry, Biophysics and BiotechnologyJagiellonian UniversityKrakowPoland
| |
Collapse
|
13
|
Kisoi M, Moritsugu M, Imai M, Fukumoto K, Sakaguchi Y, Murata S, Kawai S, Ichikawa A, Kinoshita K. Rapid and Cost-Effective Genotyping Protocol for Angiotensin-Converting Enzyme Insertion/Deletion (Ins/Del) Polymorphism from Saliva. Biol Pharm Bull 2019; 42:1345-1349. [PMID: 31366869 DOI: 10.1248/bpb.b19-00110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
DNA extraction and purification have been generally considered to be required for PCR assay. We demonstrated a new protocol using biological specimens directly as templates for real-time PCR with melting curve analysis. We confirmed the melting curve analysis was particularly suitable for the identification of the insertion/deletion (Ins/Del) polymorphism of the angiotensin-converting enzyme (ACE) gene. The new protocol we developed can be set up using simple and complete PCR analysis including data interpretation in under four hours with additional advantages of application for large-scale clinical research, diagnostics, and epidemiological studies at low cost.
Collapse
Affiliation(s)
- Madoka Kisoi
- School of Pharmaceutical Sciences, Mukogawa Women's University
| | | | - Miho Imai
- School of Pharmaceutical Sciences, Mukogawa Women's University
| | - Kae Fukumoto
- School of Pharmaceutical Sciences, Mukogawa Women's University
| | - Yui Sakaguchi
- School of Pharmaceutical Sciences, Mukogawa Women's University
| | - Shigenori Murata
- School of Pharmaceutical Sciences, Mukogawa Women's University.,Institute of Biosciences, Mukogawa Women's University
| | - Sayuri Kawai
- Institute of Biosciences, Mukogawa Women's University
| | | | - Kenji Kinoshita
- School of Pharmaceutical Sciences, Mukogawa Women's University.,Institute of Biosciences, Mukogawa Women's University
| |
Collapse
|
14
|
Bachelet D, Albert T, Mbogning C, Hässler S, Zhang Y, Schultze-Strasser S, Repessé Y, Rayes J, Pavlova A, Pezeshkpoor B, Liphardt K, Davidson JE, Hincelin-Méry A, Dönnes P, Lacroix-Desmazes S, Königs C, Oldenburg J, Broët P. Risk stratification integrating genetic data for factor VIII inhibitor development in patients with severe hemophilia A. PLoS One 2019; 14:e0218258. [PMID: 31194850 PMCID: PMC6564000 DOI: 10.1371/journal.pone.0218258] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/29/2019] [Indexed: 12/20/2022] Open
Abstract
Replacement therapy in severe hemophilia A leads to factor VIII (FVIII) inhibitors in 30% of patients. Factor VIII gene (F8) mutation type, a family history of inhibitors, ethnicity and intensity of treatment are established risk factors, and were included in two published prediction tools based on regression models. Recently investigated immune regulatory genes could also play a part in immunogenicity. Our objective is to identify bio-clinical and genetic markers for FVIII inhibitor development, taking into account potential genetic high order interactions. The study population consisted of 593 and 79 patients with hemophilia A from centers in Bonn and Frankfurt respectively. Data was collected in the European ABIRISK tranSMART database. A subset of 125 severely affected patients from Bonn with reliable information on first treatment was selected as eligible for risk stratification using a hybrid tree-based regression model (GPLTR). In the eligible subset, 58 (46%) patients developed FVIII inhibitors. Among them, 49 (84%) were “high risk” F8 mutation type. 19 (33%) had a family history of inhibitors. The GPLTR model, taking into account F8 mutation risk, family history of inhibitors and product type, distinguishes two groups of patients: a high-risk group for immunogenicity, including patients with positive HLA-DRB1*15 and genotype G/A and A/A for IL-10 rs1800896, and a low-risk group of patients with negative HLA-DRB1*15 / HLA-DQB1*02 and T/T or G/T for CD86 rs2681401. We show associations between genetic factors and the occurrence of FVIII inhibitor development in severe hemophilia A patients taking into account for high-order interactions using a generalized partially linear tree-based approach.
Collapse
Affiliation(s)
- Delphine Bachelet
- CESP, INSERM UMR 1018, Faculty of Medicine, Paris-Sud University, UVSQ, Paris-Saclay University, Villejuif, France
| | - Thilo Albert
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Bonn, Germany
| | - Cyprien Mbogning
- CESP, INSERM UMR 1018, Faculty of Medicine, Paris-Sud University, UVSQ, Paris-Saclay University, Villejuif, France
| | - Signe Hässler
- CESP, INSERM UMR 1018, Faculty of Medicine, Paris-Sud University, UVSQ, Paris-Saclay University, Villejuif, France
| | - Yuan Zhang
- CESP, INSERM UMR 1018, Faculty of Medicine, Paris-Sud University, UVSQ, Paris-Saclay University, Villejuif, France
| | - Stephan Schultze-Strasser
- University Hospital Frankfurt, Goethe University, Department of Pediatrics, Molecular Haemostasis and Immunodeficiency, Frankfurt am Main, Germany
| | | | - Julie Rayes
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Anna Pavlova
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Bonn, Germany
| | - Behnaz Pezeshkpoor
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Bonn, Germany
| | - Kerstin Liphardt
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Bonn, Germany
| | | | | | | | - Sébastien Lacroix-Desmazes
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Université Paris Descartes, Sorbonne Paris Cité, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Christoph Königs
- University Hospital Frankfurt, Goethe University, Department of Pediatrics, Molecular Haemostasis and Immunodeficiency, Frankfurt am Main, Germany
| | - Johannes Oldenburg
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, Bonn, Germany
| | - Philippe Broët
- CESP, INSERM UMR 1018, Faculty of Medicine, Paris-Sud University, UVSQ, Paris-Saclay University, Villejuif, France
- AP-HP, Paris-Sud University Hospitals, Villejuif, France
- * E-mail:
| | | |
Collapse
|
15
|
Lenglet H, Schmitt C, Grange T, Manceau H, Karboul N, Bouchet-Crivat F, Robreau AM, Nicolas G, Lamoril J, Simonin S, Mirmiran A, Karim Z, Casalino E, Deybach JC, Puy H, Peoc'h K, Gouya L. From a dominant to an oligogenic model of inheritance with environmental modifiers in acute intermittent porphyria. Hum Mol Genet 2019; 27:1164-1173. [PMID: 29360981 DOI: 10.1093/hmg/ddy030] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/09/2018] [Indexed: 01/02/2023] Open
Abstract
Acute intermittent porphyria (AIP) is a disease affecting the heme biosynthesis pathway caused by mutations of the hydroxymethylbilane synthase (HMBS) gene. AIP is thought to display autosomal dominant inheritance with incomplete penetrance. We evaluated the prevalence, penetrance and heritability of AIP, in families with the disease from the French reference center for porphyria (CFP) (602 overt patients; 1968 relatives) and the general population, using Exome Variant Server (EVS; 12 990 alleles) data. The pathogenicity of the 42 missense variants identified was assessed in silico, and in vitro, by measuring residual HMBS activity of the recombinant protein. The minimal estimated prevalence of AIP in the general population was 1/1299. Thus, 50 000 subjects would be expected to carry the AIP genetic trait in France. Penetrance was estimated at 22.9% in families with AIP, but at only 0.5-1% in the general population. Intrafamily correlation studies showed correlations to be strong overall and modulated by kinship and the area in which the person was living, demonstrating strong influences of genetic and environmental modifiers on inheritance. Null alleles were associated with a more severe phenotype and a higher penetrance than for other mutant alleles. In conclusion, the striking difference in the penetrance of HMBS mutations between the general population and the French AIP families suggests that AIP inheritance does not follow the classical autosomal dominant model, instead of being modulated by strong environmental and genetic factors independent from HMBS. An oligogenic inheritance model with environmental modifiers might better explain AIP penetrance and heritability.
Collapse
Affiliation(s)
- Hugo Lenglet
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Département des Urgences, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Bichat, F-75018 Paris, France
| | - Caroline Schmitt
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Université Paris Diderot, F-75018 Paris, France.,Centre Français des Porphyries, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Louis Mourier, F-92701 Colombes, France
| | - Thomas Grange
- INSERM UMR_S1048 Laboratory for Vascular Translational Science (LVTS) Université Paris Diderot, F-75018 Paris, France
| | - Hana Manceau
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Laboratoire de Biochimie, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Beaujon, 92110 Clichy, France and DHU Unity
| | - Narjesse Karboul
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France
| | - Florian Bouchet-Crivat
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France
| | - Anne-Marie Robreau
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Centre Français des Porphyries, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Louis Mourier, F-92701 Colombes, France
| | - Gael Nicolas
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France
| | - Jerôme Lamoril
- Département de Génétique, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Bichat, F-75018 Paris, France
| | - Sylvie Simonin
- Centre Français des Porphyries, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Louis Mourier, F-92701 Colombes, France
| | - Arienne Mirmiran
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France
| | - Zoubida Karim
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France
| | - Enrique Casalino
- Département des Urgences, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Bichat, F-75018 Paris, France.,Université Paris Diderot, F-75018 Paris, France
| | - Jean-Charles Deybach
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Université Paris Diderot, F-75018 Paris, France.,Centre Français des Porphyries, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Louis Mourier, F-92701 Colombes, France
| | - Hervé Puy
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Université Paris Diderot, F-75018 Paris, France.,Centre Français des Porphyries, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Louis Mourier, F-92701 Colombes, France.,Laboratory of Excellence GR-Ex, F-75015 Paris, France
| | - Katell Peoc'h
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Université Paris Diderot, F-75018 Paris, France.,Laboratoire de Biochimie, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Beaujon, 92110 Clichy, France and DHU Unity.,Laboratory of Excellence GR-Ex, F-75015 Paris, France
| | - Laurent Gouya
- UMRs 1149, Centre de Recherche sur l'Inflammation, Institut National de la Santé et de la Recherche Médicale, F-75018 Paris, France.,Université Paris Diderot, F-75018 Paris, France.,Centre Français des Porphyries, Assistance Publique-Hôpitaux de Paris, HUPNVS, Hôpital Louis Mourier, F-92701 Colombes, France.,Laboratory of Excellence GR-Ex, F-75015 Paris, France
| |
Collapse
|
16
|
The sinister face of heme oxygenase-1 in brain aging and disease. Prog Neurobiol 2019; 172:40-70. [DOI: 10.1016/j.pneurobio.2018.06.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 11/23/2022]
|
17
|
Tsai MT, Tarng DC. Beyond a Measure of Liver Function-Bilirubin Acts as a Potential Cardiovascular Protector in Chronic Kidney Disease Patients. Int J Mol Sci 2018; 20:ijms20010117. [PMID: 30597982 PMCID: PMC6337523 DOI: 10.3390/ijms20010117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/17/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023] Open
Abstract
Bilirubin is a well-known neurotoxin in newborn infants; however, current evidence has shown that a higher serum bilirubin concentration in physiological ranges is associated with a lower risk for the development and progression of both chronic kidney disease (CKD) and cardiovascular disease (CVD) in adults. The protective mechanisms of bilirubin in CVD, CKD, and associated mortality may be ascribed to its antioxidant and anti-inflammatory properties. Bilirubin further improves insulin sensitivity, reduces low-density lipoprotein cholesterol levels and inhibits platelet activation in at-risk individuals. These effects are expected to maintain normal vascular homeostasis and thus reduce the incidence of CKD and the risks of cardiovascular complications and death. In this review, we highlight the recent advances in the biological actions of bilirubin in the pathogenesis of CVD and CKD progression, and further propose that targeting bilirubin metabolism could be a potential approach to ameliorate morbidity and mortality in CKD patients.
Collapse
Affiliation(s)
- Ming-Tsun Tsai
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Department and Institute of Physiology, National Yang-Ming University, Taipei 11217, Taiwan.
| |
Collapse
|
18
|
Heme Oxygenase 1 in the Nervous System: Does It Favor Neuronal Cell Survival or Induce Neurodegeneration? Int J Mol Sci 2018; 19:ijms19082260. [PMID: 30071692 PMCID: PMC6121636 DOI: 10.3390/ijms19082260] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023] Open
Abstract
Heme oxygenase 1 (HO-1) up-regulation is recognized as a pivotal mechanism of cell adaptation to stress. Under control of different transcription factors but with a prominent role played by Nrf2, HO-1 induction is crucial also in nervous system response to damage. However, several lines of evidence have highlighted that HO-1 expression is associated to neuronal damage and neurodegeneration especially in Alzheimer’s and Parkinson’s diseases. In this review, we summarize the current literature regarding the role of HO-1 in nervous system pointing out different molecular mechanisms possibly responsible for HO-1 up-regulation in nervous system homeostasis and neurodegeneration.
Collapse
|
19
|
Ibuki A, Minematsu T, Yoshida M, Iizaka S, Matsumoto M, Sugama J, Sanada H. Microsatellite polymorphism in the Heme oxygenase-1 gene promoter is associated with dermal collagen density in Japanese obese male subjects. PLoS One 2018; 13:e0199994. [PMID: 30024897 PMCID: PMC6053161 DOI: 10.1371/journal.pone.0199994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 06/18/2018] [Indexed: 11/19/2022] Open
Abstract
We previously reported elevated oxidative stress-related mechanical vulnerabilities of the skin as sparse distributions of hyperechoic areas. Although this helped establish a personalized skin care system to prevent skin disorders related to mechanical stress, obesity-related skin vulnerability involves individual differences. Here, we hypothesized that individual differences are caused by polymorphisms of GT repetitive sequences in the heme oxygenase1 (HMOX1) promoter region, which encodes an antioxidant enzyme. This cross-sectional study enrolled healthy male volunteers in a walking classroom aimed at weight control. Subjects with a body mass index <25 kg/m2 were classified as non-obese and those with body mass index ≥25 kg/m2 were classified as obese. Subject skin was categorized into sparse dermis or normal groups according to the distribution of hyperechoic areas by high-resolution skin ultrasonography (20 MHz). Genomic DNA and mRNA extracted from three body hairs with attached follicle cells were used to analyze GT repetitive sequences of the HMOX1 promoter, HMOX1 mRNA expression levels, and oxidative stress levels (8-hydroxy-2’-deoxyguanosine). Classifications of GT repetitive sequence of HMOX1 promoter were Short (<27 times) and Long (≥27 times). Higher numbers of subjects with sparse dermis were in the obese group compared with the non-obese group. In obese subjects, the number of subjects that had the Long allele of the HMOX1 promoter with sparse dermis was significantly higher compared with the normal group, whereas no association was observed between the polymorphism and ultrasonographic features in non-obese subjects. Thus, HMOX1 polymorphisms detected a risk of low collagen density in Japanese obese male subjects.
Collapse
Affiliation(s)
- Ai Ibuki
- Department of Biological Science and Nursing, School of Medicine, Yokohama City University, Kanagawa, Japan
- Department of Gerontological Nursing/ Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeo Minematsu
- Department of Skincare Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mikako Yoshida
- Department of Imaging Nursing Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinji Iizaka
- School of Nutrition, College of Nursing and Nutrition, Shukutoku University, Chiba, Japan
| | - Masaru Matsumoto
- Department of Imaging Nursing Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junko Sugama
- Advanced Health Care Science Research Unit Innovative Integrated Bio-Research Core Institute for Frontier Science Initiative, Kanazawa University, Ishikawa, Japan
| | - Hiromi Sanada
- Department of Gerontological Nursing/ Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
20
|
The macrophage heme-heme oxygenase-1 system and its role in inflammation. Biochem Pharmacol 2018; 153:159-167. [PMID: 29452096 DOI: 10.1016/j.bcp.2018.02.010] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/12/2018] [Indexed: 02/07/2023]
Abstract
Heme oxygenase (HO)-1, the inducible isoform of the heme-degrading enzyme HO, plays a critical role in inflammation and iron homeostasis. Regulatory functions of HO-1 are mediated via the catalytic breakdown of heme, which is an iron-containing tetrapyrrole complex with potential pro-oxidant and pro-inflammatory effects. In addition, the HO reaction produces the antioxidant and anti-inflammatory compounds carbon monoxide (CO) and biliverdin, subsequently converted into bilirubin, along with iron, which is reutilized for erythropoiesis. HO-1 is up-regulated by a plethora of stimuli and injuries in most cell types and tissues and provides salutary effects by restoring physiological homeostasis. Notably, HO-1 exhibits critical immuno-modulatory functions in macrophages, which are a major cell population of the mononuclear phagocyte system. Macrophages play key roles as sentinels and regulators of the immune system and HO-1 in these cells appears to be of critical importance for driving resolution of inflammatory responses. In this review, the complex functions and regulatory mechanisms of HO-1 in macrophages will be high-lighted. A particular focus will be the intricate interactions of HO-1 with its substrate heme, which play a contradictory role in distinct physiological and pathophysiological settings. The therapeutic potential of targeted modulation of the macrophage heme-HO-1 system will be discussed in the context of inflammatory disorders.
Collapse
|
21
|
Kah J, Volz T, Lütgehetmann M, Groth A, Lohse AW, Tiegs G, Sass G, Dandri M. Haem oxygenase-1 polymorphisms can affect HCV replication and treatment responses with different efficacy in humanized mice. Liver Int 2017; 37:1128-1137. [PMID: 27992676 DOI: 10.1111/liv.13347] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS Enhancement of host anti-oxidant enzymes, such as haemoxygenase-1, may attenuate virus-mediated hepatocyte injury, while the induction of HO-1 by cobalt-protoporphyrin-IX (CoPP) administration, as the application of its haem degradation product biliverdin (BV), was shown to hinder HCV replication in vitro. In addition, (GT)n -repeats length in the polymorphic region of the HO-1 promoter may affect HO-1 expression and responsiveness to infection and disease severity. Aim of this study was to investigate the antiviral and hepatoprotective effects of CoPP-mediated HO-1 induction, alone or in combination with interferon alpha (peg-IFNα), in HCV-infected mice harbouring hepatocytes from donors with different HO-1-promoter polymorphisms. METHODS Upon establishment of HCV infection, CoPP, BV and peg-IFNα were given alone or in combination. Viraemia changes and intrahepatic human gene expression were determined by qRT-PCR and immunohistochemistry. RESULTS CoPP administration increased human HO-1 expression and significantly reduced viraemia, although changes correlated with promoter length (Δ0.5log and Δ2log reduction with medium- and short-polymorphism respectively). Polymorphisms did not influence BV-mediated antiviral effects (Δ1log). Notably, HO-1 induction attenuated basal HCV-driven enhancement of interferon genes and pro-inflammatory cytokines, both in cells with short- or medium-polymorphisms. Moreover, simultaneous administration of CoPP and peg-IFNα reduced viraemia even stronger (median 3log), whereas 1log viraemia reduction was determined in mice receiving peg-IFNα monotherapy. CONCLUSIONS Although the protective function of HO-1 could be elicited in vivo with both host polymorphisms, the strength of HO-1 induction and suppression of HCV occurred in a polymorphism-dependent manner, indicating that host-genetic determinants may affect disease progression and infection outcome.
Collapse
Affiliation(s)
- Janine Kah
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tassilo Volz
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Groth
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, Hamburg, Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Sass
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maura Dandri
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, Hamburg, Germany
| |
Collapse
|
22
|
Freystaetter K, Andreas M, Bilban M, Perkmann T, Kaider A, Masetti M, Kocher A, Wolzt M, Zuckermann A. The recipient's heme oxygenase-1 promoter region polymorphism is associated with cardiac allograft vasculopathy. Transpl Int 2017; 30:510-518. [DOI: 10.1111/tri.12935] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/25/2016] [Accepted: 01/31/2017] [Indexed: 01/18/2023]
Affiliation(s)
| | - Martin Andreas
- Division of Cardiac Surgery; Medical University of Vienna; Vienna Austria
| | - Martin Bilban
- Department of Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine; Medical University of Vienna; Vienna Austria
| | - Alexandra Kaider
- Section for Clinical Biometrics; Center for Medical Statistics Informatics and Intelligent Systems; Medical University of Vienna; Vienna Austria
| | - Marco Masetti
- Department of Cardiology; Bologna University Hospital; Bologna Italy
| | - Alfred Kocher
- Division of Cardiac Surgery; Medical University of Vienna; Vienna Austria
| | - Michael Wolzt
- Department of Clinical Pharmacology; Medical University of Vienna; Vienna Austria
| | - Andreas Zuckermann
- Division of Cardiac Surgery; Medical University of Vienna; Vienna Austria
| |
Collapse
|
23
|
(GT)n Repeat Polymorphism in Heme Oxygenase-1 (HO-1) Correlates with Clinical Outcome after Myeloablative or Nonmyeloablative Allogeneic Hematopoietic Cell Transplantation. PLoS One 2016; 11:e0168210. [PMID: 27997582 PMCID: PMC5172582 DOI: 10.1371/journal.pone.0168210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/28/2016] [Indexed: 11/19/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is a treatment for various hematologic diseases where efficacy of treatment is in part based on the graft versus tumour (GVT) activity of cells in the transplant. The cytoprotective enzyme heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation and it has been shown to exert anti-inflammatory functions. In humans a (GT)n repeat polymorphism regulates the expression of HO-1. We conducted fragment length analyses of the (GT)n repeat in the promotor region of the gene for HO-1 in DNA from donors and recipients receiving allogeneic myeloablative- (MA) (n = 110) or nonmyeloablative- (NMA-) (n = 250) HCT. Subsequently, we compared the length of the (GT)n repeat with clinical outcome after HCT. We demonstrated that transplants from a HO-1high donor after MA-conditioning (n = 13) is associated with higher relapse incidence at 3 years (p = 0.01, n = 110). In the NMA-conditioning setting transplantation of HO-1low donor cells into HO-1low recipients correlated significantly with decreased relapse related mortality (RRM) and longer progression free survival (PFS) (p = 0.03 and p = 0.008, respectively). Overall, our findings suggest that HO-1 may play a role for the induction of GVT effect after allogeneic HCT.
Collapse
|
24
|
Zhou H, Ying X, Liu Y, Ye S, Yan J, 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 2016; 21:894-903. [PMID: 27998018 PMCID: PMC5387120 DOI: 10.1111/jcmm.13028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 10/05/2016] [Indexed: 11/27/2022] Open
Abstract
Heme oxygenase 1 (HMOX1) plays an important role in the development of chronic obstructive pulmonary disease (COPD). However, the association of HMOX1 length polymorphism in promoter region to the risk and severity of COPD has not been well studied. In this study, we searched the databases including PubMed, EMBASE, Cochrane Library and China National Knowledge Infrastructure (CNKI) and extracted the information from related articles. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to study the effect of HMOX1 polymorphism on the risk and severity of COPD. As a result, nine studies were included for this meta‐analysis. Higher frequencies of L allele and type I genotype (containing at least one L allele) were found in patients with COPD (for L allele, OR 2.02, 95% CI: 1.32–3.11, P = 0.001; for type I genotype, OR 1.82, 95% CI: 1.28–2.61, P = 0.001), especially in Asian population (for L allele, OR 2.23, 95% CI: 1.68–2.95, P < 0.001; for type I genotype, OR 2.02, 95% CI: 1.51–2.70, P < 0.001). Genotyping method, source of control subjects, literature quality and language also affected the results to some extent. However, there was little difference in HMOX1 genotypes distribution in patients with COPD with different severity. Our study indicated L allele and type I genotype were related to the susceptibility but not the severity of COPD.
Collapse
Affiliation(s)
- Hongbin Zhou
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Xiwang Ying
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Yuanshun Liu
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Sa Ye
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Jianping Yan
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Yaqing Li
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Enhancement of DEN-induced liver tumorigenesis in heme oxygenase-1 G143H mutant transgenic mice. Biochem Biophys Res Commun 2016; 481:169-175. [PMID: 27810363 DOI: 10.1016/j.bbrc.2016.10.148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 12/25/2022]
Abstract
Heme oxygenase (HO) is the rate-limiting enzyme in heme metabolism. HO-1 exhibits anti-oxidative and anti-inflammatory function via the actions of its metabolite, respectively. A growing body of evidence demonstrates that HO-1 is implicated in the pathogenesis and progression of several types of cancer. However, whether HO-1 takes part in healthy-premalignant-malignant transformation is still undefined. In this study, we took advantage of transgenic mice which over-expressed HO-1 dominant negative mutant (HO-1 G143H) and observed its susceptibility to DEN-induced hepatocarcinogenesis. Our results indicate that HO-1 G143H mutant accelerates the progression of tumorigenesis and tumor growth. The mechanism is closely related to enhancement of ROS production which induce more hepatocytes death and secretion of inflammatory cytokines, proliferation of surviving hepatocytes. Our result provides the direct evidence that HO-1 plays an important protective role in liver carcinogenesis. Alternatively, we suggest the possible explanation on effect of HO-1 promoter polymorphism which involved in tumorigenesis.
Collapse
|
26
|
Abstract
Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. While initially considered to be waste products, CO and biliverdin/bilirubin have been shown over the last 20 years to modulate key cellular processes, such as inflammation, cell proliferation, and apoptosis, as well as antioxidant defense. This shift in paradigm has led to the importance of heme oxygenases and their products in cell physiology now being well accepted. The identification of the two human cases thus far of heme oxygenase deficiency and the generation of mice deficient in Hmox1 or Hmox2 have reiterated a role for these enzymes in both normal cell function and disease pathogenesis, especially in the context of cardiovascular disease. This review covers the current knowledge on the function of both Hmox1 and Hmox2 at both a cellular and tissue level in the cardiovascular system. Initially, the roles of heme oxygenases in vascular health and the regulation of processes central to vascular diseases are outlined, followed by an evaluation of the role(s) of Hmox1 and Hmox2 in various diseases such as atherosclerosis, intimal hyperplasia, myocardial infarction, and angiogenesis. Finally, the therapeutic potential of heme oxygenases and their products are examined in a cardiovascular disease context, with a focus on how the knowledge we have gained on these enzymes may be capitalized in future clinical studies.
Collapse
Affiliation(s)
- Anita Ayer
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Abolfazl Zarjou
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Anupam Agarwal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
27
|
Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: an evolutionarily conserved mechanism. Cell Mol Life Sci 2016; 73:3221-47. [PMID: 27100828 PMCID: PMC4967105 DOI: 10.1007/s00018-016-2223-0] [Citation(s) in RCA: 1665] [Impact Index Per Article: 208.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/12/2022]
Abstract
The multifunctional regulator nuclear factor erythroid 2-related factor (Nrf2) is considered not only as a cytoprotective factor regulating the expression of genes coding for anti-oxidant, anti-inflammatory and detoxifying proteins, but it is also a powerful modulator of species longevity. The vertebrate Nrf2 belongs to Cap 'n' Collar (Cnc) bZIP family of transcription factors and shares a high homology with SKN-1 from Caenorhabditis elegans or CncC found in Drosophila melanogaster. The major characteristics of Nrf2 are to some extent mimicked by Nrf2-dependent genes and their proteins including heme oxygenase-1 (HO-1), which besides removing toxic heme, produces biliverdin, iron ions and carbon monoxide. HO-1 and their products exert beneficial effects through the protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. On the other hand, the disturbances in the proper HO-1 level are associated with the pathogenesis of some age-dependent disorders, including neurodegeneration, cancer or macular degeneration. This review summarizes our knowledge about Nrf2 and HO-1 across different phyla suggesting their conservative role as stress-protective and anti-aging factors.
Collapse
Affiliation(s)
- Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Milena Damulewicz
- Department of Cell Biology and Imaging, Faculty of Biology and Earth Sciences, Jagiellonian University, Krakow, Poland
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Faculty of Biology and Earth Sciences, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
28
|
Abstract
SIGNIFICANCE Acute kidney injury (AKI) and chronic kidney disease (CKD) represent a considerable burden in healthcare. The heme oxygenase (HO) system plays an important role in regulating oxidative stress and is protective in a variety of human and animal models of kidney disease. Preclinical studies of the HO system have led to the development of several clinical trials targeting the enzyme or its products. RECENT ADVANCES Connection of HO, ferritin, and other proteins involved in iron regulation has provided important insight into mechanisms of damage in AKI. Also, HO-1 expression is important in the pathogenesis of hypertension, diabetic kidney disease, and progression to end-stage renal disease. CRITICAL ISSUES Despite intriguing discoveries, no drugs targeting the HO system have been translated to the clinic. Meanwhile, treatments for AKI and CKD are urgently needed. Many factors have likely contributed to challenges in clinical translation, including variation in animal models, difficulties in obtaining human tissue, and complexity of the disease processes being studied. FUTURE DIRECTIONS The HO system represents a promising avenue of investigation that may lead to targeted therapeutics. Tissue-specific gene modulation, widening the scope of animal studies, and continued clinical research will provide valuable insight into the role HO plays in kidney homeostasis and disease. Antioxid. Redox Signal. 25, 165-183.
Collapse
Affiliation(s)
- Jeremie M Lever
- 1 Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham , Birmingham, Alabama
| | - Ravindra Boddu
- 1 Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham , Birmingham, Alabama
| | - James F George
- 2 Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham , Birmingham, Alabama
| | - Anupam Agarwal
- 1 Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham , Birmingham, Alabama.,3 Birmingham Veterans Administration Medical Center , Birmingham, Alabama
| |
Collapse
|
29
|
Adams RH, Blackmon H, Reyes-Velasco J, Schield DR, Card DC, Andrew AL, Waynewood N, Castoe TA. Microsatellite landscape evolutionary dynamics across 450 million years of vertebrate genome evolution. Genome 2016; 59:295-310. [PMID: 27064176 DOI: 10.1139/gen-2015-0124] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The evolutionary dynamics of simple sequence repeats (SSRs or microsatellites) across the vertebrate tree of life remain largely undocumented and poorly understood. In this study, we analyzed patterns of genomic microsatellite abundance and evolution across 71 vertebrate genomes. The highest abundances of microsatellites exist in the genomes of ray-finned fishes, squamate reptiles, and mammals, while crocodilian, turtle, and avian genomes exhibit reduced microsatellite landscapes. We used comparative methods to infer evolutionary rates of change in microsatellite abundance across vertebrates and to highlight particular lineages that have experienced unusually high or low rates of change in genomic microsatellite abundance. Overall, most variation in microsatellite content, abundance, and evolutionary rate is observed among major lineages of reptiles, yet we found that several deeply divergent clades (i.e., squamate reptiles and mammals) contained relatively similar genomic microsatellite compositions. Archosauromorph reptiles (turtles, crocodilians, and birds) exhibit reduced genomic microsatellite content and the slowest rates of microsatellite evolution, in contrast to squamate reptile genomes that have among the highest rates of microsatellite evolution. Substantial branch-specific shifts in SSR content in primates, monotremes, rodents, snakes, and fish are also evident. Collectively, our results support multiple major shifts in microsatellite genomic landscapes among vertebrates.
Collapse
Affiliation(s)
- Richard H Adams
- a Department of Biology, 501 S. Nedderman Dr., University of Texas at Arlington, TX 76019, USA
| | - Heath Blackmon
- b Department of Ecology, Evolution & Behavior, 1987 Upper Buford Cir., University of Minnesota, Saint Paul, MN 55108-6097, USA
| | - Jacobo Reyes-Velasco
- a Department of Biology, 501 S. Nedderman Dr., University of Texas at Arlington, TX 76019, USA
| | - Drew R Schield
- a Department of Biology, 501 S. Nedderman Dr., University of Texas at Arlington, TX 76019, USA
| | - Daren C Card
- a Department of Biology, 501 S. Nedderman Dr., University of Texas at Arlington, TX 76019, USA
| | - Audra L Andrew
- a Department of Biology, 501 S. Nedderman Dr., University of Texas at Arlington, TX 76019, USA
| | - Nyimah Waynewood
- a Department of Biology, 501 S. Nedderman Dr., University of Texas at Arlington, TX 76019, USA
| | - Todd A Castoe
- a Department of Biology, 501 S. Nedderman Dr., University of Texas at Arlington, TX 76019, USA
| |
Collapse
|
30
|
Wu MM, Lee CH, Hsu LI, Cheng WF, Lee TC, Wang YH, Chiou HY, Chen CJ. Effect of heme oxygenase-1 gene promoter polymorphism on cancer risk by histological subtype: A prospective study in arseniasis-endemic areas in Taiwan. Int J Cancer 2015; 138:1875-86. [PMID: 26566708 DOI: 10.1002/ijc.29926] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 11/03/2015] [Indexed: 11/07/2022]
Abstract
Heme oxygenase (HO)-1 is upregulated by many stressful stimuli, including arsenic. A GT-repeat ((GT)n) polymorphism in the HO-1 gene promoter inversely modulates the levels of HO-1 induction. Previous HO-1 (GT)n polymorphism studies in relation to cancer risk have shown disparate results. We prospectively investigated the associations between HO-1 (GT)n polymorphism and cancer risk related to arsenic from drinking water. Totally, 1,013 participants from community-based cohorts of arseniasis-endemic areas in Taiwan were followed for 13 years. Allelic polymorphisms were classified into long (L, ≥ 27 (GT)n) and short (S, <27 (GT)n). Newly developed cases were identified through linkage with National Cancer Registry of Taiwan. Multivariate Cox proportional hazard methods were used to evaluate effects of the HO-1 polymorphism alone or combined with arsenic exposure. Results showed that participants with the S/S genotype had an increased risk of Bowen's disease (HR = 10.49; 95% CI: 2.77-39.7), invasive skin cancer (HR = 2.99; 95% CI: 1.13-7.87), and lung squamous cell carcinoma (HR = 3.39; 95% CI: 1.15-9.95) versus those with L/S or L/L genotype. The S/S genotype combined with high arsenic exposure (>300 μg/L) had a greater risk of skin cancer compared to the genotype alone. Consistent with previous findings, participants with the S-allele had a reduced risk of lung adenocarcinoma (HR = 0.21; 95% CI: 0.03-0.68) versus those with L/L genotype. There were no significant differences in risk of urothelial carcinoma among the three genotypes. Associations of HO-1 (GT)n polymorphism with cancer risk differs by histological subtype and the polymorphism should be considered a modifier in the risk assessment of arsenic exposure.
Collapse
Affiliation(s)
- Meei-Maan Wu
- School of Public Health, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Hung Lee
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung and Chang Gung University College of Medicine, Taiwan
| | - Ling-I Hsu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wen-Fang Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Te-Chang Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuang-Hung Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hung-Yi Chiou
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Chien-Jen Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
31
|
Saraf SL, Zhang X, Shah B, Kanias T, Gudehithlu KP, Kittles R, Machado RF, Arruda JAL, Gladwin MT, Singh AK, Gordeuk VR. Genetic variants and cell-free hemoglobin processing in sickle cell nephropathy. Haematologica 2015. [PMID: 26206798 DOI: 10.3324/haematol.2015.124875] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intravascular hemolysis and hemoglobinuria are associated with sickle cell nephropathy. ApoL1 is involved in cell-free hemoglobin scavenging through association with haptoglobin-related protein. APOL1 G1/G2 variants are the strongest genetic predictors of kidney disease in the general African-American population. A single report associated APOL1 G1/G2 with sickle cell nephropathy. In 221 patients with sickle cell disease at the University of Illinois at Chicago, we replicated the finding of an association of APOL1 G1/G2 with proteinuria, specifically with urine albumin concentration (β=1.1, P=0.003), observed an even stronger association with hemoglobinuria (OR=2.5, P=4.3×10(-6)), and also replicated the finding of an association with hemoglobinuria in 487 patients from the Walk-Treatment of Pulmonary Hypertension and Sickle cell Disease with Sildenafil Therapy study (OR=2.6, P=0.003). In 25 University of Illinois sickle cell disease patients, concentrations of urine kidney injury molecule-1 correlated with urine cell-free hemoglobin concentrations (r=0.59, P=0.002). Exposing human proximal tubular cells to increasing cell-free hemoglobin led to increasing concentrations of supernatant kidney injury molecule-1 (P=0.01), reduced viability (P=0.01) and induction of HMOX1 and SOD2. HMOX1 rs743811 associated with chronic kidney disease stage (OR=3.0, P=0.0001) in the University of Illinois cohort and end-stage renal disease (OR=10.0, P=0.0003) in the Walk-Treatment of Pulmonary Hypertension and Sickle cell Disease with Sildenafil Therapy cohort. Longer HMOX1 GT-tandem repeats (>25) were associated with lower estimated glomerular filtration rate in the University of Illinois cohort (P=0.01). Our findings point to an association of APOL1 G1/G2 with kidney disease in sickle cell disease, possibly through increased risk of hemoglobinuria, and associations of HMOX1 variants with kidney disease, possibly through reduced protection of the kidney from hemoglobin-mediated toxicity.
Collapse
Affiliation(s)
- Santosh L Saraf
- Division of Hematology & Oncology, Department of Medicine, Comprehensive Sickle Cell Center, University of Illinois at Chicago, IL
| | - Xu Zhang
- Division of Hematology & Oncology, Department of Medicine, Comprehensive Sickle Cell Center, University of Illinois at Chicago, IL
| | - Binal Shah
- Division of Hematology & Oncology, Department of Medicine, Comprehensive Sickle Cell Center, University of Illinois at Chicago, IL
| | - Tamir Kanias
- Division of Pulmonary, Allergy, and Critical Care Medicine, Vascular Medicine Institute, University of Pittsburgh, PA
| | - Krishnamurthy P Gudehithlu
- Division of Nephrology, Department of Medicine, John H. Stroger, Jr Hospital of Cook County, Chicago, IL
| | - Rick Kittles
- Department of Surgery, University of Arizona, Tucson, AZ
| | - Roberto F Machado
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, IL
| | - Jose A L Arruda
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, IL, USA
| | - Mark T Gladwin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Vascular Medicine Institute, University of Pittsburgh, PA
| | - Ashok K Singh
- Division of Nephrology, Department of Medicine, John H. Stroger, Jr Hospital of Cook County, Chicago, IL
| | - Victor R Gordeuk
- Division of Hematology & Oncology, Department of Medicine, Comprehensive Sickle Cell Center, University of Illinois at Chicago, IL
| |
Collapse
|
32
|
Zhang L, Song FF, Huang YB, Zheng H, Song FJ, 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 2015; 15:4617-22. [PMID: 24969894 DOI: 10.7314/apjcp.2014.15.11.4617] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Several studies have previously focused on associations between the (GT)n repeat polymorphism of the heme oxygenase-1 (HO-1) gene promoter region and risk of cancers, but results are complex. We conducted the present meta-analysis to integrate relevant findings and evaluate the association between HO-1 (GT)n repeat polymorphism and cancer susceptibility. MATERIALS AND METHODS Published literature was retrieved from the PubMed/MEDLINE, EMBASE and ISI Web of Science databases before November 2013. For all alleles and genotypes, odds ratios were pooled to assess the strength of the associations using either fixed-effects or random-effects models according to heterogeneity. Subgroup analysis was conducted according to ethnicity and histopathology. RESULTS A total of 10 studies involving 2,367 cases and 2,870 controls were identified. The results showed there was no association between HO-1 (GT)n repeat polymorphism and the cancer risk both at the allelic and genotypic level. However, in the stratified analysis, we observed an increased risk of squamous cell carcinoma in persons carrying the LL genotype and the LL+LS genotype as compared with those carrying the SS genotype. When the LS and SS genotypes were combined, the odds ratio for squamous cell carcinoma in LL-genotype carriers, were also significantly increased. No publication bias was observed. CONCLUSIONS The LL genotype and L-allele carrying genotypes (LL+LS) of HO-1 (GT)n repeat polymorphism are potential genetic factors for developing squamous cell carcinoma. More large and well-designed studies are required for further validations.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China E-mail :
| | | | | | | | | | | |
Collapse
|
33
|
Ayuso P, Martínez C, Pastor P, Lorenzo-Betancor O, Luengo A, Jiménez-Jiménez FJ, Alonso-Navarro H, Agúndez JAG, García-Martín E. An association study between Heme oxygenase-1 genetic variants and Parkinson's disease. Front Cell Neurosci 2014; 8:298. [PMID: 25309329 PMCID: PMC4173932 DOI: 10.3389/fncel.2014.00298] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/04/2014] [Indexed: 01/25/2023] Open
Abstract
The blood-brain barrier (BBB) supplies brain tissues with nutrients, filters harmful compounds from the brain back to the bloodstream, and plays a key role in iron homeostasis in the human brain. Disruptions of the BBB are associated with several neurodegenerative conditions including Parkinson's disease (PD). Oxidative stress, iron deposition and mitochondrial impaired function are considered as risk factors for degeneration of the central nervous system. Heme oxygenase (HMOX) degrades heme ring to biliverdin, free ferrous iron and carbon monoxide being the rate-limiting activity in heme catabolism. The isoform HMOX1 is highly inducible in response to reactive oxygen species, which induce an increase in BBB permeability and impair its pathophysiology. Consequently, an over- expression of this enzyme may contribute to the marked iron deposition found in PD. We analyzed the HMOX1 SNPs rs2071746, rs2071747, and rs9282702, a microsatellite (GT)n polymorphism and copy number variations in 691 patients suffering from PD and 766 healthy control individuals. Copy number variations in the HMOX1 gene exist, but these do not seem to be associated with PD risk. In contrast two polymorphisms that modify the transcriptional activity of the gene, namely a VNTR (GT)n and the SNP rs2071746, are strongly associated with PD risk, particularly with the classic PD phenotype and with early onset of the disease. This study indicates that HMOX1 gene variants are associated to the risk of developing some forms of PD, thus adding new information that supports association of HMOX gene variations with PD risk.
Collapse
Affiliation(s)
- Pedro Ayuso
- Department of Biochemistry, Molecular Biology, and Genetic, University of Extremadura Cáceres, Spain ; Redes Temáticas de Investigación Cooperativa en Salud (RIRAAF/RETICS), Instituto de Salud Carlos III Madrid, Spain
| | - Carmen Martínez
- Redes Temáticas de Investigación Cooperativa en Salud (RIRAAF/RETICS), Instituto de Salud Carlos III Madrid, Spain ; Department of Pharmacology, University of Extremadura Cáceres, Spain
| | - Pau Pastor
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra Pamplona, Spain ; Department of Neurology, School of Medicine, Clínica Universidad de Navarra, University of Navarra Pamplona, Spain ; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III Madrid, Spain
| | - Oswaldo Lorenzo-Betancor
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research (CIMA), University of Navarra Pamplona, Spain ; Department of Neurology, School of Medicine, Clínica Universidad de Navarra, University of Navarra Pamplona, Spain
| | - Antonio Luengo
- Department of Pharmacology, University of Extremadura Cáceres, Spain
| | | | | | - José A G Agúndez
- Redes Temáticas de Investigación Cooperativa en Salud (RIRAAF/RETICS), Instituto de Salud Carlos III Madrid, Spain ; Department of Pharmacology, University of Extremadura Cáceres, Spain
| | - Elena García-Martín
- Department of Biochemistry, Molecular Biology, and Genetic, University of Extremadura Cáceres, Spain ; Redes Temáticas de Investigación Cooperativa en Salud (RIRAAF/RETICS), Instituto de Salud Carlos III Madrid, Spain
| |
Collapse
|
34
|
Han SW, Song W, Kim HS, Shin KS, Kang H, Cho HC, Ki CS, Park MJ. HMOX1 gene promoter polymorphism is not associated with coronary artery disease in Koreans. Ann Lab Med 2014; 34:337-44. [PMID: 25187885 PMCID: PMC4151001 DOI: 10.3343/alm.2014.34.5.337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 05/21/2014] [Accepted: 07/28/2014] [Indexed: 01/09/2023] Open
Abstract
Background The heme oxygenase-1 gene (HMOX1) promoter polymorphisms modulate its transcription in response to oxidative stress. This study screened for HMOX1 polymorphisms and investigated the association between HMOX1 polymorphisms and coronary artery disease (CAD) in the Korean population. Methods The study population consisted of patients with CAD with obstructive lesions (n=110), CAD with minimal or no lesions (n=40), and controls (n=107). Thirty-nine patients with CAD with obstructive lesions underwent follow-up coronary angiography after six months for the presence of restenosis. The 5'-flanking region containing (GT)n repeats of the HMOX1 gene was analyzed by PCR. Results The numbers of (GT)n repeats in the HMOX1 promoter showed a bimodal distribution. The alleles were divided into two subclasses, S25 and L25, depending on whether there were less than or equal to and more than 25 (GT)n repeats, respectively. The allele and genotype frequencies among groups were statistically not different. More subjects in the S25-carrier group had the low risk levels of high sensitivity C-reactive protein (hsCRP) for the CAD than those in the non-S25 carrier group (P=0.034). Multivariate logistic regression analysis revealed that the genotypes of (GT)n repeats were not related to CAD status. The restenosis group in the coronary angiography follow-up did not show any significant difference in HMOX1 genotype frequency. Conclusions The HMOX1 genotypes were not found to be associated with CAD, but the short allele carrier group contained more individuals with hsCRP values reflecting low risk of cardiovascular disease in the Korean population.
Collapse
Affiliation(s)
- Seong Woo Han
- Department of Cardiology, Hallym University College of Medicine, Seoul, Korea
| | - Wonkeun Song
- Department of Laboratory Medicine, Hallym University College of Medicine, Seoul, Korea
| | - Han-Sung Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Seoul, Korea
| | - Kyu-Sung Shin
- Department of Laboratory Medicine, Hallym University College of Medicine, Seoul, Korea
| | - Heejung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Seoul, Korea
| | - Hyoun Chan Cho
- Department of Laboratory Medicine, Hallym University College of Medicine, Seoul, Korea
| | - Chang-Seok Ki
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min-Jeong Park
- Department of Laboratory Medicine, Hallym University College of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Chen YH, Kuo KL, Hung SC, Hsu CC, Chen YH, Tarng DC. Length polymorphism in heme oxygenase-1 and risk of CKD among patients with coronary artery disease. J Am Soc Nephrol 2014; 25:2669-77. [PMID: 24762402 DOI: 10.1681/asn.2013111205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The length polymorphism of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter is associated with cardiovascular events and mortality in high-risk populations. Experimental data suggest that heme oxygenase-1 protects against kidney disease. However, the association between this polymorphism and long-term risk of CKD in high-risk patients is unknown. We analyzed the allelic frequencies of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter in 386 patients with coronary artery disease recruited from January 1999 to July 2001 and followed until August 31, 2012. The S allele represents short repeats (<27), and the L allele represents long repeats (≥27). The primary renal end points consisted of sustained serum creatinine doubling and/or ESRD requiring long-term RRT. The secondary end points were major adverse cardiovascular events and mortality. At the end of study, the adjusted hazard ratios (95% confidence intervals) for each L allele in the additive model were 1.99 (1.27 to 3.14; P=0.003) for the renal end points, 1.70 (1.27 to 2.27; P<0.001) for major adverse cardiovascular events, and 1.36 (1.04 to 1.79; P=0.03) for mortality. With cardiac events as time-dependent covariates, the adjusted hazard ratio for each L allele in the additive model was 1.91 (1.20 to 3.06; P=0.01) for the renal end points. In conclusion, a greater number of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter is associated with higher risk for CKD, cardiovascular events, and mortality among patients with coronary artery disease.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Faculty of Medicine, Division of Nephrology, Department of Internal Medicine, Taipei City Hospital, Yang-Ming Branch, Taipei, Taiwan
| | - Ko-Lin Kuo
- Division of Nephrology, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taipei, Taiwan
| | - Szu-Chun Hung
- Division of Nephrology, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taipei, Taiwan
| | - Chih-Cheng Hsu
- Division of Geriatrics and Gerontology, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan; and
| | | | - Der-Cherng Tarng
- Faculty of Medicine, Institute of Clinical Medicine, and Department and Institute of Physiology, National Yang-Ming University, Taipei, Taiwan; Nephrology, Department of Medicine and Immunology Center, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
36
|
Chiabrando D, Vinchi F, Fiorito V, Mercurio S, Tolosano E. Heme in pathophysiology: a matter of scavenging, metabolism and trafficking across cell membranes. Front Pharmacol 2014; 5:61. [PMID: 24782769 PMCID: PMC3986552 DOI: 10.3389/fphar.2014.00061] [Citation(s) in RCA: 289] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/18/2014] [Indexed: 01/19/2023] Open
Abstract
Heme (iron-protoporphyrin IX) is an essential co-factor involved in multiple biological processes: oxygen transport and storage, electron transfer, drug and steroid metabolism, signal transduction, and micro RNA processing. However, excess free-heme is highly toxic due to its ability to promote oxidative stress and lipid peroxidation, thus leading to membrane injury and, ultimately, apoptosis. Thus, heme metabolism needs to be finely regulated. Intracellular heme amount is controlled at multiple levels: synthesis, utilization by hemoproteins, degradation and both intracellular and intercellular trafficking. This review focuses on recent findings highlighting the importance of controlling intracellular heme levels to counteract heme-induced oxidative stress. The contributions of heme scavenging from the extracellular environment, heme synthesis and incorporation into hemoproteins, heme catabolism and heme transport in maintaining adequate intracellular heme content are discussed. Particular attention is put on the recently described mechanisms of heme trafficking through the plasma membrane mediated by specific heme importers and exporters. Finally, the involvement of genes orchestrating heme metabolism in several pathological conditions is illustrated and new therapeutic approaches aimed at controlling heme metabolism are discussed.
Collapse
Affiliation(s)
- Deborah Chiabrando
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Francesca Vinchi
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Veronica Fiorito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Sonia Mercurio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| | - Emanuela Tolosano
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin Turin, Italy
| |
Collapse
|
37
|
Lee JP, Kim DH, Yang SH, Hwang JH, An JN, Min SI, Ha J, Oh YK, Kim YS, Lim CS. Serum bilirubin affects graft outcomes through UDP-glucuronosyltransferase sequence variation in kidney transplantation. PLoS One 2014; 9:e93633. [PMID: 24690955 PMCID: PMC3972238 DOI: 10.1371/journal.pone.0093633] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 03/04/2014] [Indexed: 12/20/2022] Open
Abstract
Background Oxidative stress is a major mediator of adverse outcome after kidney transplantation. Bilirubin is produced by heme oxygenase-1 (HO-1), catalyzed by UDP-glucuronosyltransferase (UGT1A1), and has potential as an antioxidant. In this study, we investigated the effects of HO-1 and UGT1A1 sequence variations on kidney allograft outcomes. Methods Clinical data were collected from 429 Korean recipients who underwent kidney transplantation from 1990–2008. Genotyping for UGT1A1*28 and HO-1 (A−413T) was performed. Acute rejection and graft survival were monitored as end-points. Results Serum levels of total bilirubin were significantly increased after transplantation (0.41±0.19 mg/dL to 0.80±0.33 mg/dL, P<0.001). Post-transplant 1-year bilirubin level was higher in 6/7 or 7/7 carriers compared with 6/6 homozygotes in terms of the UGT1A1*28 polymorphism (6/6 vs. 6/7 vs. 7/7: 0.71±0.27 vs. 1.06±0.36 vs. 1.10±0.45 mg/dL, P<0.001). According to an additive model of genotype analysis, the 7-allele genotype had a protective effect on the development of acute rejection compared with the 6-allele (odds ratio 0.43, 95% CI 0.25–0.73, P for trend = 0.006). Multivariate Cox regression analysis revealed that individuals carrying the 7-allele had a decreased risk of graft loss, by a factor of 0.36 (95% CI 0.15–0.85, P = 0.019). The HO-1 (A−413T) polymorphism had no effect on serum bilirubin levels or graft outcomes. Conclusions The UGT1A1*28 polymorphism is associated with changes in serum bilirubin and with graft outcome after kidney transplantation.
Collapse
Affiliation(s)
- Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Do Hyoung Kim
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Korea
| | - Seung Hee Yang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Ho Hwang
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Korea
| | - Jung Nam An
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Il Min
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jongwon Ha
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| |
Collapse
|
38
|
Okano Y, Nezu U, Enokida Y, Lee MTM, Kinoshita H, Lezhava A, Hayashizaki Y, Morita S, Taguri M, Ichikawa Y, Kaneko T, Natsumeda Y, Yokose T, Nakayama H, Miyagi Y, Ishikawa T. SNP (-617C>A) in ARE-like loci of the NRF2 gene: a new biomarker for prognosis of lung adenocarcinoma in Japanese non-smoking women. PLoS One 2013; 8:e73794. [PMID: 24040073 PMCID: PMC3770684 DOI: 10.1371/journal.pone.0073794] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 07/26/2013] [Indexed: 12/30/2022] Open
Abstract
Purpose The transcription factor NRF2 plays a pivotal role in protecting normal cells from external toxic challenges and oxidative stress, whereas it can also endow cancer cells resistance to anticancer drugs. At present little information is available about the genetic polymorphisms of the NRF2 gene and their clinical relevance. We aimed to investigate the single nucleotide polymorphisms in the NRF2 gene as a prognostic biomarker in lung cancer. Experimental Design We prepared genomic DNA samples from 387 Japanese patients with primary lung cancer and detected SNP (c.–617C>A; rs6721961) in the ARE-like loci of the human NRF2 gene by the rapid genetic testing method we developed in this study. We then analyzed the association between the SNP in the NRF2 gene and patients’ overall survival. Results Patients harboring wild-type (WT) homozygous (c.–617C/C), SNP heterozygous (c.–617C/A), and SNP homozygous (c.–617A/A) alleles numbered 216 (55.8%), 147 (38.0%), and 24 (6.2%), respectively. Multivariate logistic regression models revealed that SNP homozygote (c.–617A/A) was significantly related to gender. Its frequency was four-fold higher in female patients than in males (10.8% female vs 2.7% male) and was associated with female non-smokers with adenocarcinoma. Interestingly, lung cancer patients carrying NRF2 SNP homozygous alleles (c.–617A/A) and the 309T (WT) allele in the MDM2 gene exhibited remarkable survival over 1,700 days after surgical operation (log-rank p = 0.021). Conclusion SNP homozygous (c.–617A/A) alleles in the NRF2 gene are associated with female non-smokers with adenocarcinoma and regarded as a prognostic biomarker for assessing overall survival of patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Yasuko Okano
- Omics Science Center, RIKEN Yokohama Institute, Yokohama, Japan
- Department of Clinical Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Uru Nezu
- Omics Science Center, RIKEN Yokohama Institute, Yokohama, Japan
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yasuaki Enokida
- Omics Science Center, RIKEN Yokohama Institute, Yokohama, Japan
- Division of Thoracic and Visceral Organ Surgery, Gunma University Graduate School of Medicine, Maehashi, Japan
| | - Ming Ta Michael Lee
- Laboratory for International Alliance on Genomic Research, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | | | | | - Satoshi Morita
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Masataka Taguri
- Department of Biostatistics and Epidemiology, Yokohama City University Medical Center, Yokohama, Japan
| | - Yasushi Ichikawa
- Department of Clinical Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Kaneko
- Omics Science Center, RIKEN Yokohama Institute, Yokohama, Japan
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Yutaka Natsumeda
- Department of Clinical Research, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Haruhiko Nakayama
- Department of Thoracic Surgery, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yohei Miyagi
- Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Toshihisa Ishikawa
- Omics Science Center, RIKEN Yokohama Institute, Yokohama, Japan
- * E-mail:
| |
Collapse
|
39
|
Chen YH, Hung SC, Tarng DC. Length polymorphism in heme oxygenase-1 and cardiovascular events and mortality in hemodialysis patients. Clin J Am Soc Nephrol 2013; 8:1756-63. [PMID: 23813560 DOI: 10.2215/cjn.01110113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Persistent inflammation and oxidative stress play a pathogenic role in the high cardiovascular morbidity and mortality of hemodialysis patients. Heme oxygenase-1 is considered to have anti-inflammatory and antioxidant properties. This study assessed the association between the length of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene microsatellite promoter and cardiovascular events and mortality among hemodialysis patients. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Study participants were recruited from October 1, 2006 to December 31, 2006. The allelic frequencies of the length of guanosine thymidine dinucleotide repeats (the S allele represents shorter [<27] repeats, and the L allele represents longer [≥ 27] repeats) in the heme oxygenase-1 gene promoter were analyzed in 1080 unrelated chronic hemodialysis patients and 365 healthy controls for distribution comparison. Cardiovascular events and mortality were the study outcomes, and the hemodialysis patients were followed until June 30, 2011. RESULTS The genotype proportions were 20.6%, 48.8%, and 30.6% for S/S, S/L, and L/L, respectively, in the hemodialysis patients and comparable with those proportions in healthy controls. The patients with the L/L genotype had significantly higher baseline serum high-sensitivity C-reactive protein and malondialdehyde levels than the patients with the S/S or S/L genotypes. During a median follow-up of 50 months, 307 patients died. A Kaplan-Meier survival analysis showed the highest cardiovascular events and all-cause mortality in patients with the L/L genotype. The adjusted hazard ratios (95% confidence intervals) for each L allele in additive model were 1.42 (1.20 to 1.67 [P<0.001]) for cardiovascular events and 1.19 (1.01 to 1.40 [P=0.03]) for all-cause mortality. CONCLUSIONS Chronic hemodialysis patients with longer lengths of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter exhibit higher inflammation and oxidative stress. These patients have higher risk of long-term cardiovascular events and mortality.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Department of Medicine, Taipei Veterans General Hospital, Su-Ao Branch, Yilan, Taiwan;, †Faculty of Medicine and, ‖Department and Institute of Physiology, National Yang-Ming University, Taipei, Taiwan;, ‡Division of Nephrology, Buddhist Tzu Chi General Hospital, Taipei Branch, Taipei, Taiwan, §Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | |
Collapse
|
40
|
Repessé Y, Peyron I, Dimitrov JD, Dasgupta S, Moshai EF, Costa C, Borel-Derlon A, Guillet B, D'Oiron R, Aouba A, Rothschild C, Oldenburg J, Pavlova A, Kaveri SV, Lacroix-Desmazes S. Development of inhibitory antibodies to therapeutic factor VIII in severe hemophilia A is associated with microsatellite polymorphisms in the HMOX1 promoter. Haematologica 2013; 98:1650-5. [PMID: 23716558 DOI: 10.3324/haematol.2013.084665] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Induction of heme oxygenase-1, a stress-inducible enzyme with anti-inflammatory activity, reduces the immunogenicity of therapeutic factor VIII in experimental hemophilia A. In humans, heme oxygenase-1 expression is modulated by polymorphisms in the promoter of the heme oxygenase-1-encoding gene (HMOX1). We investigated the relationship between polymorphisms in the HMOX1 promoter and factor VIII inhibitor development in severe hemophilia A. We performed a case-control study on 99 inhibitor-positive patients and 263 patients who did not develop inhibitors within the first 150 cumulative days of exposure to therapeutic factor VIII. Direct sequencing and DNA fragment analysis were used to study (GT)n polymorphism and single nucleotide polymorphisms located at -1135 and -413 in the promoter of HMOX1. We assessed associations between the individual allele frequencies or genotypes, and inhibitor development. Our results demonstrate that inhibitor-positive patients had a higher frequency of alleles with large (GT)n repeats (L: n≥30), which are associated with lesser heme oxygenase-1 expression (odds ratio 2.31; 95% confidence interval 1.46-3.66; P<0.001]. Six genotypes (L/L, L/M, L/S, M/M, M/S and S/S) of (GT)n repeats were identified (S: n<21; M: 21≤n<30). The genotype group including L alleles (L/L, L/M and L/S) was statistically more frequent among inhibitor-positive than inhibitor-negative patients, as compared to the other genotypes (33.3% versus 17.1%) (odds ratio 2.21, 95% confidence interval 1.30-3.76; P<0.01). To our knowledge, this is the first association identified between HMOX1 promoter polymorphism and development of anti-drug antibodies. Our study paves the way towards modulation of the endogenous anti-inflammatory machinery of hemophilia patients to reduce the risk of inhibitor development.
Collapse
|
41
|
Li B, Takeda K, Ishikawa K, Yoshizawa M, Sato M, Shibahara S, Furuyama K. Coordinated expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 and heme oxygenase 2: evidence for a regulatory link between glycolysis and heme catabolism. TOHOKU J EXP MED 2013; 228:27-41. [PMID: 22892400 DOI: 10.1620/tjem.228.27] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Heme is an essential requirement for cell survival. Heme oxygenase (HO) is the rate-limiting enzyme in heme catabolism and consists of two isozymes, HO-1 and HO-2. To identify the protein that regulates the expression or function of HO-1 or HO-2, we searched for proteins that interact with both isozymes, using protein microarrays. We thus identified 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 (PFKFB4) that synthesizes or degrades fructose-2,6-bisphosphate, a key activator of glycolysis, depending on cellular microenvironments. Importantly, HO-2 and PFKFB4 are predominantly expressed in haploid spermatids. Here, we show a drastic reduction in expression levels of PFKFB4 mRNA and protein and HO-2 mRNA in HepG2 human hepatoma cells in responses to glucose deprivation (≤ 2.5 mM), which occurred concurrently with remarkable induction of HO-1 mRNA and protein. Knockdown of HO-2 expression in HepG2 cells, using small interfering RNA, caused PFKFB4 mRNA levels to decrease with a concurrent increase in HO-1 expression. Thus, in HepG2 cells, HO-1 expression was increased, when expression levels of HO-2 and PFKFB4 mRNAs were decreased. Conversely, overexpression of HO-2 in HepG2 cells caused the level of co-expressed PFKFB4 protein to increase. These results suggest a potential regulatory role for HO-2 in ensuring PFKFB4 expression. Moreover, in D407 human retinal pigment epithelial cells, glucose deprivation decreased the expression levels of PFKFB4, HO-1, and HO-2 mRNAs. Thus, glucose deprivation consistently down-regulated the expression of PFKFB4 and HO-2 mRNAs in both HepG2 cells and RPE cells. We therefore postulate that PFKFB4 and HO-2 are expressed in a coordinated manner to maintain glucose homeostasis.
Collapse
Affiliation(s)
- Bin Li
- Department of Molecular Biology and Applied Physiology, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Heme and its breakdown products CO, Fe, and bilirubin are being recognized as signaling molecules or even therapeutic agents, but also exert adverse effects when released at high concentrations. Manipulating the pathway confers protection in rodent sepsis models via both control of free heme and formation of its first and higher-order products. Thus, regulatory elements present in human heme oxygenase 1 (HMOX1) and biliverdin reductases (BLVRA/B) genes might impact outcome. We tested whether a highly polymorphic (GT)n microsatellite and single-nucleotide polymorphisms in HMOX1 and BLVRA/B genes are associated with outcome of sepsis. Two cohorts (n = 430 and 398 patients) with severe sepsis were screened for single-nucleotide polymorphisms and/or the microsatellite by fragment length analysis and genotyping techniques. Heme oxygenase 1 plasma levels were determined in additional patients with severe sepsis (n = 92) by enzyme-linked immunosorbent assay. Based on mean Sepsis-related Organ Failure Assessment scores, patients homozygous for rs2071746 A allele or medium length (GT)n microsatellites of HMOX1 showed higher 28-day mortality (P = 0.047 and P = 0.033) in one cohort compared with other genotypes, whereas 90-day mortality rates showed no association. The T allele was less frequently observed in both cohorts than would be expected according to Hardy-Weinberg equilibrium. Heme oxygenase 1 plasma levels were elevated in septic patients, independent of the genotype. Single-nucleotide polymorphisms within BLVRA/B showed no association with outcome. Short (GT)n repeats that are in linkage disequilibrium with the T allele of rs2071746 in HMOX1 are associated with favorable outcome, whereas no association with gene variants of BLVRA/B, involved in the generation of higher-order metabolites, was noticed.
Collapse
|
43
|
Gregorek AC, Gornik KC, Polancec DS, Dabelic S. GT microsatellite repeats in the heme oxygenase-1 gene promoter associated with abdominal aortic aneurysm in Croatian patients. Biochem Genet 2013; 51:482-92. [PMID: 23430116 DOI: 10.1007/s10528-013-9579-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 10/16/2012] [Indexed: 12/20/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a complex genetic disorder caused by the interplay of genetic and environmental risk factors. The number of (GT)(n) repeats in the heme oxygenase-1 (HO-1) gene promoter modulates transcription of this enzyme, which might have anti-inflammatory, antioxidant, antiapoptotic, and antiproliferative effect. The distribution of alleles and genotypes in Croatian individuals genotyped for the (GT)(n) HO-1 polymorphism was similar to that in other European populations. Frequency of the short (S) alleles (GT < 25) was higher in AAA patients (41.9%) than in non-AAA individuals (28.2%, p = 0.0026) because there were more SL heterozygotes among the AAA patients. The SL genotype appeared to increase the risk for AAA, but the increase was not statistically significant after adjustment for age, sex, smoking, hypertension, and hyperlipidemia (OR = 1.53, 95% CI 0.90-3.09, p = 0.062). These findings contradict those of the only other study performed so far on the association of (GT)(n) HO-1 polymorphism and AAA.
Collapse
Affiliation(s)
- Andrea Crkvenac Gregorek
- Division of Vascular Surgery, Clinic of Surgery, University Hospital Center Zagreb, Zagreb, Croatia
| | | | | | | |
Collapse
|
44
|
Vázquez-Armenta G, González-Leal N, J Vázquez-de la Torre M, Francisco Muñoz-Valle J, E Ramos-Márquez M, Hernández-Cañaveral I, Plascencia-Hernández A, Siller-López F. Short (GT)n Microsatellite Repeats in the Heme Oxygenase-1 Gene Promoter Are Associated with Antioxidant and Anti-Inflammatory Status in Mexican Pediatric Patients with Sepsis. TOHOKU J EXP MED 2013; 231:201-9. [DOI: 10.1620/tjem.231.201] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Gabriela Vázquez-Armenta
- Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| | - Natalia González-Leal
- Servicio Infectología Pediátrica, Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Universidad de Guadalajara
| | - Mayra J Vázquez-de la Torre
- Servicio Infectología Pediátrica, Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Universidad de Guadalajara
| | - José Francisco Muñoz-Valle
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| | - Martha E Ramos-Márquez
- Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| | - Iván Hernández-Cañaveral
- Departamento de Microbiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| | - Arturo Plascencia-Hernández
- Servicio Infectología Pediátrica, Hospital Civil de Guadalajara “Fray Antonio Alcalde”, Universidad de Guadalajara
| | - Fernando Siller-López
- Facultad de Medicina, Fundación Universitaria Autónoma de las Américas
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| |
Collapse
|
45
|
The host genetic diversity in malaria infection. J Trop Med 2012; 2012:940616. [PMID: 23316245 PMCID: PMC3532872 DOI: 10.1155/2012/940616] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/06/2012] [Accepted: 11/19/2012] [Indexed: 02/06/2023] Open
Abstract
Populations exposed to Plasmodium infection develop genetic mechanisms of protection against severe disease. The clinical manifestation of malaria results primarily from the lysis of infected erythrocytes and subsequent immune and inflammatory responses. Herein, we review the genetic alterations associated with erythrocytes or mediators of the immune system, which might influence malaria outcome. Moreover, polymorphisms in genes related to molecules involved in mechanisms of cytoadherence and their influence on malaria pathology are also discussed. The results of some studies have suggested that the combinatorial effects of a set of genetic factors in the erythrocyte-immunology pathway might be relevant to host resistance or susceptibility against Plasmodium infection. However, these results must be interpreted with caution because of the differences observed in the functionality and frequency of polymorphisms within different populations. With the recent advances in molecular biology techniques, more robust studies with reliable data have been reported, and the results of these studies have identified individual genetic factors for consideration in preventing severe disease and the individual response to treatment.
Collapse
|
46
|
Schipper HM. Biomarker potential of heme oxygenase-1 in Alzheimer's disease and mild cognitive impairment. Biomark Med 2012; 1:375-85. [PMID: 20477381 DOI: 10.2217/17520363.1.3.375] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The advent of an accessible chemical biological marker that differentiates early, sporadic Alzheimer's disease (AD) from normal aging and other dementing illnesses, and identifies individuals with mild cognitive impairment who are destined to deteriorate to Alzheimer's dementia, would represent a major achievement in the evaluation and management of this common neurodegenerative disorder. Although several candidate biomarkers of sporadic AD have been identified and commercialized, none currently fulfill the criteria for an ideal test. In this article, we review evidence implicating blood heme oxygenase-1 mRNA/protein levels and a recently identified plasma heme oxygenase-1 suppressor factor as potential biomarkers of AD and mild cognitive impairment.
Collapse
Affiliation(s)
- Hyman M Schipper
- McGill University, Centre for Neurotranslational Research, Lady Davis Institute for Medical Research, Sir Mortimer B Davis Jewish General Hospital, 3755 Cote St Catherine Road, Montreal, Quebec H3T 1E2, Canada.
| |
Collapse
|
47
|
Microsatellite polymorphism in the heme oxygenase-1 gene promoter and the risk of psoriasis in Taiwanese. Arch Dermatol Res 2012; 304:739-44. [PMID: 22965812 DOI: 10.1007/s00403-012-1289-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 08/02/2012] [Accepted: 08/29/2012] [Indexed: 10/27/2022]
Abstract
Psoriasis is a chronic disease characterized by inflammation of the skin. The expression of heme oxygenase-1 (HO-1), the rate-limiting enzyme involved in heme degradation, correlates well with the severity of psoriasis, and is a heritable trait. This study aimed to assess the role of (GT)(n) dinucleotide repeat polymorphisms in the promoter region of the HO-1 gene in Chinese-Taiwanese patients with psoriasis. In total, 288 patients with psoriasis and 542 control subjects were analyzed for the presence of the HO-1 microsatellite polymorphism by using polymerase chain reaction. The alleles were classified as the S and L alleles according to the number of (GT)(n) repeats, with the alleles with ≤26 repeats designated as S and alleles with ≥27 repeats designated as L alleles. The subjects were then classified as having S/S, S/L, or L/L genotypes according to each of their HO-1 alleles. No significant difference was observed in either the genotype or allele distribution between the patients and healthy controls. However, the average number of repeats of both alleles in psoriasis patients with late disease onset was lower than that of psoriasis patients with early disease onset (26.7 ± 3.2 vs. 27.5 ± 3.4; P = 0.043, adjusted for age and sex), but the difference was not significant after additional adjustment for body mass index, smoking, diabetes, and hypertension (P = 0.189). Our results suggest that the HO-1 microsatellite polymorphism may not contribute to the genetic background of psoriasis in Chinese-Taiwanese patients.
Collapse
|
48
|
Choi SW, Yeung VTF, Benzie IFF. Heme oxygenase microsatellite polymorphism, oxidative stress, glycemic control, and complication development in type 2 diabetes patients. Free Radic Biol Med 2012; 53:60-3. [PMID: 22583702 DOI: 10.1016/j.freeradbiomed.2012.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 04/11/2012] [Accepted: 04/13/2012] [Indexed: 11/15/2022]
Abstract
Heme oxygenase-1 (HMOX-1) is activated by oxidative stress, and gene responsiveness is reportedly determined by the number of dinucleotide (GT(n)) repeats in its highly polymorphic promoter region. "Short" (S; GT(n)<25) alleles reportedly associate with higher response, lower oxidative stress, lower risk of type 2 diabetes mellitus (type 2DM), and better glycemic control and outcome, but data are conflicting. We investigated GT(n) in type 2DM subjects (all ethnic Chinese) in relation to basal glycemic control, oxidative stress, and outcome during up to 9 years' follow-up. Fasting blood from 418 type 2 DM subjects was collected at entry for GT(n) genotyping, glycated hemoglobin, glucose, lipids, and biomarkers of oxidative stress and antioxidants. A subset (n=368) was followed for up to 9 years for incident complications or death. GT(n) genotype distribution was 128, 182, and 108 for, respectively, S/S, S/L, and L/L. No significant differences in glycemic control, lipids, or oxidative stress were seen across genotypes. During follow-up, 168/368 subjects developed complications. No association was seen with GT(n). No difference in plasma HO-1 was seen between genotypes in a small substudy (S/S n=21 vs L/L n=23). Glycated hemoglobin and lymphocytic DNA damage was higher (p<0.05) at entry in the incident complications group. No other significant differences were seen in oxidative stress or antioxidants. Data do not support the postulated link between HMOX-1 microsatellite polymorphism and type 2 DM or the putative beneficial effect of the S allele on glycemic control, oxidative stress, or outcome in type 2 DM patients, at least in this particular population.
Collapse
Affiliation(s)
- Siu-Wai Choi
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | | | | |
Collapse
|
49
|
Weis S, Jesinghaus M, Kovacs P, Schleinitz D, Schober R, Ruffert C, Herms M, Wittenburg H, Stumvoll M, Blüher M, Grützmann R, Schulz HU, Keim V, Mössner J, Bugert P, Witt H, Drenth JPH, Krohn K, Rosendahl J. Genetic analyses of heme oxygenase 1 (HMOX1) in different forms of pancreatitis. PLoS One 2012; 7:e37981. [PMID: 22666428 PMCID: PMC3364204 DOI: 10.1371/journal.pone.0037981] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/01/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Heme oxygenase 1 (HMOX1) is the rate limiting enzyme in heme degradation and a key regulator of inflammatory processes. In animal models the course of pancreatitis was ameliorated by up-regulation of HMOX1 expression. Additionally, carbon monoxide released during heme breakdown inhibited proliferation of pancreatic stellate cells and might thereby prevent the development of chronic pancreatitis (CP). Transcription of HMOX1 in humans is influenced by a GT-repeat located in the promoter. As such, HMOX1 variants might be of importance in the pathogenesis of pancreatitis. METHODS The GT-repeat and SNP rs2071746 were investigated with fluorescence labelled primers and by melting curve analysis in 285 patients with acute pancreatitis, 208 patients with alcoholic CP, 207 patients with idiopathic/hereditary CP, 147 patients with alcoholic liver cirrhosis, and in 289 controls, respectively. GT-repeat analysis was extended to a total of 446 alcoholic CP patients. In addition, we performed DNA sequencing in 145 patients with alcoholic CP, 138 patients with idiopathic/hereditary CP, 147 patients with alcoholic liver cirrhosis, and 151 controls. Exon 3 screening was extended to additional patients and controls. RESULTS S- and L-alleles of the GT-repeat, genotypes and alleles of SNP rs2071746 and non-synonymous variants detected by sequencing were found with similar frequencies in all groups. CONCLUSIONS Although functional data implicate a potential influence of HMOX1 variants on the pathogenesis of pancreatitis, we did not find any association. As rare non-synonymous HMOX1 variants were found in patients and controls, it is rather unlikely that they will have functional consequences essential for pancreatitis development.
Collapse
Affiliation(s)
- Sebastian Weis
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Moritz Jesinghaus
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Department of Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Leipzig, Germany
| | - Dorit Schleinitz
- Department of Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Leipzig, Germany
| | - Robert Schober
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Claudia Ruffert
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Max Herms
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Henning Wittenburg
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Department of Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Internal Medicine, Neurology and Dermatology, Division of Endocrinology, University of Leipzig, Leipzig, Germany
- IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Robert Grützmann
- Department of General, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hans-Ulrich Schulz
- Department of Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Volker Keim
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Joachim Mössner
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg-Hessen, Mannheim, Germany
| | - Heiko Witt
- Department of Pediatrics, Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ) & Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL); Technische Universität München, Munich, Germany
| | - Joost P. H. Drenth
- Department of Gastroenterology and Hepatology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Knut Krohn
- Interdisciplinary Centre for Clinical Research Leipzig, Core-Unit DNA Technologies, University of Leipzig, Leipzig, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
50
|
Chen M, Zhou L, Ding H, Huang S, He M, Zhang X, Cheng L, Wang D, Hu FB, Wu T. Short (GT) ( n ) repeats in heme oxygenase-1 gene promoter are associated with lower risk of coronary heart disease in subjects with high levels of oxidative stress. Cell Stress Chaperones 2012; 17:329-38. [PMID: 22120665 PMCID: PMC3312960 DOI: 10.1007/s12192-011-0309-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 10/20/2011] [Accepted: 10/21/2011] [Indexed: 01/30/2023] Open
Abstract
Although (GT) ( n ) repeats in heme oxygenase-1 (HO-1) promoter may modulate gene transcriptional activity, the association between (GT) ( n ) repeats polymorphism and risk of coronary heart disease (CHD) from different levels of oxidative stress (OS) is unknown. We determined the allelic frequencies of (GT) ( n ) repeats in the HO-1 gene promoter and plasma malonaldehyde (MDA) as biomarkers of OS in 2,298 pairs of CHD patients and controls in the Chinese population. Furthermore, we measured MDA in culture mediums and HO-1 expressions levels in cell lysates of endothelial cells carrying various (GT) ( n ) genotypes under different concentrations of H(2)O(2). Compared with L/L genotype (>25 repeats) carriers, the adjusted odd ratios for S/S genotype (≤25 repeats) in subjects with different levels of OS (MDA < 1.83, 1.83-2.91, >2.91 μmol/L) were 1.06 (95%CI, 0.75 to 1.49), 0.79 (95%CI, 0.55 to 1.12), and 0.60 (95%CI, 0.44 to 0.81), respectively (P (interaction) = 0.002). In biological experiments, compared with endothelial cells carrying L/L genotype, cells with S/S genotype did not have a significantly higher HO-1 expression under 0 μmol/L H(2)O(2), but displayed a significantly higher HO-1 expression under 50 μmol/L H(2)O(2) (P (interaction) = 0.003). S/S genotype in HO-1 gene promoter is associated with a lower risk of CHD in subjects with higher levels of OS, because under conditions of high OS, the S/S genotype has higher levels of HO-1, an antioxidant.
Collapse
Affiliation(s)
- Mu Chen
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Li Zhou
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Hu Ding
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030 China
| | - Suli Huang
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Meian He
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Xiaomin Zhang
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| | - Longxian Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 32 Xinhua Rd, Wuhan, Hubei 430030 China
| | - Daowen Wang
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030 China
| | - Frank B. Hu
- Departments of Nutrition and Epidemiology, Harvard School of Public Health, Boston, MA 02115 USA
| | - Tangchun Wu
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd, Wuhan, Hubei 430030 China
| |
Collapse
|