1
|
Mancuso C. The Heme Oxygenase/Biliverdin Reductase System and Its Genetic Variants in Physiology and Diseases. Antioxidants (Basel) 2025; 14:187. [PMID: 40002374 PMCID: PMC11852105 DOI: 10.3390/antiox14020187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Heme oxygenase (HO) metabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin-IXα (BV), the latter being reduced into bilirubin-IXα (BR) by the biliverdin reductase-A (BVR). Heme oxygenase exists as two isoforms, HO-1, inducible and involved in the cell stress response, and HO-2, constitutive and committed to the physiologic turnover of heme and in the intracellular oxygen sensing. Many studies have identified genetic variants of the HO/BVR system and suggested their connection in free radical-induced diseases. The most common genetic variants include (GT)n dinucleotide length polymorphisms and single nucleotide polymorphisms. Gain-of-function mutations in the HO-1 and HO-2 genes foster the ventilator response to hypoxia and reduce the risk of coronary heart disease and age-related macular degeneration but increase the risk of neonatal jaundice, sickle cell disease, and Parkinson's disease. Conversely, loss-of-function mutations in the HO-1 gene increase the risk of type 2 diabetes mellitus, chronic obstructive pulmonary disease, and some types of cancers. Regarding BVR, the reported loss-of-function mutations increase the risk of green jaundice. Unfortunately, the physiological role of the HO/BVR system does not allow for the hypothesis gene silencing/induction strategies, but knowledge of these mutations can certainly facilitate a medical approach that enables early diagnoses and tailored treatments.
Collapse
Affiliation(s)
- Cesare Mancuso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito, 1, 00168 Rome, Italy;
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Largo F. Vito, 1, 00168 Rome, Italy
| |
Collapse
|
2
|
Nakajima K, Nishizawa H, Chen G, Tsuge S, Yamanaka M, Kiyohara M, Irikura R, Matsumoto M, Tanaka K, Narikawa R, Igarashi K. Intracellular biliverdin dynamics during ferroptosis. J Biochem 2024; 176:472-483. [PMID: 39340324 PMCID: PMC11638335 DOI: 10.1093/jb/mvae067] [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: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Ferroptosis is a cell death mechanism mediated by iron-dependent lipid peroxidation. Although ferroptosis has garnered attention as a cancer-suppressing mechanism, there are still limited markers available for identifying ferroptotic cells or assessing their sensitivity to ferroptosis. The study focused on biliverdin, an endogenous reducing substance in cells, and examined the dynamics of intracellular biliverdin during ferroptosis using a biliverdin-binding cyanobacteriochrome. It was found that intracellular biliverdin decreases during ferroptosis and that this decrease is specific to ferroptosis amongst different forms of cell death. Furthermore, the feasibility of predicting sensitivity to ferroptosis by measuring intracellular biliverdin was demonstrated using a ferroptosis model induced by the re-expression of the transcription factor BACH1. These findings provide further insight into ferroptosis research and are expected to contribute to the development of cancer therapies that exploit ferroptosis.
Collapse
Affiliation(s)
- Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Guan Chen
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Shunichi Tsuge
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Gladstone Institute of Neurological Disease, Gladstone Institutes, 1650 Owens street, San Francisco, CA 94158, USA
| | - Machi Kiyohara
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Rei Narikawa
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Ohsawa, Hachioji, Tokyo 192-0397, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
3
|
Satarug S. Is Environmental Cadmium Exposure Causally Related to Diabetes and Obesity? Cells 2023; 13:83. [PMID: 38201287 PMCID: PMC10778334 DOI: 10.3390/cells13010083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Cadmium (Cd) is a pervasive toxic metal, present in most food types, cigarette smoke, and air. Most cells in the body will assimilate Cd, as its charge and ionic radius are similar to the essential metals, iron, zinc, and calcium (Fe, Zn, and Ca). Cd preferentially accumulates in the proximal tubular epithelium of the kidney, and is excreted in urine when these cells die. Thus, excretion of Cd reflects renal accumulation (body burden) and the current toxicity of Cd. The kidney is the only organ other than liver that produces and releases glucose into the circulation. Also, the kidney is responsible for filtration and the re-absorption of glucose. Cd is the least recognized diabetogenic substance although research performed in the 1980s demonstrated the diabetogenic effects of chronic oral Cd administration in neonatal rats. Approximately 10% of the global population are now living with diabetes and over 80% of these are overweight or obese. This association has fueled an intense search for any exogenous chemicals and lifestyle factors that could induce excessive weight gain. However, whilst epidemiological studies have clearly linked diabetes to Cd exposure, this appears to be independent of adiposity. This review highlights Cd exposure sources and levels associated with diabetes type 2 and the mechanisms by which Cd disrupts glucose metabolism. Special emphasis is on roles of the liver and kidney, and cellular stress responses and defenses, involving heme oxygenase-1 and -2 (HO-1 and HO-2). From heme degradation, both HO-1 and HO-2 release Fe, carbon monoxide, and a precursor substrate for producing a potent antioxidant, bilirubin. HO-2 appears to have also anti-diabetic and anti-obese actions. In old age, HO-2 deficient mice display a symptomatic spectrum of human diabetes, including hyperglycemia, insulin resistance, increased fat deposition, and hypertension.
Collapse
Affiliation(s)
- Soisungwan Satarug
- Kidney Disease Research Collaborative, Translational Research Institute, Woolloongabba, Brisbane, QLD 4102, Australia
| |
Collapse
|
4
|
Mitigation of Cadmium Toxicity through Modulation of the Frontline Cellular Stress Response. STRESSES 2022. [DOI: 10.3390/stresses2030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cadmium (Cd) is an environmental toxicant of public health significance worldwide. Diet is the main Cd exposure source in the non-occupationally exposed and non-smoking populations. Metal transporters for iron (Fe), zinc (Zn), calcium (Ca), and manganese (Mn) are involved in the assimilation and distribution of Cd to cells throughout the body. Due to an extremely slow elimination rate, most Cd is retained by cells, where it exerts toxicity through its interaction with sulfur-containing ligands, notably the thiol (-SH) functional group of cysteine, glutathione, and many Zn-dependent enzymes and transcription factors. The simultaneous induction of heme oxygenase-1 and the metal-binding protein metallothionein by Cd adversely affected the cellular redox state and caused the dysregulation of Fe, Zn, and copper. Experimental data indicate that Cd causes mitochondrial dysfunction via disrupting the metal homeostasis of this organelle. The present review focuses on the adverse metabolic outcomes of chronic exposure to low-dose Cd. Current epidemiologic data indicate that chronic exposure to Cd raises the risk of type 2 diabetes by several mechanisms, such as increased oxidative stress, inflammation, adipose tissue dysfunction, increased insulin resistance, and dysregulated cellular intermediary metabolism. The cellular stress response mechanisms involving the catabolism of heme, mediated by heme oxygenase-1 and -2 (HO-1 and HO-2), may mitigate the cytotoxicity of Cd. The products of their physiologic heme degradation, bilirubin and carbon monoxide, have antioxidative, anti-inflammatory, and anti-apoptotic properties.
Collapse
|
5
|
Mancuso C. The brain heme oxygenase/biliverdin reductase system as a target in drug research and development. Expert Opin Ther Targets 2022; 26:361-374. [PMID: 35285395 DOI: 10.1080/14728222.2022.2052848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The heme oxygenase/biliverdin reductase (HO/BVR) system is involved in heme metabolism. The inducible isoform of HO (HO-1) and BVR both exert cytoprotective effects by enhancing cell stress response. In this context, some xenobiotics, which target HO-1, including herbal products, behave as neuroprotectants in several experimental models of neurodegeneration. Despite this, no drug having either HO-1 or BVR as a main target is currently available. AREAS COVERED After a description of the brain HO/BVR system, the paper analyzes the main classes of drugs acting on the nervous system, with HO as second-level target, and their neuroprotective potential. Finally, the difficulties that exist for the development of drugs acting on HO/BVR and the possible ways to overcome these hurdles are examined. EXPERT OPINION Although the limited clinical evidence has restricted the translational research on the HO/BVR system, mainly because of the dual nature of its by-products, there has been growing interest in the therapeutic potential of these enzymes. Scientists should boost the translational research on the HO/BVR system which could be supported by the significant evidence provided by preclinical studies.
Collapse
Affiliation(s)
- Cesare Mancuso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
6
|
Ma LL, Sun L, Wang YX, Sun BH, Li YF, Jin YL. Association between HO‑1 gene promoter polymorphisms and diseases (Review). Mol Med Rep 2021; 25:29. [PMID: 34841438 PMCID: PMC8669660 DOI: 10.3892/mmr.2021.12545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/11/2021] [Indexed: 12/21/2022] Open
Abstract
Heme oxygenase‑1 (HO‑1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. These degradation products serve an important role in the regulation of inflammation, oxidative stress and apoptosis. While the expression level of HO‑1 is typically low in most cells, it may be highly expressed when induced by a variety of stimulating factors, a process that contributes to the regulation of cell homeostasis. In the 5'‑non‑coding region of the HO‑1 gene, there are two polymorphic sites, namely the (GT)n dinucleotide and T(‑413)A single nucleotide polymorphism sites, which regulate the transcriptional activity of HO‑1. These polymorphisms have been shown to be closely associated with the occurrence and progression of numerous diseases, including cardiovascular, pulmonary, liver and kidney, various types of cancer and viral diseases. The present article reviews the progress that has been made in research on the association between the two types of polymorphisms and these diseases, which is expected to provide novel strategies for the diagnosis, treatment and prognosis of various diseases.
Collapse
Affiliation(s)
- Lin-Lin Ma
- School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai 201318, P.R. China
| | - Lei Sun
- School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai 201318, P.R. China
| | - Yu-Xi Wang
- School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai 201318, P.R. China
| | - Bai-He Sun
- School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai 201318, P.R. China
| | - Yan-Fei Li
- School of Medical Technology, Shanghai University of Medicine & Health Sciences, Shanghai 201318, P.R. China
| | - Yue-Ling Jin
- Management Department of Scientific Research, Shanghai Science and Technology Museum, Shanghai 200127, P.R. China
| |
Collapse
|
7
|
Wu J, Li S, Li C, Cui L, Ma J, Hui Y. The non-canonical effects of heme oxygenase-1, a classical fighter against oxidative stress. Redox Biol 2021; 47:102170. [PMID: 34688156 PMCID: PMC8577501 DOI: 10.1016/j.redox.2021.102170] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 08/29/2021] [Indexed: 12/30/2022] Open
Abstract
The role of heme oxygenase-1 in resisting oxidative stress and cell protection has always been a hot research topic. With the continuous deepening of research, in addition to directly regulating redox by catalyzing the degradation of heme, HO-1 protein also participates in the gene expression level in a great diversity of methods, thereby initiating cell defense. Particularly the non-canonical nuclear-localized HO-1 and HO-1 protein interactions play the role of a warrior against oxidative stress. Besides, HO-1 may be a promising marker for disease prediction and detection in many clinical trials. Especially for malignant diseases, there may be new advances in the treatment of HO-1 by regulating abnormal ROS and metabolic signaling. The purpose of this review is to systematically sort out and describe several aspects of research to facilitate further detailed mechanism research and clinical application promotion in the future. The different subcellular localizations ofHO-1 implies that it has special functions. Nuclear HO-1 plays an indispensable role in gene regulation and other aspects. The interactions between HO-1 and others provide the possibility to participate in vital physiological processes. HO-1 may become a potential disease assessment marker.
Collapse
Affiliation(s)
- Jiajia Wu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Siyu Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Cheng Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Liying Cui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiajia Ma
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China; Basic Medical Institute of Heilongjiang Medical Science Academy, PR China; Translational Medicine Center of Northern China, PR China.
| |
Collapse
|
8
|
Distinct Regulations of HO-1 Gene Expression for Stress Response and Substrate Induction. Mol Cell Biol 2021; 41:e0023621. [PMID: 34398680 DOI: 10.1128/mcb.00236-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is the key enzyme for heme catabolism and cytoprotection. Whereas HO-1 gene expression in response to various stresses has been investigated extensively, the precise mechanisms by which HO-1 gene expression is regulated by the HO-1 substrate heme remain elusive. To systematically examine whether stress-mediated induction and substrate-mediated induction of HO-1 utilize similar or distinct regulatory pathways, we developed an HO-1-DsRed-knock-in reporter mouse in which the HO-1 gene is floxed by loxP sites and the DsRed gene has been inserted. Myeloid lineage-specific recombination of the floxed locus led to fluorescence derived from expression of the HO-1-DsRed fusion protein in peritoneal macrophages. We also challenged general recombination of the locus and generated mice harboring heterozygous recombinant alleles, which enabled us to monitor HO-1-DsRed expression in the whole body in vivo and ex vivo. HO-1 inducers upregulated HO-1-DsRed expression in myeloid lineage cells isolated from the mice. Notably, analyses of peritoneal macrophages from HO-1-DsRed mice lacking NRF2, a major regulator of the oxidative/electrophilic stress response, led us to identify NRF2-dependent stress response-mediated HO-1 induction and NRF2-independent substrate-mediated HO-1 induction. Thus, the HO-1 gene is subjected to at least two distinct levels of regulation, and the available lines of evidence suggest that substrate induction in peritoneal macrophages is independent of CNC family-based regulation.
Collapse
|
9
|
Zhao X, Wang Y, Liu C, Zhou P, Sheng Z, Li J, Zhou J, Chen R, Chen Y, Zhao H, Yan H. Prognostic Value of Total Bilirubin in Patients With ST-Segment Elevation Acute Myocardial Infarction Undergoing Primary Coronary Intervention. Front Cardiovasc Med 2021; 7:615254. [PMID: 33392275 PMCID: PMC7773653 DOI: 10.3389/fcvm.2020.615254] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/24/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Bilirubin, a natural product of heme catabolism, has antioxidant and anti-inflammatory activities and is inversely associated with stable coronary artery disease. However, the relationship between the bilirubin levels and long-term outcomes in patients with ST-segment elevation myocardial infarction (STEMI) who underwent primary percutaneous coronary intervention (PPCI) remains unknown. This study aimed to establish a score model based on bilirubin for predicting major adverse cardiovascular events (MACEs) and stratify patients to the level of care. Methods and Results: Data of 4,151 consecutive patients with STEMI who underwent PPCI were evaluated, and 3,708 cases were analyzed. The total bilirubin (TBil) levels were measured during admission, and the study population was divided into two groups. The high TBil group (n = 143) comprised patients who had a TBil level of ≥22 μmmol/L, and the low TBil group (n = 3,565) comprised patients who had a TBil level of <22 μmmol/L. The median follow-up period was 754 days (2.066 years). The MACE was significantly lower in the high TBil group than in the low TBil group (3.5% vs. 11.0%, p = 0.001). In the multivariate Cox regression analysis, a significant association was noted between the TBil levels and adjusted risk of MACE (hazard ratio, 0.279; 95% confidence interval, 0.088-0.877; p = 0.029). A prediction score model composed of TBil, age, hypertension history, and other eight variables was developed, with scores ranging from 0 to 500. The scores categorized patients into low-, medium-, and high-risk categories. The cumulative survival rate was significantly higher in the low-risk group than in the medium- and high-risk groups for MACE, all-cause death, cardiac death, recurrent myocardial infarction, and ischemic stroke (p < 0.001, p < 0.001, p < 0.001, p = 0.030, and p = 0.001, respectively). The area under the curve of the TBil score was 0.768; this was significantly greater in the pairwise comparison with the Global Registry of Acute Coronary Events score (p = 0.0012). Conclusion: The new prediction score model based on TBil could be used in clinical practice to support risk stratification as recommended in the clinical guidelines.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Ying Wang
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Chen Liu
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Peng Zhou
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Zhaoxue Sheng
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Jiannan Li
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Jinying Zhou
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Runzhen Chen
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Yi Chen
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Hanjun Zhao
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China
| | - Hongbing Yan
- Department of Cardiology, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Fuwai Hospital, Beijing, China.,Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
| |
Collapse
|
10
|
Funes SC, Rios M, Fernández-Fierro A, Covián C, Bueno SM, Riedel CA, Mackern-Oberti JP, Kalergis AM. Naturally Derived Heme-Oxygenase 1 Inducers and Their Therapeutic Application to Immune-Mediated Diseases. Front Immunol 2020; 11:1467. [PMID: 32849503 PMCID: PMC7396584 DOI: 10.3389/fimmu.2020.01467] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
Heme oxygenase (HO) is the primary antioxidant enzyme involved in heme group degradation. A variety of stimuli triggers the expression of the inducible HO-1 isoform, which is modulated by its substrate and cellular stressors. A major anti-inflammatory role has been assigned to the HO-1 activity. Therefore, in recent years HO-1 induction has been employed as an approach to treating several disorders displaying some immune alterations components, such as exacerbated inflammation or self-reactivity. Many natural compounds have shown to be effective inductors of HO-1 without cytotoxic effects; among them, most are chemicals present in plants used as food, flavoring, and medicine. Here we discuss some naturally derived compounds involved in HO-1 induction, their impact in the immune response modulation, and the beneficial effect in diverse autoimmune disorders. We conclude that the use of some compounds from natural sources able to induce HO-1 is an attractive lifestyle toward promoting human health. This review opens a new outlook on the investigation of naturally derived HO-1 inducers, mainly concerning autoimmunity.
Collapse
Affiliation(s)
- Samanta C Funes
- Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Rios
- Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ayleen Fernández-Fierro
- Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Covián
- Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas, Millenium Institute on Immunolgy and Immunotherapy, Facultad Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Juan Pablo Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo, IMBECU CCT Mendoza- CONICET, Mendoza, Argentina.,Facultad de Ciencias Médicas, Instituto de Fisiología, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Millenium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
11
|
After haemin treatment intracellular non-haem iron increases prior to haem oxygenase-1 induction: A study in human monocytic cell line THP-1. Transfus Apher Sci 2019; 58:102662. [PMID: 31727545 DOI: 10.1016/j.transci.2019.10.004] [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: 08/27/2019] [Revised: 10/09/2019] [Accepted: 10/15/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Iron overload is a major health concern for transfusion-dependent patients. Repeated transfusions result in the loading of large amounts of haem-derived iron on macrophages, in turn, inducing cell death. We previously demonstrated that haemin-induced cell death in human monocytic THP-1 cells is consistent with ferroptosis, an iron-dependent cell death regulation mechanism. However, direct measurement of iron after haemin treatment has not yet been conducted. In this study, we measured intracellular non-haem iron concentration and haem oxygenase levels after haemin treatment. MATERIAL AND METHODS Human monocytic THP-1 cells were treated with haemin, and the cell lysate was prepared. Non-haem iron concentration of the cell lysate was measured using the Nitroso-PSAP method. Expression of haem oxygenase-1 (HO-1) and haem oxygenase-2 (HO-2) was quantified by western blotting. RESULTS We measured intracellular non-haem iron and the expression of haem oxygenases post-haemin treatment. Concentration of non-haem iron post-haemin treatment increased dependently with time and dose. HO-1 expression was detected 4 h after haemin treatment, whereas HO-2 expression was constitutive. DISCUSSION Increase in non-haem iron prior to induction of HO-1 expression suggests the involvement of HO-2 in haem-induced cytotoxicity. (184 words).
Collapse
|
12
|
Mhillaj E, Cuomo V, Trabace L, Mancuso C. The Heme Oxygenase/Biliverdin Reductase System as Effector of the Neuroprotective Outcomes of Herb-Based Nutritional Supplements. Front Pharmacol 2019; 10:1298. [PMID: 31780933 PMCID: PMC6859463 DOI: 10.3389/fphar.2019.01298] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/10/2019] [Indexed: 12/28/2022] Open
Abstract
Over the last few years, several preclinical studies have shown that some herbal products, such as ferulic acid, Ginkgo biloba, and resveratrol, exert neuroprotective effects through the modulation of the heme oxygenase/biliverdin reductase system. Unfortunately, sufficient data supporting the shift of knowledge from preclinical studies to humans, particularly in neurodegenerative diseases, are not yet available in the literature. The purpose of this review is to summarize the studies and the main results achieved on the potential therapeutic role of the interaction between the heme oxygenase/biliverdin reductase system with ferulic acid, G. biloba, and resveratrol. Some critical issues have also been reported, mainly concerning the safety profile and the toxicological sequelae associated to the supplementation with the herbs mentioned above, based on both current literature and specific reports issued by the competent Regulatory Authorities.
Collapse
Affiliation(s)
- Emanuela Mhillaj
- Institute of Pharmacology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Vincenzo Cuomo
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome, Rome, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Cesare Mancuso
- Institute of Pharmacology, Università Cattolica del Sacro Cuore, Roma, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| |
Collapse
|
13
|
Concetta Scuto M, Mancuso C, Tomasello B, Laura Ontario M, Cavallaro A, Frasca F, Maiolino L, Trovato Salinaro A, Calabrese EJ, Calabrese V. Curcumin, Hormesis and the Nervous System. Nutrients 2019; 11:2417. [PMID: 31658697 PMCID: PMC6835324 DOI: 10.3390/nu11102417] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
Curcumin is a polyphenol compound extracted from the rhizome of Curcuma longa Linn (family Zingiberaceae) commonly used as a spice to color and flavor food. Several preclinical studies have suggested beneficial roles for curcumin as an adjuvant therapy in free radical-based diseases, mainly neurodegenerative disorders. Indeed, curcumin belongs to the family of hormetins and the enhancement of the cell stress response, mainly the heme oxygenase-1 system, is actually considered the common denominator for this dual response. However, evidence-based medicine has clearly demonstrated the lack of any therapeutic effect of curcumin to contrast the onset or progression of neurodegeneration and related diseases. Finally, the curcumin safety profile imposes a careful analysis of the risk/benefit balance prior to proposing chronic supplementation with curcumin.
Collapse
Affiliation(s)
- Maria Concetta Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Cesare Mancuso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
- Institute of Pharmacology, Catholic University of Sacred Heart, 00168 Roma, Italy.
| | - Barbara Tomasello
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Maria Laura Ontario
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Andrea Cavallaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Francesco Frasca
- Department of Clinical and experimental Medicine, Division of Endocrinology, University of Catania, 95125 Catania, Italy.
| | - Luigi Maiolino
- Department of Medical and Surgery Sciences, University of Catania, 95125 Catania, Italy.
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| | - Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Science, University of Massachusetts, Amherst, MA 01003, USA.
| | - Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Torre Biologica, Via Santa Sofia, 97-95125 Catania, Italy.
| |
Collapse
|
14
|
Daenen K, Andries A, Mekahli D, Van Schepdael A, Jouret F, Bammens B. Oxidative stress in chronic kidney disease. Pediatr Nephrol 2019; 34:975-991. [PMID: 30105414 DOI: 10.1007/s00467-018-4005-4] [Citation(s) in RCA: 491] [Impact Index Per Article: 81.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 06/03/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS), defined as disturbances in the pro-/antioxidant balance, is harmful to cells due to the excessive generation of highly reactive oxygen (ROS) and nitrogen (RNS) species. When the balance is not disturbed, OS has a role in physiological adaptations and signal transduction. However, an excessive amount of ROS and RNS results in the oxidation of biological molecules such as lipids, proteins, and DNA. Oxidative stress has been reported in kidney disease, due to both antioxidant depletions as well as increased ROS production. The kidney is a highly metabolic organ, rich in oxidation reactions in mitochondria, which makes it vulnerable to damage caused by OS, and several studies have shown that OS can accelerate kidney disease progression. Also, in patients at advanced stages of chronic kidney disease (CKD), increased OS is associated with complications such as hypertension, atherosclerosis, inflammation, and anemia. In this review, we aim to describe OS and its influence on CKD progression and its complications. We also discuss the potential role of various antioxidants and pharmacological agents, which may represent potential therapeutic targets to reduce OS in both pediatric and adult CKD patients.
Collapse
Affiliation(s)
- Kristien Daenen
- Department of Microbiology and Immunology, Laboratory of Nephrology, KU Leuven - University of Leuven, 3000, Leuven, Belgium.
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, 3000, Leuven, Belgium.
- Department of Nephrology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Asmin Andries
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, KU Leuven - University of Leuven, 3000, Leuven, Belgium
| | - Djalila Mekahli
- Department of Development and Regeneration, Laboratory of Pediatrics, PKD Group, KU Leuven - University of Leuven, 3000, Leuven, Belgium
- Department of Pediatric Nephrology, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Ann Van Schepdael
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, KU Leuven - University of Leuven, 3000, Leuven, Belgium
| | - François Jouret
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital (ULg CHU), Liège, Belgium
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Science, University of Liège, Liège, Belgium
| | - Bert Bammens
- Department of Microbiology and Immunology, Laboratory of Nephrology, KU Leuven - University of Leuven, 3000, Leuven, Belgium
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, 3000, Leuven, Belgium
| |
Collapse
|
15
|
Mahan VL. Neurointegrity and neurophysiology: astrocyte, glutamate, and carbon monoxide interactions. Med Gas Res 2019; 9:24-45. [PMID: 30950417 PMCID: PMC6463446 DOI: 10.4103/2045-9912.254639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022] Open
Abstract
Astrocyte contributions to brain function and prevention of neuropathologies are as extensive as that of neurons. Astroglial regulation of glutamate, a primary neurotransmitter, is through uptake, release through vesicular and non-vesicular pathways, and catabolism to intermediates. Homeostasis by astrocytes is considered to be of primary importance in determining normal central nervous system health and central nervous system physiology - glutamate is central to dynamic physiologic changes and central nervous system stability. Gasotransmitters may affect diverse glutamate interactions positively or negatively. The effect of carbon monoxide, an intrinsic central nervous system gasotransmitter, in the complex astrocyte homeostasis of glutamate may offer insights to normal brain development, protection, and its use as a neuromodulator and neurotherapeutic. In this article, we will review the effects of carbon monoxide on astrocyte homeostasis of glutamate.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Division of Pediatric Cardiothoracic Surgery in the Department of Surgery, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
16
|
NRF2 and HSF1 coordinately regulate heme oxygenase-1 expression. Biochem Biophys Res Commun 2018; 506:7-11. [DOI: 10.1016/j.bbrc.2018.10.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/05/2018] [Indexed: 01/13/2023]
|
17
|
Aztatzi-Aguilar OG, Valdés-Arzate A, Debray-García Y, Calderón-Aranda ES, Uribe-Ramirez M, Acosta-Saavedra L, Gonsebatt ME, Maciel-Ruiz JA, Petrosyan P, Mugica-Alvarez V, Gutiérrez-Ruiz MC, Gómez-Quiroz LE, Osornio-Vargas A, Froines J, Kleinman MT, De Vizcaya-Ruiz A. Exposure to ambient particulate matter induces oxidative stress in lung and aorta in a size- and time-dependent manner in rats. TOXICOLOGY RESEARCH AND APPLICATION 2018. [DOI: 10.1177/2397847318794859] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Exposure to particulate matter (PM) has been implicated in oxidative stress (OxS) and inflammation as underlying mechanisms of lung damage and cardiovascular alterations. PM is a chemical mixture that can be subdivided according to their aerodynamic size into coarse (CP), fine (FP), and ultrafine (UFP) particulates. We investigated, in a rat model, the induction of OxS (protein oxidation and antioxidant response), carcinogen-DNA adduct formation, and inflammatory mediators in lung in response to different airborne particulate fractions, CP, FP, and UFP, after an acute and subchronic exposure. In addition, OxS was evaluated in the aorta to assess the effects beyond the lungs. Exposure to CP, FP, and UFP induced time- and size-dependent lung protein oxidation and DNA adduct formation. After acute and subchronic exposure, nuclear factor erythroid-2 (Nrf2) activation was observed in the lung, by electrophoretic mobility shift assay, and the induction of mRNA antioxidant enzymes in the FP and UFP groups, but not in the CP. Cytokine concentration of interleukin 1β, interleukin 6, and macrophage inflammatory protein-2 was significantly increased in bronchoalveolar lavage fluid after acute exposure to FP and UFP. Activation of Nrf2 and expression of mRNA antioxidant enzymes were observed only after the subchronic exposure to FP and UFP in the aorta. Our results indicate that FP and UFP were mainly accountable for the oxidant toxic effects in the lung; OxS is spread from the lung to the cardiovascular system. We conclude that the biological mechanisms associated with transient OxS and inflammation are particle size and time-dependent exposure resulting in acute lung injury, which later reaches the vascular system.
Collapse
Affiliation(s)
- OG Aztatzi-Aguilar
- Cátedras-CONACYT
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, CDMX, Mexico
| | - A Valdés-Arzate
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, CDMX, México
| | - Y Debray-García
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, CDMX, México
| | - ES Calderón-Aranda
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, CDMX, México
| | - M Uribe-Ramirez
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, CDMX, México
| | - L Acosta-Saavedra
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, CDMX, México
| | - ME Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, México
| | - JA Maciel-Ruiz
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, México
| | - P Petrosyan
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, CDMX, México
| | - V Mugica-Alvarez
- Área de Química Aplicada, Universidad Autónoma Metropolitana, CDMX, México
| | - MC Gutiérrez-Ruiz
- Departamento Ciencias de la Salud, Universidad Autónoma Metropolitana, and Unidad de Medicina Traslacional UNAM/INCICH, Instituto de Investigaciones Biomédicas, CDMX, México
| | - LE Gómez-Quiroz
- Departamento Ciencias de la Salud, Universidad Autónoma Metropolitana, and Unidad de Medicina Traslacional UNAM/INCICH, Instituto de Investigaciones Biomédicas, CDMX, México
| | - A Osornio-Vargas
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - J Froines
- Center for Occupational and Environmental Health, School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - MT Kleinman
- Department of Medicine, School of Medicine, University of California-Irvine, Irvine, CA, USA
| | - A De Vizcaya-Ruiz
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional. Av. Instituto Politécnico Nacional, CDMX, México
| |
Collapse
|
18
|
Kuesap J, Na-Bangchang K. The Effect of ABO Blood Groups, Hemoglobinopathy, and Heme Oxygenase-1 Polymorphisms on Malaria Susceptibility and Severity. THE KOREAN JOURNAL OF PARASITOLOGY 2018; 56:167-173. [PMID: 29742871 PMCID: PMC5976019 DOI: 10.3347/kjp.2018.56.2.167] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 03/20/2018] [Accepted: 04/10/2018] [Indexed: 11/23/2022]
Abstract
Malaria is one of the most important public health problems in tropical areas on the globe. Several factors are associated with susceptibility to malaria and disease severity, including innate immunity such as blood group, hemoglobinopathy, and heme oxygenase-1 (HO-1) polymorphisms. This study was carried out to investigate association among ABO blood group, thalassemia types and HO-1 polymorphisms in malaria. The malarial blood samples were collected from patients along the Thai-Myanmar border. Determination of ABO blood group, thalassemia variants, and HO-1 polymorphisms were performed using agglutination test, low pressure liquid chromatography and polymerase chain reaction, respectively. Plasmodium vivax was the major infected malaria species in the study samples. Distribution of ABO blood type in the malaria-infected samples was similar to that in healthy subjects, of which blood type O being most prevalent. Association between blood group A and decreased risk of severe malaria was significant. Six thalassemia types (30%) were detected, i.e., hemoglobin E (HbE), β-thalassemia, α-thalassemia 1, α-thalassemia 2, HbE with α-thalassemia 2, and β-thalassemia with α-thalassemia 2. Malaria infected samples without thalassemia showed significantly higher risk to severe malaria. The prevalence of HO-1 polymorphisms, S/S, S/L and L/L were 25, 62, and 13%, respectively. Further study with larger sample size is required to confirm the impact of these 3 host genetic factors in malaria patients.
Collapse
Affiliation(s)
- Jiraporn Kuesap
- Faculty of Allied Health Sciences, Thammasat University. Pathumthani, Thailand
| | - Kesara Na-Bangchang
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
19
|
Vanella L, Barbagallo I, Tibullo D, Forte S, Zappalà A, Li Volti G. The non-canonical functions of the heme oxygenases. Oncotarget 2018; 7:69075-69086. [PMID: 27626166 PMCID: PMC5356613 DOI: 10.18632/oncotarget.11923] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/05/2016] [Indexed: 11/25/2022] Open
Abstract
Heme oxygenase (HO) isoforms catalyze the conversion of heme to carbon monoxide (CO) and biliverdin with a concurrent release of iron, which can drive the synthesis of ferritin for iron sequestration. Most of the studies so far were directed at evaluating the protective effect of these enzymes because of their ability to generate antioxidant and antiapoptotic molecules such as CO and bilirubin. Recent evidences are suggesting that HO may possess other important physiological functions, which are not related to its enzymatic activity and for which we would like to introduce for the first time the term “non canonical functions”. Recent evidence suggest that both HO isoforms may form protein-protein interactions (i.e. cytochrome P450, adiponectin, CD91) thus serving as chaperone-like protein. In addition, truncated HO-1 isoform was localized in the nuclear compartment under certain experimental conditions (i.e. excitotoxicity, hypoxia) regulating the activity of important nuclear transcription factors (i.e. Nrf2) and DNA repair. In the present review, we discuss three potential signaling mechanisms that we refer to as the non-canonical functions of the HO isoforms: protein-protein interaction, intracellular compartmentalization, and extracellular secretion. The aim of the present review is to describe each of this mechanism and all the aspects warranting additional studies in order to unravel all the functions of the HO system.
Collapse
Affiliation(s)
- Luca Vanella
- Department of Drug Sciences, University of Catania, Catania, Italy
| | | | - Daniele Tibullo
- Division of Haematology, AOU "Policlinico - Vittorio Emanuele", University of Catania, Catania, Italy
| | - Stefano Forte
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Istituto Oncologico del Mediterraneo Ricerca srl Viagrande, Catania, Italy
| | - Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,EuroMediterranean Institute of Science and Technology, Palermo, Italy
| |
Collapse
|
20
|
Kırıs T, Avcı E, Çelik A. Combined value of left ventricular ejection fraction and the Model for End-Stage Liver Disease (MELD) score for predicting mortality in patients with acute coronary syndrome who were undergoing percutaneous coronary intervention. BMC Cardiovasc Disord 2018; 18:44. [PMID: 29499644 PMCID: PMC5833061 DOI: 10.1186/s12872-018-0782-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 02/23/2018] [Indexed: 12/16/2022] Open
Abstract
Background The purpose of the study was to investigate whether the addition of left ventricular ejection fraction (LVEF) to the MELD score enhances the prediction of mortality in patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI). Methods This retrospective study analyzed 846 consecutive patients with ACS undergoing PCI who were not receiving previous anticoagulant therapy. The patients were grouped as survivors or non-survivors. The MELD score and LVEF were calculated in all patients. The primary end point was all-cause death during the median follow-up of 28 months. Results During the follow-up, there were 183 deaths (21.6%). MELD score was significantly higher in non-survivors than survivors (10.1 ± 4.4 vs 7.8 ± 2.4, p < 0.001). LVEF was lower in non-survivors compared with survivors (41.3 ± 11.8% vs. 47.5 ± 10.0%, p < 0.001). In multivariate analysis, both MELD score and LVEF were independent predictors of total mortality. (HR: 1.116, 95%CI: 1.069–1.164, p < 0.001; HR: 0.972, 95%CI: 0.958–0.986, p < 0.001, respectively). The addition of LVEF to MELD score was associated with significant improvement in predicting mortality compared with the MELD score alone (AUC:0.733 vs 0.690, p < 0.05). Also, the combining LVEF with MELD score improved the reclassification (NRI:24.6%, p < 0.001) and integrated discrimination (IDI:0.045, p < 0.001) of patients compared with MELD score alone. Conclusions Our study demonstrated that the combining LVEF with MELD score may be useful to predict long-term survival in patients with ACS who were undergoing PCI. Electronic supplementary material The online version of this article (10.1186/s12872-018-0782-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tuncay Kırıs
- Department of Cardiology, Izmir Katip Celebi University, Ataturk Training and Research Hospital, 35360, Izmir, Turkey
| | - Eyüp Avcı
- Department of Cardiology, Balikesir University Faculty of Medicine, 10345, Balikesir, Turkey.
| | - Aykan Çelik
- Department of Cardiology, Izmir Katip Celebi University, Ataturk Training and Research Hospital, 35360, Izmir, Turkey
| |
Collapse
|
21
|
Bifurcated BACH2 control coordinates mantle cell lymphoma survival and dispersal during hypoxia. Blood 2017; 130:763-776. [PMID: 28592433 DOI: 10.1182/blood-2017-02-767293] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/31/2017] [Indexed: 02/07/2023] Open
Abstract
BACH2, a B-cell-specific transcription factor, plays a critical role in oxidative stress-mediated drug resistance in mantle cell lymphoma (MCL); however, the biological functions of BACH2 and its regulation of B-cell malignancies in chronic hypoxic microenvironment have not been studied. Here, we found that silencing BACH2 led to not only increased tumor formation and colony formation but also increased tumor dispersal to spleen and bone marrow. Decreased BACH2 levels in patients were also correlated with bone marrow and gastrointestinal dispersal of MCL and blastoid subtypes of MCL. Unexpectedly, decreased BACH2 levels in dispersed MCL cells were due to direct transcriptional repression by hypoxia-induced factor 1α (HIF-1α) and increased heme-mediated protein degradation. In normoxic conditions, BACH2 was able to modulate HIF-1α degradation by suppressing prolyl hydroxylase 3 expression. Bifurcated BACH2 controls during hypoxia and normoxia coordinate not only MCL tumor dispersal but also drug resistance, including bortezomib resistance, via plasmacytic differentiation. Our data highlight an interactive relationship between tumor cells and local microenvironment and the mechanisms of B-cell transcription factor in the regulation of MCL dispersal.
Collapse
|
22
|
Gu Q, Gu Y, Shi Q, Yang H. Hypoxia Promotes Osteogenesis of Human Placental-Derived Mesenchymal Stem Cells. TOHOKU J EXP MED 2017; 239:287-96. [PMID: 27477937 DOI: 10.1620/tjem.239.287] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Placental-derived mesenchymal stem cells (pMSCs) are promising candidates for regenerative medicine because they possess high proliferative capacity and multi-differentiation potential. Human pMSCs are residing in an environment with low oxygen tension in the body. Heme oxygenase-1 (HO-1) is known to participate in the regulation of MSC differentiation. The present study aimed to investigate the impact of hypoxia on the osteogenic differentiation of human pMSCs, and to elucidate the role of HO-1 in the osteogenic differentiation of hypoxic pMSCs. Human pMSCs were cultured under normoxia (21% O2) or hypoxia (5% O2) for 3 days. We found that hypoxia maintained the morphology and immunophenotype of human pMSCs. The expression of stemness markers Oct4, Nanog, and Sox2 was increased under hypoxia. After a 5-day hypoxic culture, the proliferation ability of pMSCs was increased, which might be correlated with the increased expression of stem cell factor. During osteogenic induction, hypoxia increased the expression of osteogenic genes including osteopontin, osteocalcin, and alkaline phosphatase (ALP). Moreover, hypoxia increased the mineralization and ALP levels of human pMSCs as evidenced by Alizarin Red staining and ALP staining. Upregulation of HO-1 by cobalt-protoporphyrin treatment increased the osteogenic differentiation of pMSCs under hypoxia, while inhibition of HO-1 by Zn-protoporphyrin reduced the osteogenic differentiation of hypoxic pMSCs. Taken together, our data suggest that hypoxia can promote the osteogenic differentiation of human pMSCs. Upregulation of HO-1 can further increase the osteogenesis of human pMSCs under hypoxia. Our findings will highlight the therapeutic potential of MSCs in the tissue engineering of bones.
Collapse
Affiliation(s)
- Qiaoli Gu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University
| | | | | | | |
Collapse
|
23
|
Mancuso C. Bilirubin and brain: A pharmacological approach. Neuropharmacology 2017; 118:113-123. [PMID: 28315352 DOI: 10.1016/j.neuropharm.2017.03.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/09/2017] [Accepted: 03/12/2017] [Indexed: 01/01/2023]
Abstract
For many decades, the world scientific literature has accounted for a number of works on the biological effects of bilirubin-IXalpha (BR). The first studies focused on the neurotoxic effects of the excessive production of BR, in particular regarding both physiological neonatal jaundice and the more severe ones, typically as consequences of severe hemolysis or other underlying diseases. Only since 1987, has significant evidence, however, underlined the neuroprotective role of BR linked to the scavenging effect of free radicals as reactive oxygen species and nitric oxide and its congeners. Despite the presence in the literature of many excellent papers dealing with the multiple roles played by BR in health and disease, there were very few and somewhat dated reviews that summarize the key findings related to the neuroprotective and neurotoxic effects of the bile pigment and underlying mechanisms. In light of the previous statements, the aim of this review is to provide a summary of the main discoveries in the last years on the effects of BR on the central nervous system. An analytical description about the synthesis of BR, its distribution in the systemic circulation, liver metabolism and elimination through feces and urine will be provided, together with the main mechanisms claimed to describe the neurotoxicity and neuroprotection by the bile pigment. Finally, the possible translational aspects of pharmacological modulation in the production of BR in order to prevent or counteract toxic effects or enhance the protective actions, will be discussed.
Collapse
Affiliation(s)
- Cesare Mancuso
- Institute of Pharmacology, Catholic University School of Medicine, Largo F. Vito, 1-00168 Rome, Italy.
| |
Collapse
|
24
|
Satarug S, Vesey DA, Gobe GC. Kidney Cadmium Toxicity, Diabetes and High Blood Pressure: The Perfect Storm. TOHOKU J EXP MED 2017; 241:65-87. [DOI: 10.1620/tjem.241.65] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Soisungwan Satarug
- Centre for Kidney Disease Research, Translational Research Institute and The University of Queensland Diamantina Institute
| | - David A. Vesey
- Centre for Kidney Disease Research, Translational Research Institute and The University of Queensland Diamantina Institute
| | - Glenda C. Gobe
- Centre for Kidney Disease Research, Translational Research Institute and The University of Queensland Diamantina Institute
| |
Collapse
|
25
|
|
26
|
Matsumoto M, Kondo K, Shiraki T, Brydun A, Funayama R, Nakayama K, Yaegashi N, Katagiri H, Igarashi K. Genomewide approaches for BACH1 target genes in mouse embryonic fibroblasts showed BACH1-Ppargpathway in adipogenesis. Genes Cells 2016; 21:553-67. [DOI: 10.1111/gtc.12365] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/23/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Mitsuyo Matsumoto
- Department of Biochemistry; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| | - Keiichi Kondo
- Division of Metabolism and Diabetes; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| | - Takuma Shiraki
- Department of Biochemistry; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
- Faculty of Biology-Oriented Science and Technology; Kinki University; Nishimitani Kinokawashi Wakayama 649-6493 Japan
| | - Andrey Brydun
- Department of Biochemistry; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| | - Ryo Funayama
- Division of Cell Proliferation; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| | - Keiko Nakayama
- Division of Cell Proliferation; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| | - Hideki Katagiri
- Division of Metabolism and Diabetes; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
- AMED-CREST; Japan Agency for Medical Research and Development; Tokyo 100-0004 Japan
- Center for Regulatory Epigenome and Diseases; Tohoku University Graduate School of Medicine; 2-1, Seiryo-machi Aoba-ku Sendai 980-8575 Japan
| |
Collapse
|
27
|
Samatiwat P, Takeda K, Satarug S, Ohba K, Kukongviriyapan V, Shibahara S. Induction of MITF expression in human cholangiocarcinoma cells and hepatocellular carcinoma cells by cyclopamine, an inhibitor of the Hedgehog signaling. Biochem Biophys Res Commun 2016; 470:144-149. [PMID: 26773496 DOI: 10.1016/j.bbrc.2016.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/04/2016] [Indexed: 12/23/2022]
Abstract
Microphthalmia-associated transcription factor (MITF) is a key regulator of differentiation of melanocytes and retinal pigment epithelial cells, but it also has functions in non-pigment cells. MITF consists of multiple isoforms, including widely expressed MITF-A and MITF-H. In the present study, we explored the potential role played by the Hedgehog signaling on MITF expression in two common types of primary liver cancer, using human cholangiocarcinoma cell lines, the KKU-100 and HuCCT1, along with the HepG2 human hepatocellular carcinoma cell line. Importantly, cholangiocarcinoma is characterized by the activated Hedgehog signaling. Here we show that MITF-A mRNA is predominantly expressed in all three human liver cancer cell lines examined. Moreover, cyclopamine, an inhibitor of the Hedgehog signalling, increased the expression levels of MITF proteins in HuCCT1 and HepG2 cells, but not in KKU-100 cells, suggesting that MITF expression may be down-regulated in some liver cancer cases.
Collapse
Affiliation(s)
- Papavee Samatiwat
- Department of Molecular Biology and Applied Physiology, Tohoku University School of Medicine, Sendai, Japan; Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kazuhisa Takeda
- Department of Molecular Biology and Applied Physiology, Tohoku University School of Medicine, Sendai, Japan.
| | - Soisungwan Satarug
- Department of Molecular Biology and Applied Physiology, Tohoku University School of Medicine, Sendai, Japan; Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland School of Medicine, Woolloongabba, Brisbane, Queensland, Australia
| | - Koji Ohba
- Department of Molecular Biology and Applied Physiology, Tohoku University School of Medicine, Sendai, Japan
| | | | - Shigeki Shibahara
- Department of Molecular Biology and Applied Physiology, Tohoku University School of Medicine, Sendai, Japan
| |
Collapse
|
28
|
Li BZ, Guo B, Zhang HY, Liu J, Tao SS, Pan HF, Ye DQ. Therapeutic potential of HO-1 in autoimmune diseases. Inflammation 2015; 37:1779-88. [PMID: 24818708 DOI: 10.1007/s10753-014-9908-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Heme oxygenase-1 (HO-1), the inducible isoform of heme oxygenase (HO), has raised a lot of concerns in recent years due to its multiple functions. HO-1 was found to be a pivotal cytoprotective, antioxidant, anti-apoptotic, immunosuppressive, as well as anti-inflammatory molecule. Recent studies have clarified its significant functions in many diseases with substantial findings. In autoimmune diseases, HO-1 may have promising therapeutic potential. Here, we briefly reviewed recent advances in this field, aiming at hopefully exploring the potential therapeutic roles of HO-1, and design HO-1-based strategies for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Daenen KEL, Martens P, Bammens B. Association of HO-1 (GT)n Promoter Polymorphism and Cardiovascular Disease: A Reanalysis of the Literature. Can J Cardiol 2015; 32:160-8. [PMID: 26483091 DOI: 10.1016/j.cjca.2015.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 05/27/2015] [Accepted: 06/11/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Heme-oxygenase 1 (HO-1), an inducible heme-degrading enzyme, has antiatherogenic effects through its enzymatic end products. HO-1 gene expression is modulated by a guanidine thymidine dinucleotide ([GT]n) repeat polymorphism in the promoter region. Shorter repeats with (GT)n < 25 are associated with higher inducibility and activity of HO-1. METHODS We performed a systematic review of all literature from 1997 to 2013 on the association of the HO-1 (GT)n and cardiovascular disease (CVD). On the basis of predefined criteria (patient characteristics, genotype data format, allelic distribution, repeat length cutoff) 41 articles were selected. Patients were redistributed into 4 homogeneous subpopulations: patients with CVD (CVD group), patients without CVD (nonCVD), 'controls' with unknown cardiovascular status (unspecified) and children younger than 20 years of age (unselected). Genotype distributions (homozygous short [SS] or long [LL], and heterozygous) of the 4 patient categories were compared and odds ratios (ORs) for CVD were calculated using logistic regression analysis. RESULTS Overall, the proportion of the SS genotype was lower in CVD compared with nonCVD and unspecified. The ORs for CVD was highest in patients carrying the LL genotype (OR LL vs SS, 1.769 [95% confidence interval, 1.594-1.963]). Furthermore, genotype distribution differed between Caucasian and Asian individuals, the latter having a much higher proportion of the SS genotype (22% vs 11%). CONCLUSIONS This review of the available literature on the epidemiological association between the HO-1 (GT)n repeat polymorphism and CVD supports the presumed protective effects of HO-1. The second but probably even more relevant finding of our review is that racial disparities in HO-1 (GT)n repeat length distribution exist and might influence the associations of the genotype with CVD status.
Collapse
Affiliation(s)
- Kristien E L Daenen
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium; Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Martens
- Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Bert Bammens
- Laboratory of Nephrology, Department of Immunology and Microbiology, KU Leuven, Leuven, Belgium; Department of Nephrology, Dialysis and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
30
|
Baumann S, Huseynov A, Koepp J, Jabbour C, Behnes M, Becher T, Renker M, Lang S, Borggrefe M, Lehmann R, Akin I. Comparison of Serum Uric Acid, Bilirubin, and C-Reactive Protein as Prognostic Biomarkers of In-Hospital MACE Between Women and Men With ST-Segment Elevation Myocardial Infarction. Angiology 2015; 67:272-80. [PMID: 26032849 DOI: 10.1177/0003319715589246] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Levels of C-reactive protein (CRP), uric acid (UA), and total bilirubin (TB) are associated with coronary artery disease and major adverse cardiac events (MACEs) in patients with ST-segment elevation myocardial infarction (STEMI). We retrospectively included 1167 patients with STEMI who underwent percutaneous coronary intervention and routine blood sampling. The study cohort consisted of 803 patients (73.1% male, mean age 62.5 ± 13.4 years). In men, the levels of CRP, TB, and UA were significantly higher in the MACE than in the non-MACE group (P < .05). The receiver-operating characteristic (ROC) analysis shows that CRP (area under the curve [AUC]: 0.59; 95% confidence interval [CI]: 0.53-0.66; P = .014) and TB (AUC: 0.58; 95% CI: 0.51-0.65; P = .019) are significantly associated with MACE but not UA (AUC: 0.61; 95% CI: 0.42-0.76; P = .083). Logistic regression revealed CRP (odds ratio [OR] 1.01; 95% CI: 1.00-1.01; P = .006) and TB (OR 2.03; 95% CI: 1.12-3.40; P = .007) as an independent predictor for MACE. In women, none of the biomarkers was associated with MACE by ROC analysis or logistic regression analysis. This study demonstrated that high CRP and TB serum levels have a prognostic association with in-hospital MACE in male patients with STEMI.
Collapse
Affiliation(s)
- Stefan Baumann
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Aydin Huseynov
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Johanna Koepp
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Claude Jabbour
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Michael Behnes
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Tobias Becher
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Matthias Renker
- Department of Internal Medicine I, Cardiology and Angiology, Giessen University, Giessen, Germany
| | - Siegfried Lang
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Martin Borggrefe
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ralf Lehmann
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| |
Collapse
|
31
|
Gaucher C, Domingues-Hamdi É, Prin-Mathieu C, Menu P, Baudin-Creuza V. Interaction of recombinant octameric hemoglobin with endothelial cells. C R Biol 2015; 338:95-102. [DOI: 10.1016/j.crvi.2014.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/12/2014] [Accepted: 11/12/2014] [Indexed: 10/24/2022]
|
32
|
Microphthalmia-associated transcription factor as the molecular target of cadmium toxicity in human melanocytes. Biochem Biophys Res Commun 2014; 454:594-9. [DOI: 10.1016/j.bbrc.2014.10.141] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 10/28/2014] [Indexed: 12/26/2022]
|
33
|
Kühl T, Imhof D. Regulatory Fe(II/III) heme: the reconstruction of a molecule's biography. Chembiochem 2014; 15:2024-35. [PMID: 25196849 DOI: 10.1002/cbic.201402218] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Indexed: 11/10/2022]
Abstract
More than 20 years of research on heme as a temporary effector molecule of proteins have revealed its widespread impact on virtually all primary functions in the human organism. As our understanding of this influence is still growing, a comprehensive overview of compiled data will give fresh impetus for creativity and developing new strategies in heme-related research. From known data concerning heme-regulated proteins and their involvement in the development of diseases, we provide concise information of Fe(II/III) heme as a regulator and the availability of "regulatory heme". The latter is dependent on the balance between free and bound Fe(II/III) heme, here termed "hemeostasis". Imbalance of this system can lead to the development of diseases that were not always attributed to this small molecule. Diseases such as cancer or Alzheimer's disease highlight the reawakened interest in heme, whose function was previously believed to be completely understood.
Collapse
Affiliation(s)
- Toni Kühl
- Pharmaceutical Chemistry I, Pharmaceutical Institute, University of Bonn, Brühler Strasse 7, 53119 Bonn (Germany).
| | | |
Collapse
|
34
|
Stulnig G, Frisch MT, Crnkovic S, Stiegler P, Sereinigg M, Stacher E, Olschewski H, Olschewski A, Frank S. Docosahexaenoic acid (DHA)-induced heme oxygenase-1 attenuates cytotoxic effects of DHA in vascular smooth muscle cells. Atherosclerosis 2013; 230:406-13. [PMID: 24075775 DOI: 10.1016/j.atherosclerosis.2013.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 07/19/2013] [Accepted: 08/03/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Docosahexaenoic acid (DHA), a member of n-3 polyunsaturated fatty acids (n-3 PUFA) is a potent regulator of molecular events implicated in cardiovascular health. In a previous study we found that Ca(2+)-dependent oxidative stress is the central and initial event responsible for induction of unfolded protein response (UPR), cell cycle arrest and apoptosis in DHA treated primary human smooth muscle cells isolated from small pulmonary artery (hPASMC). In the present study we examined the impact of heme oxygenase (HO)-1, induced by DHA, on DHA-induced oxidative stress, UPR, cell proliferation and apoptosis in hPASMC. METHODS & RESULTS DHA led to a time- and concentration-dependent increase in HO-1 mRNA and protein levels in hPASMC. The DHA-induced HO-1 upregulation could be attenuated by preincubation of cells with a strong antioxidant Tempol or by siRNA-mediated depletion of nuclear factor erythroid 2-related factor-2 (Nrf2). In DHA-treated hPASMC, depletion of HO-1 by siRNA-mediated silencing resulted in increased levels of reactive oxygen species (ROS) and increased duration of UPR, the latter revealed by monitoring of spliced X-box binding protein 1 (XBP-1) variant. Moreover, HO-1 silencing augmented apoptosis in DHA-treated hPASMC as found by increased numbers of cleaved caspase-3-positive cells. HO-1 silencing did not affect proliferation of hPASMC exposed to DHA. CONCLUSION Our results indicate that DHA-induced, ROS-dependent upregulation of HO-1 attenuates oxidative stress, UPR and apoptosis in DHA-treated hPASMC.
Collapse
Affiliation(s)
- Gabriel Stulnig
- Institute of Molecular Biology and Biochemistry, Center of Molecular Medicine, Medical University of Graz, Harrachgasse 21/III, A-8010 Graz, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
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: 1.2] [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
|
36
|
Gul M, Uyarel H, Ergelen M, Akgul O, Karaca G, Turen S, Ugur M, Ertürk M, Kul S, Surgit O, Bozbay M, Uslu N. Prognostic value of total bilirubin in patients with ST-segment elevation acute myocardial infarction undergoing primary coronary intervention. Am J Cardiol 2013; 111:166-171. [PMID: 23102877 DOI: 10.1016/j.amjcard.2012.09.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Revised: 09/25/2012] [Accepted: 09/25/2012] [Indexed: 02/07/2023]
Abstract
Previous studies have shown that the serum total bilirubin (TB) concentration was inversely related with stable coronary artery disease, diabetes mellitus, hypertension, and metabolic syndromes. The relation between TB levels and in-hospital and long-term outcomes in patients with ST-segment elevation myocardial infarction (STEMI) who undergo primary percutaneous coronary intervention (PCI) is not known. Data from 1,624 consecutive patients with STEMI who underwent primary PCI were evaluated. TB was measured after primary PCI, and the study population was divided into tertiles. The high TB group (n = 450) was defined as a value in the upper third tertile (>0.9 mg/dl) and the low TB group (n = 1,174) as any value in the lower 2 tertiles (≤0.9 mg/dl). The in-hospital mortality rate was significantly greater in the high TB group than in the low TB group (4% vs 1.5%, p = 0.003). In the multivariate analyses, a significant association was noted between high TB levels and the adjusted risk of in-hospital cardiovascular mortality (odds ratio 3.24, 95% confidence interval 1.27 to 8.27, p = 0.014). In the receiver operating characteristic curve analysis, TB >0.90 mg/dl was identified as an effective cutpoint in patients with STEMI for in-hospital cardiovascular mortality (area under the curve 0.66, 95% confidence interval 0.55 to 0.76, p = 0.001). The mean follow-up period was 26.2 months. No differences were seen in the long-term mortality rates between the 2 groups. In conclusion, high TB is independently associated with in-hospital adverse outcomes in patients with STEMI who undergo primary PCI. However, no association was found with long-term mortality.
Collapse
Affiliation(s)
- Mehmet Gul
- Department of Cardiology, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Satarug S, Moore MR. Emerging roles of cadmium and heme oxygenase in type-2 diabetes and cancer susceptibility. TOHOKU J EXP MED 2012; 228:267-88. [PMID: 23117262 DOI: 10.1620/tjem.228.267] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Many decades after an outbreak of severe cadmium poisoning, known as Itai-itai disease, cadmium continues to pose a significant threat to human health worldwide. This review provides an update on the effects of this environmental toxicant cadmium, observed in numerous populations despite modest exposure levels. In addition, it describes the current knowledge on the link between heme catabolism and glycolysis. It examines novel functions of heme oxygenase-2 (HO-2) that protect against type 2-diabetes and obesity, which have emerged from diabetic/obese phenotypes of the HO-2 knockout mouse model. Increased cancer susceptibility in type-2 diabetes has been noted in several large cohorts. This is a cause for concern, given the high prevalence of type-2 diabetes worldwide. A lifetime exposure to cadmium is associated with pre-diabetes, diabetes, and overall cancer mortality with sex-related differences in specific types of cancer. Liver and kidney are target organs for the toxic effects of cadmium. These two organs are central to the maintenance of blood glucose levels. Further, inhibition of gluconeogenesis is a known effect of heme, while cadmium has the propensity to alter heme catabolism. This raises the possibility that cadmium may mimic certain HO-2 deficiency conditions, resulting in diabetic symptoms. Intriguingly, evidence has emerged from a recent study to suggest the potential interaction and co-regulation of HO-2 with the key regulator of glycolysis: 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 (PFKFB4). HO-2 could thus be critical to a metabolic switch to cancer-prone cells because the enzyme PFKFB and glycolysis are metabolic requirements for cell proliferation and resistance to apoptosis.
Collapse
Affiliation(s)
- Soisungwan Satarug
- Center for Kidney Disease Research, University of Queensland School of Medicine, Brisbane, Australia.
| | | |
Collapse
|
38
|
Elias RM, Correa-Costa M, Barreto CR, Silva RC, Hayashida CY, Castoldi Â, Gonçalves GM, Braga TT, Barboza R, Rios FJ, Keller AC, Cenedeze MA, Hyane MI, D'Império-Lima MR, Figueiredo-Neto AM, Reis MA, Marinho CRF, Pacheco-Silva A, Câmara NOS. Oxidative stress and modification of renal vascular permeability are associated with acute kidney injury during P. berghei ANKA infection. PLoS One 2012; 7:e44004. [PMID: 22952850 PMCID: PMC3432099 DOI: 10.1371/journal.pone.0044004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/27/2012] [Indexed: 12/21/2022] Open
Abstract
Malaria associated-acute kidney injury (AKI) is associated with 45% of mortality in adult patients hospitalized with severe form of the disease. However, the causes that lead to a framework of malaria-associated AKI are still poorly characterized. Some clinical studies speculate that oxidative stress products, a characteristic of Plasmodium infection, as well as proinflammatory response induced by the parasite are involved in its pathophysiology. Therefore, we aimed to investigate the development of malaria-associated AKI during infection by P. berghei ANKA, with special attention to the role played by the inflammatory response and the involvement of oxidative stress. For that, we took advantage of an experimental model of severe malaria that showed significant changes in the renal pathophysiology to investigate the role of malaria infection in the renal microvascular permeability and tissue injury. Therefore, BALB/c mice were infected with P. berghei ANKA. To assess renal function, creatinine, blood urea nitrogen, and ratio of proteinuria and creatininuria were evaluated. The products of oxidative stress, as well as cytokine profile were quantified in plasma and renal tissue. The change of renal microvascular permeability, tissue hypoxia and cellular apoptosis were also evaluated. Parasite infection resulted in renal dysfunction. Furthermore, we observed increased expression of adhesion molecule, proinflammatory cytokines and products of oxidative stress, associated with a decrease mRNA expression of HO-1 in kidney tissue of infected mice. The measurement of lipoprotein oxidizability also showed a significant increase in plasma of infected animals. Together, our findings support the idea that products of oxidative stress, as well as the immune response against the parasite are crucial to changes in kidney architecture and microvascular endothelial permeability of BALB/c mice infected with P. berghei ANKA.
Collapse
Affiliation(s)
- Rosa Maria Elias
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Matheus Correa-Costa
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Reinaldo Correia Silva
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Caroline Y. Hayashida
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Ângela Castoldi
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Giselle Martins Gonçalves
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Tarcio Teodoro Braga
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Renato Barboza
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Francisco José Rios
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Marcos Antonio Cenedeze
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Meire Ioshie Hyane
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | - Maria Regina D'Império-Lima
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Marlene Antônia Reis
- Divisão de Patologia, Universidade Federal do Triângulo Mineiro, Uberaba, Brazil
| | | | - Alvaro Pacheco-Silva
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto Israelita de Ensino e Pesquisa Albert Einstein, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Universidade de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
39
|
Barone E, Di Domenico F, Sultana R, Coccia R, Mancuso C, Perluigi M, Butterfield DA. Heme oxygenase-1 posttranslational modifications in the brain of subjects with Alzheimer disease and mild cognitive impairment. Free Radic Biol Med 2012; 52:2292-301. [PMID: 22549002 PMCID: PMC3377854 DOI: 10.1016/j.freeradbiomed.2012.03.020] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 02/07/2012] [Accepted: 03/19/2012] [Indexed: 01/21/2023]
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder characterized by progressive cognitive impairment and neuropathology. Oxidative and nitrosative stress plays a principal role in the pathogenesis of AD. The induction of the heme oxygenase-1/biliverdin reductase-A (HO-1/BVR-A) system in the brain represents one of the earliest mechanisms activated by cells to counteract the noxious effects of increased reactive oxygen species and reactive nitrogen species. Although initially proposed as a neuroprotective system in AD brain, the HO-1/BVR-A pathophysiological features are under debate. We previously reported alterations in BVR activity along with decreased phosphorylation and increased oxidative/nitrosative posttranslational modifications in the brain of subjects with AD and those with mild cognitive impairment (MCI). Furthermore, other groups proposed the observed increase in HO-1 in AD brain as a possible neurotoxic mechanism. Here we provide new insights about HO-1 in the brain of subjects with AD and MCI, the latter condition being the transitional phase between normal aging and early AD. HO-1 protein levels were significantly increased in the hippocampus of AD subjects, whereas HO-2 protein levels were significantly decreased in both AD and MCI hippocampi. In addition, significant increases in Ser-residue phosphorylation together with increased oxidative posttranslational modifications were found in the hippocampus of AD subjects. Interestingly, despite the lack of oxidative stress-induced AD neuropathology in cerebellum, HO-1 demonstrated increased Ser-residue phosphorylation and oxidative posttranslational modifications in this brain area, suggesting HO-1 as a target of oxidative damage even in the cerebellum. The significance of these findings is profound and opens new avenues into the comprehension of the role of HO-1 in the pathogenesis of AD.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
- Institute of Pharmacology, Catholic University School of Medicine, Largo F. Vito, 1, 00168 Roma, Italy
| | - Fabio Di Domenico
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | - Raffaella Coccia
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Cesare Mancuso
- Institute of Pharmacology, Catholic University School of Medicine, Largo F. Vito, 1, 00168 Roma, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - D. Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
- Address correspondence to: Prof. D. Allan Butterfield, Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA. Tel: 859-257-3184, Fax: 859-259-5876,
| |
Collapse
|
40
|
Sahin O, Akpek M, Elcik D, Karadavut S, Simsek V, Tulmac M, Orscelik O, Calapkorur B, Ergin A, Kaya MG. Bilirubin levels and the burden of coronary atherosclerosis in patients with STEMI. Angiology 2012; 64:200-4. [PMID: 22492252 DOI: 10.1177/0003319712440619] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated whether serum bilirubin level (a marker of heme oxygenase activity) is a predictor of high levels of SYNTAX score (SXscore) in patients with acute myocardial infarction. Patients (n = 281; male 77%; mean age 60 ± 12) who were admitted with ST-elevation myocardial infarctions (STEMIs) were enrolled. Patients were divided into 2 groups. Group 1 was defined as SXscore <22 and group 2 was defined as SXscore ≥22. Total bilirubin levels were significantly higher in the high-SXscore group than in the low-SXscore group (0.86 ± 0.42 vs 1.02 ± 0.51, P = .005). A significant correlation was detected between total bilirubin and SXscore (r = .42; P = .001). At multivariate analysis, total bilirubin (odds ratio: 1.86, 95% confidence interval 1.04-3.35; P = .038) was an independent risk factor for high SXscore in patients with STEMI. In conclusion, serum bilirubin level is independently associated with SXscore in patients with STEMI.
Collapse
Affiliation(s)
- Omer Sahin
- Department of Cardiology, Erciyes University School of Medicine, Kayseri, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Walther M, De Caul A, Aka P, Njie M, Amambua-Ngwa A, Walther B, Predazzi IM, Cunnington A, Deininger S, Takem EN, Ebonyi A, Weis S, Walton R, Rowland-Jones S, Sirugo G, Williams SM, Conway DJ. HMOX1 gene promoter alleles and high HO-1 levels are associated with severe malaria in Gambian children. PLoS Pathog 2012; 8:e1002579. [PMID: 22438807 PMCID: PMC3305414 DOI: 10.1371/journal.ppat.1002579] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 01/26/2012] [Indexed: 01/16/2023] Open
Abstract
Heme oxygenase 1 (HO-1) is an essential enzyme induced by heme and multiple stimuli associated with critical illness. In humans, polymorphisms in the HMOX1 gene promoter may influence the magnitude of HO-1 expression. In many diseases including murine malaria, HO-1 induction produces protective anti-inflammatory effects, but observations from patients suggest these may be limited to a narrow range of HO-1 induction, prompting us to investigate the role of HO-1 in malaria infection. In 307 Gambian children with either severe or uncomplicated P. falciparum malaria, we characterized the associations of HMOX1 promoter polymorphisms, HMOX1 mRNA inducibility, HO-1 protein levels in leucocytes (flow cytometry), and plasma (ELISA) with disease severity. The (GT)(n) repeat polymorphism in the HMOX1 promoter was associated with HMOX1 mRNA expression in white blood cells in vitro, and with severe disease and death, while high HO-1 levels were associated with severe disease. Neutrophils were the main HO-1-expressing cells in peripheral blood, and HMOX1 mRNA expression was upregulated by heme-moieties of lysed erythrocytes. We provide mechanistic evidence that induction of HMOX1 expression in neutrophils potentiates the respiratory burst, and propose this may be part of the causal pathway explaining the association between short (GT)(n) repeats and increased disease severity in malaria and other critical illnesses. Our findings suggest a genetic predisposition to higher levels of HO-1 is associated with severe illness, and enhances the neutrophil burst leading to oxidative damage of endothelial cells. These add important information to the discussion about possible therapeutic manipulation of HO-1 in critically ill patients.
Collapse
Affiliation(s)
- Michael Walther
- Medical Research Council Laboratories, Fajara, Banjul, Gambia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Murakami A, Fujimori Y, Yoshikawa Y, Yamada S, Tamura K, Hirayama N, Terada T, Kuribayashi K, Tabata C, Fukuoka K, Tamaoki T, Nakano T. Heme oxygenase-1 promoter polymorphism is associated with risk of malignant mesothelioma. Lung 2012; 190:333-7. [PMID: 22271370 DOI: 10.1007/s00408-012-9371-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 01/04/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND Malignant mesothelioma is an aggressive tumor of serosal surfaces that is closely associated with asbestos exposure which induces oxidative stress. Heme oxygenase (HO)-1, a rate-limiting enzyme of heme degradation, plays a protective role against oxidative stress. The HO-1 gene promoter carries (GT)n repeats whose number is inversely related to transcriptional activity of the HO-1 gene. METHODS To investigate the relationship between the length polymorphism of (GT)n repeats and mesothelioma susceptibility, we analyzed the HO-1 promoter in 44 asbestos-exposed subjects without mesothelioma and 78 asbestos-exposed subjects with mesothelioma using PCR-based genotyping. RESULTS The number of repeats ranged from 16 to 38, with two peaks at 23 and 30 repeats. Polymorphisms of (GT)n repeats were grouped into two classes of alleles, short (S) (<24) and long (L) (≥24), and three genotypes: L/L, L/S, and S/S. The proportions of allele frequencies in class L as well as genotypic frequencies of L allele carriers (L/L and L/S) were significantly higher in the asbestos-exposed subjects with mesothelioma than in those without mesothelioma. CONCLUSION The findings of this study suggest that long (GT)n repeats in the HO-1 gene promoter are associated with a higher risk of malignant mesothelioma in the Japanese population.
Collapse
Affiliation(s)
- Aki Murakami
- Division of Respiratory Medicine, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yang JJ, Yang X, Liu ZQ, Hu SY, Du ZY, Feng LL, Liu JF, Chen YD. Transplantation of Adipose Tissue-Derived Stem Cells Overexpressing Heme Oxygenase-1 Improves Functions and Remodeling of Infarcted Myocardium in Rabbits. TOHOKU J EXP MED 2012; 226:231-41. [DOI: 10.1620/tjem.226.231] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Jun-jie Yang
- Department of Cardiology, PLA General Hospital
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences
| | - Xia Yang
- Department of Cardiology, PLA General Hospital
| | - Zhi-qiang Liu
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences
| | - Shun-yin Hu
- Department of Cardiology, PLA General Hospital
| | - Zhi-yan Du
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences
| | - Lan-lan Feng
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences
| | | | | |
Collapse
|
44
|
Calabrese V, Cornelius C, Dinkova-Kostova AT, Iavicoli I, Di Paola R, Koverech A, Cuzzocrea S, Rizzarelli E, Calabrese EJ. Cellular stress responses, hormetic phytochemicals and vitagenes in aging and longevity. Biochim Biophys Acta Mol Basis Dis 2011; 1822:753-83. [PMID: 22108204 DOI: 10.1016/j.bbadis.2011.11.002] [Citation(s) in RCA: 293] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 10/31/2011] [Accepted: 11/01/2011] [Indexed: 12/30/2022]
Abstract
Modulation of endogenous cellular defense mechanisms represents an innovative approach to therapeutic intervention in diseases causing chronic tissue damage, such as in neurodegeneration. This paper introduces the emerging role of exogenous molecules in hormetic-based neuroprotection and the mitochondrial redox signaling concept of hormesis and its applications to the field of neuroprotection and longevity. Maintenance of optimal long-term health conditions is accomplished by a complex network of longevity assurance processes that are controlled by vitagenes, a group of genes involved in preserving cellular homeostasis during stressful conditions. Vitagenes encode for heat shock proteins (Hsp) Hsp32, Hsp70, the thioredoxin and the sirtuin protein systems. Dietary antioxidants, such as polyphenols and L-carnitine/acetyl-L-carnitine, have recently been demonstrated to be neuroprotective through the activation of hormetic pathways, including vitagenes. Hormesis provides the central underpinning of neuroprotective responses, providing a framework for explaining the common quantitative features of their dose response relationships, their mechanistic foundations, their relationship to the concept of biological plasticity as well as providing a key insight for improving the accuracy of the therapeutic dose of pharmaceutical agents within the highly heterogeneous human population. This paper describes in mechanistic detail how hormetic dose responses are mediated for endogenous cellular defense pathways including sirtuin, Nrfs and related pathways that integrate adaptive stress responses in the prevention of neurodegenerative diseases. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.
Collapse
|
45
|
Calabrese V, Cornelius C, Cuzzocrea S, Iavicoli I, Rizzarelli E, Calabrese EJ. Hormesis, cellular stress response and vitagenes as critical determinants in aging and longevity. Mol Aspects Med 2011; 32:279-304. [PMID: 22020114 DOI: 10.1016/j.mam.2011.10.007] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 10/11/2011] [Indexed: 12/14/2022]
Abstract
Understanding mechanisms of aging and determinants of life span will help to reduce age-related morbidity and facilitate healthy aging. Average lifespan has increased over the last centuries, as a consequence of medical and environmental factors, but maximal life span remains unchanged. Extension of maximal life span is currently possible in animal models with measures such as genetic manipulations and caloric restriction (CR). CR appears to prolong life by reducing reactive oxygen species (ROS)-mediated oxidative damage. But ROS formation, which is positively implicated in cellular stress response mechanisms, is a highly regulated process controlled by a complex network of intracellular signaling pathways. By sensing the intracellular nutrient and energy status, the functional state of mitochondria, and the concentration of ROS produced in mitochondria, the longevity network regulates life span across species by co-ordinating information flow along its convergent, divergent and multiply branched signaling pathways, including vitagenes which are genes involved in preserving cellular homeostasis during stressful conditions. Vitagenes encode for heat shock proteins (Hsp) Hsp32, Hsp70, the thioredoxin and the sirtuin protein systems. Dietary antioxidants, such as carnosine, carnitines or polyphenols, have recently been demonstrated to be neuroprotective through the activation of hormetic pathways, including vitagenes. The hormetic dose-response, challenges long-standing beliefs about the nature of the dose-response in a lowdose zone, having the potential to affect significantly the design of pre-clinical studies and clinical trials as well as strategies for optimal patient dosing in the treatment of numerous diseases. Given the broad cytoprotective properties of the heat shock response there is now strong interest in discovering and developing pharmacological agents capable of inducing stress responses. In this review we discuss the most current and up to date understanding of the possible signaling mechanisms by which caloric restriction, as well hormetic caloric restriction-mimetics compounds by activating vitagenes can enhance defensive systems involved in bioenergetic and stress resistance homeostasis with consequent impact on longevity processes.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Chemistry, University of Catania, Viale Andrea Doria, 95100 Catania, Italy.
| | | | | | | | | | | |
Collapse
|
46
|
Halilovic A, Patil KA, Bellner L, Marrazzo G, Castellano K, Cullaro G, Dunn MW, Schwartzman ML. Knockdown of heme oxygenase-2 impairs corneal epithelial cell wound healing. J Cell Physiol 2011; 226:1732-40. [PMID: 21506105 PMCID: PMC3078528 DOI: 10.1002/jcp.22502] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Heme oxygenase (HO) represents an intrinsic cytoprotective system based on its anti-oxidative and anti-inflammatory properties mediated via its products biliverdin/bilirubin and carbon monoxide (CO). We showed that deletion of HO-2 results in impaired corneal wound healing with associated chronic inflammatory complications. This study was undertaken to examine the role of HO activity and the contribution of HO-1 and HO-2 to corneal wound healing in an in vitro epithelial scratch injury model. A scratch wound model was established using human corneal epithelial (HCE) cells. These cells expressed both HO-1 and HO-2 proteins. Injury elicited a rapid and transient increase in HO-1 and HO activity; HO-2 expression was unchanged. Treatment with biliverdin or CORM-A1, a CO donor, accelerated wound closure by 10% at 24 h. Inhibition of HO activity impaired wound closure by more than 50%. However, addition of biliverdin or CORM-A1 reversed the effect of HO inhibition on wound healing. Moreover, knockdown of HO-2 expression, but not HO-1, significantly impaired wound healing. These results indicate that HO activity is required for corneal epithelial cell migration. Inhibition of HO activity impairs wound healing while amplification of its activity restores and accelerates healing. Importantly, HO-2, which is highly expressed in the corneal epithelium, appears to be critical for the wound healing process in the cornea. The mechanisms by which it contributes to cell migration in response to injury may reside in the cytoprotective properties of CO and biliverdin.
Collapse
Affiliation(s)
| | | | - Lars Bellner
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | | | | | - Giuseppe Cullaro
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | - Michael W. Dunn
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | | |
Collapse
|
47
|
Heme Oxygenase-1 Promoter Polymorphism is a Predictor of Disease Relapse in Pancreatic Neuroendocrine Tumors. J Surg Res 2011; 166:e121-7. [DOI: 10.1016/j.jss.2010.11.898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 10/28/2010] [Accepted: 11/16/2010] [Indexed: 11/23/2022]
|
48
|
Heme regulates B-cell differentiation, antibody class switch, and heme oxygenase-1 expression in B cells as a ligand of Bach2. Blood 2011; 117:5438-48. [PMID: 21444915 DOI: 10.1182/blood-2010-07-296483] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Heme binds to proteins to modulate their function, thereby functioning as a signaling molecule in a variety of biologic events. We found that heme bound to Bach2, a transcription factor essential for humoral immunity, including antibody class switch. Heme inhibited the DNA binding activity of Bach2 in vitro and reduced its half-life in B cells. When added to B-cell primary cultures, heme enhanced the transcription of Blimp-1, the master regulator of plasma cells, and skewed plasma cell differentiation toward the IgM isotype, decreasing the IgG levels in vitro. Intraperitoneal injection of heme in mice inhibited the production of antigen-specific IgM when heme was administered simultaneously with the antigen but not when it was administered after antigen exposure, suggesting that heme also modulates the early phase of B-cell responses to antigen. Heme oxygenase-1, which is known to be regulated by heme, was repressed by both Bach2 and Bach1 in B cells. Furthermore, the expression of genes for heme uptake changed in response to B-cell activation and heme administration. Our results reveal a new function for heme as a ligand of Bach2 and as a modulatory signal involved in plasma cell differentiation.
Collapse
|
49
|
Correia MA, Sinclair PR, De Matteis F. Cytochrome P450 regulation: the interplay between its heme and apoprotein moieties in synthesis, assembly, repair, and disposal. Drug Metab Rev 2011; 43:1-26. [PMID: 20860521 PMCID: PMC3034403 DOI: 10.3109/03602532.2010.515222] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Heme is vital to our aerobic universe. Heme cellular content is finely tuned through an exquisite control of synthesis and degradation. Heme deficiency is deleterious to cells, whereas excess heme is toxic. Most of the cellular heme serves as the prosthetic moiety of functionally diverse hemoproteins, including cytochromes P450 (P450s). In the liver, P450s are its major consumers, with >50% of hepatic heme committed to their synthesis. Prosthetic heme is the sine qua non of P450 catalytic biotransformation of both endo- and xenobiotics. This well-recognized functional role notwithstanding, heme also regulates P450 protein synthesis, assembly, repair, and disposal. These less well-appreciated aspects are reviewed herein.
Collapse
Affiliation(s)
- Maria Almira Correia
- Department of Cellular and Molecular Pharmacology, The Liver Center, University of California, San Francisco, 94158, USA.
| | | | | |
Collapse
|
50
|
Kim YM, Pae HO, Park JE, Lee YC, Woo JM, Kim NH, Choi YK, Lee BS, Kim SR, Chung HT. Heme oxygenase in the regulation of vascular biology: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2011; 14:137-67. [PMID: 20624029 PMCID: PMC2988629 DOI: 10.1089/ars.2010.3153] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heme oxygenases (HOs) are the rate-limiting enzymes in the catabolism of heme into biliverdin, free iron, and carbon monoxide. Two genetically distinct isoforms of HO have been characterized: an inducible form, HO-1, and a constitutively expressed form, HO-2. HO-1 is a kind of stress protein, and thus regarded as a sensitive and reliable indicator of cellular oxidative stress. The HO system acts as potent antioxidants, protects endothelial cells from apoptosis, is involved in regulating vascular tone, attenuates inflammatory response in the vessel wall, and participates in angiogenesis and vasculogenesis. Endothelial integrity and activity are thought to occupy the central position in the pathogenesis of cardiovascular diseases. Cardiovascular disease risk conditions converge in the contribution to oxidative stress. The oxidative stress leads to endothelial and vascular smooth muscle cell dysfunction with increases in vessel tone, cell growth, and gene expression that create a pro-thrombotic/pro-inflammatory environment. Subsequent formation, progression, and obstruction of atherosclerotic plaque may result in myocardial infarction, stroke, and cardiovascular death. This background provides the rationale for exploring the potential therapeutic role for HO system in the amelioration of vascular inflammation and prevention of adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Young-Myeong Kim
- Vascular System Research Center and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Kangwon-do, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|