1
|
Raqib R, Akhtar E, Ahsanul Haq M, Ahmed S, Haque F, Chowdhury MAH, Shahriar MH, Begum BA, Eunus M, Sarwar G, Parvez F, Sharker Y, Ahsan H, Yunus M. Reduction of household air pollution through clean fuel intervention and recovery of cellular immune balance. ENVIRONMENT INTERNATIONAL 2023; 179:108137. [PMID: 37579572 PMCID: PMC11062205 DOI: 10.1016/j.envint.2023.108137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND We conducted a clean fuel intervention trial (Bangladesh Global Environmental and Occupational Health (GEOHealth) (NCT02824237) with liquefied petroleum gas (LPG) for 26 months among rural Bangladeshi women chronically exposed to household air pollution (HAP) from biomass fuel (BMF) use. We aimed to evaluate the effect of HAP reduction following LPG intervention on immune response outcome. METHODS We supplied LPG cook stove and refills in cylinder in 200 households for 26 months. We measured personal exposure to HAP [particulate matter 2.5 (PM2·5), black carbon (BC) and carbon monoxide (CO)] in 200 women (main cook) by personal monitors at pre- and post-intervention. Immune function was assessed before and after intervention, in blood collected within 2 weeks of HAP measurements. Primary endpoints included reduction in HAP, lymphocyte proliferation and oxidative stress response, and alterations in T and B cell proportions. FINDINGS Exclusive LPG use for 26 months resulted in significant reduction in PM2·5 (43.5%), BC (13%) and CO (48%) exposure in the women. For one unit decrease in BC, Treg cells and memory B cells increased by 7% and 34% respectively, in the peripheral circulation. One unit decrease in CO was significantly associated with increase in early B cells and plasmablasts by 66% and 5% respectively. For one unit decrease in BC, percent-dividing cells, proliferation and expansion indices increased by 2%, 0.4%, and 1%, respectively. INTERPRETATION Reduced personal exposure to HAP through clean fuel intervention was related to a return towards cellular immune balance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mahbub Eunus
- U-Chicago Research Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - Golam Sarwar
- U-Chicago Research Bangladesh, Mohakhali, Dhaka, Bangladesh
| | - Faruque Parvez
- Mailman School of Public Health, Columbia University, New York, USA
| | | | - Habibul Ahsan
- Department of Public Health Sciences, University of Chicago, Chicago, USA; U-Chicago Research Bangladesh, Mohakhali, Dhaka, Bangladesh
| | | |
Collapse
|
2
|
Athanassiadou V, Plavoukou S, Grapsa E, Detsika MG. The Role of Heme Oxygenase-1 as an Immunomodulator in Kidney Disease. Antioxidants (Basel) 2022; 11:antiox11122454. [PMID: 36552662 PMCID: PMC9774641 DOI: 10.3390/antiox11122454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
The protein heme oxygenase (HO)-1 has been implicated in the regulations of multiple immunological processes. It is well known that kidney injury is affected by immune mechanisms and that various kidney-disease forms may be a result of autoimmune disease. The current study describes in detail the role of HO-1 in kidney disease and provides the most recent observations of the effect of HO-1 on immune pathways and responses both in animal models of immune-mediated disease forms and in patient studies.
Collapse
Affiliation(s)
- Virginia Athanassiadou
- Department of Nephrology, School of Medicine, National and Kapodistrian University of Athens, Aretaieion University Hospital, 11528 Athens, Greece
| | - Stella Plavoukou
- Department of Nephrology, School of Medicine, National and Kapodistrian University of Athens, Aretaieion University Hospital, 11528 Athens, Greece
| | - Eirini Grapsa
- Department of Nephrology, School of Medicine, National and Kapodistrian University of Athens, Aretaieion University Hospital, 11528 Athens, Greece
| | - Maria G. Detsika
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, Evangelismos Hospital, National and Kapodistrian University of Athens, 10675 Athens, Greece
- Correspondence:
| |
Collapse
|
3
|
Li Y, Ma K, Han Z, Chi M, Sai X, Zhu P, Ding Z, Song L, Liu C. Immunomodulatory Effects of Heme Oxygenase-1 in Kidney Disease. Front Med (Lausanne) 2021; 8:708453. [PMID: 34504854 PMCID: PMC8421649 DOI: 10.3389/fmed.2021.708453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/31/2021] [Indexed: 01/23/2023] Open
Abstract
Kidney disease is a general term for heterogeneous damage that affects the function and the structure of the kidneys. The rising incidence of kidney diseases represents a considerable burden on the healthcare system, so the development of new drugs and the identification of novel therapeutic targets are urgently needed. The pathophysiology of kidney diseases is complex and involves multiple processes, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Heme oxygenase-1 (HO-1), an enzyme involved in the process of heme degradation, has attracted widespread attention in recent years due to its cytoprotective properties. As an enzyme with known anti-oxidative functions, HO-1 plays an indispensable role in the regulation of oxidative stress and is involved in the pathogenesis of several kidney diseases. Moreover, current studies have revealed that HO-1 can affect cell proliferation, cell maturation, and other metabolic processes, thereby altering the function of immune cells. Many strategies, such as the administration of HO-1-overexpressing macrophages, use of phytochemicals, and carbon monoxide-based therapies, have been developed to target HO-1 in a variety of nephropathological animal models, indicating that HO-1 is a promising protein for the treatment of kidney diseases. Here, we briefly review the effects of HO-1 induction on specific immune cell populations with the aim of exploring the potential therapeutic roles of HO-1 and designing HO-1-based therapeutic strategies for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Yunlong Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kuai Ma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Zhongyu Han
- School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingxuan Chi
- School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyalatu Sai
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhaolun Ding
- Department of Emergency Surgery, Shannxi Provincial People's Hospital, Xi'an, China
| | - Linjiang Song
- School of Medical and Life Sciences, Reproductive and Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chi Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Nephrology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Miquelianin Inhibits Allergic Responses in Mice by Suppressing CD4 + T Cell Proliferation. Antioxidants (Basel) 2021; 10:antiox10071120. [PMID: 34356353 PMCID: PMC8301087 DOI: 10.3390/antiox10071120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Allergic diseases, including atopic dermatitis (AD), induce type 2 helper T (Th2) cell-dominant immune responses. Miquelianin (quercetin 3-O-glucuronide, MQL) is an active compound in Rosae multiflorae fructus extract with anti-allergic properties. Here, we investigate the anti-allergic effects of MQL in an ovalbumin (OVA)-induced Th2-dominant mouse model and the associated mechanisms. Oral MQL suppressed cytokine and IL-2 production and proliferation of Th2 cells and upregulated heme oxygenase-1 (HO-1) in splenocytes. Ex vivo MQL suppressed Th1- and Th2-related immune responses by inhibiting CD4+ T cell proliferation, and upregulated HO-1 in CD4+ T cells by activating C-Raf-ERK1/2-Nrf2 pathway via induction of reactive oxygen species generation. In a trimellitic anhydride-induced AD-like mouse model, both topical and oral MQL ameliorated AD symptoms by suppressing Th2 immune responses. Our results suggest that MQL is a potential therapeutic agent for CD4+ T cell-mediated diseases, including allergic diseases.
Collapse
|
5
|
Cyathus striatus Extract Induces Apoptosis in Human Pancreatic Cancer Cells and Inhibits Xenograft Tumor Growth In Vivo. Cancers (Basel) 2021; 13:cancers13092017. [PMID: 33922003 PMCID: PMC8122434 DOI: 10.3390/cancers13092017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary The main aim of the present study is to test the effect of Cyathus striatus extract on the cell growth of human pancreatic cancer cells in vitro and in vivo. In addition, the effect of the extract on the gene expression was detected. The results indicated that Cyathus striatus extract significantly inhibited the cell viability and induced apoptosis. The treatment of xenograft mice harboring human pancreatic cancer cells significantly inhibited tumor growth through the induction of apoptosis. RNAseq experiments revealed the involvement of the MAPK and P53 signaling pathways and pointed toward endoplasmic reticulum stress induced apoptosis. These results may suggest that Cyathus striatus extract may contain pro-apoptotic factors that can be identified and used for the treatment of human cancer. Abstract Pancreatic cancer is a highly lethal disease with limited options for effective therapy and the lowest survival rate of all cancer forms. Therefore, a new, effective strategy for cancer treatment is in need. Previously, we found that a culture liquid extract of Cyathus striatus (CS) has a potent antitumor activity. In the present study, we aimed to investigate the effects of Cyathus striatus extract (CSE) on the growth of pancreatic cancer cells, both in vitro and in vivo. The proliferation assay (XTT), cell cycle analysis, Annexin/PI staining and TUNEL assay confirmed the inhibition of cell growth and induction of apoptosis by CSE. A Western blot analysis demonstrated the involvement of both the extrinsic and intrinsic apoptosis pathways. In addition, a RNAseq analysis revealed the involvement of the MAPK and P53 signaling pathways and pointed toward endoplasmic reticulum stress induced apoptosis. The anticancer activity of the CSE was also demonstrated in mice harboring pancreatic cancer cell line-derived tumor xenografts when CSE was given for 5 weeks by weekly IV injections. Our findings suggest that CSE could potentially be useful as a new strategy for treating pancreatic cancer.
Collapse
|
6
|
Luu Hoang KN, Anstee JE, Arnold JN. The Diverse Roles of Heme Oxygenase-1 in Tumor Progression. Front Immunol 2021; 12:658315. [PMID: 33868304 PMCID: PMC8044534 DOI: 10.3389/fimmu.2021.658315] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). HO-1 is expressed across a range of cancers and has been demonstrated to promote tumor progression through a variety of mechanisms. HO-1 can be expressed in a variety of cells within the tumor microenvironment (TME), including both the malignant tumor cells as well as stromal cell populations such as macrophages, dendritic cells and regulatory T-cells. Intrinsically to the cell, HO-1 activity provides antioxidant, anti-apoptotic and cytoprotective effects via its catabolites as well as clearing toxic intracellular heme. However, the catabolites of heme degradation can also diffuse outside of the cell to extrinsically modulate the wider TME, influencing cellular functionality and biological processes which promote tumor progression, such as facilitating angiogenesis and metastasis, as well as promoting anti-inflammation and immune suppression. Pharmacological inhibition of HO-1 has been demonstrated to be a promising therapeutic approach to promote anti-tumor immune responses and inhibit metastasis. However, these biological functions might be context, TME and cell type-dependent as there is also conflicting reports for HO-1 activity facilitating anti-tumoral processes. This review will consider our current understanding of the role of HO-1 in cancer progression and as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Kim Ngan Luu Hoang
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Joanne E Anstee
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - James N Arnold
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
7
|
Choi DW, Jung SY, Lee SY, Shon DH, Shin HS. Rosae multiflorae Fructus Extract Improves Trimellitic Anhydride-Induced Atopic Dermatitis-Like Symptoms. J Med Food 2020; 23:1287-1295. [PMID: 33185498 DOI: 10.1089/jmf.2020.4767] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Allergic disorders, including atopic dermatitis (AD), are closely linked to the activation of type 2 helper T (Th2) cells. The aim of this study was to investigate the possibility of using Rosae multiflorae fructus extract (RMFE) for AD treatment in the AD-like mouse model induced by treatment with trimellitic anhydride (TMA). Oral treatment of RMFE reduced the increase in ear thickness and suppressed inflammatory cytokine expression (interleukin [IL]-1β and tumor necrosis factor [TNF]-α) and Th2-associated immune responses (immunoglobulin [Ig] E and IL-4) in mouse ears. Furthermore, messenger RNA (mRNA) expression levels such as IL-4, IL-5, and IL-13, in draining lymph nodes were decreased by RMFE. Furthermore, we found that RMFE increased the level of heme oxygenase-1 (HO-1) through ERK and p38 pathways, reducing IL-2 production and CD4+ T cell proliferation, and inhibited STAT6 phosphorylation. Therefore, this study suggested that RMFE could be an effective treatment of AD induced by Th2-mediated immune responses by suppressing proliferation of CD4+ T cells via increased HO-1.
Collapse
Affiliation(s)
- Dae Woon Choi
- Food Biotechnology Program, KFRI School, Korea University of Science and Technology, Daejeon, Korea.,Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Korea
| | - Sun Young Jung
- Food Biotechnology Program, KFRI School, Korea University of Science and Technology, Daejeon, Korea.,Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Korea
| | - So-Young Lee
- Food Biotechnology Program, KFRI School, Korea University of Science and Technology, Daejeon, Korea.,Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Korea
| | - Dong-Hwa Shon
- Department of Food Processing and Distribution, Gangneung-Wonju National University, Gangneung, Korea
| | - Hee Soon Shin
- Food Biotechnology Program, KFRI School, Korea University of Science and Technology, Daejeon, Korea.,Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Korea
| |
Collapse
|
8
|
Yan Y, Wang L, Chen S, Zhao G, Fu C, Xu B, Tan X, Xiang Y, Chen G. Carbon Monoxide Inhibits T Cell Proliferation by Suppressing Reactive Oxygen Species Signaling. Antioxid Redox Signal 2020; 32:429-446. [PMID: 31810391 DOI: 10.1089/ars.2019.7814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aims: Carbon monoxide (CO) confers antiproliferative effects on T cells; however, how these effects are produced remains unclear. Reactive oxygen species (ROS) have recently emerged as important modulators of T cell proliferation. In this study, we aimed to determine whether the inhibitory effects of CO on T cell proliferation are dependent on the inhibition of ROS signaling. Results: Pretreatment with CO-releasing molecule-2 (CORM-2) had potent inhibitory effects on mouse T cell proliferation stimulated by anti-CD3/CD28 antibodies. Interestingly, CORM-2 pretreatment markedly suppressed intracellular ROS generation as well as the activity of NADPH oxidase and mitochondrial complexes I-IV in T cells after stimulation. The inhibitory effects of CORM-2 on both ROS production and T cell proliferation were comparable with those produced by the use of antioxidant N-acetylcysteine or a combined administration of mitochondrial complex I-IV inhibitors. Moreover, increasing intracellular ROS via hydrogen peroxide supplementation largely reversed the inhibitory effect of CORM-2 on the proliferation of T cells. The inhibitory effects of CORM-2 on both cell proliferation and intracellular ROS production were also shown in a T cell proliferation model involving stimulation by allogeneic dendritic cells or phorbol 12-myristate 13-actetate/ionomycin, as well as in spontaneous cell proliferation models in EL-4 and RAW264.7 cells. In addition, CORM-2 treatment significantly inhibited T cell activation in vivo and attenuated concanavalin A-induced autoimmune hepatitis. Innovation: CO inhibits T cell proliferation via suppression of intracellular ROS production. Conclusion: The study could supply a general mechanism to explain the inhibitory effects of CO on T cell activation and proliferation, favoring its future application in T cell-mediated diseases.
Collapse
Affiliation(s)
- Yutao Yan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lu Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Song Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Guangyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Cheng Fu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingyang Xu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaosheng Tan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Ying Xiang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
9
|
Goebel U, Wollborn J. Carbon monoxide in intensive care medicine-time to start the therapeutic application?! Intensive Care Med Exp 2020; 8:2. [PMID: 31919605 PMCID: PMC6952485 DOI: 10.1186/s40635-020-0292-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/05/2020] [Indexed: 12/18/2022] Open
Abstract
Carbon monoxide (CO) is not only known as a toxic gas due to its characteristics as an odorless molecule and its rapid binding to haem-containing molecules, thus inhibiting the respiratory chain in cells resulting in hypoxia. For decades, scientists established evidence about its endogenously production in the breakdown of haem via haem-oxygenase (HO-1) and its physiological effects. Among these, the modulation of various systems inside the body are well described (e.g., anti-inflammatory, anti-oxidative, anti-apoptotic, and anti-proliferative). Carbon monoxide is able to modulate several extra- and intra-cellular signaling molecules leading to differentiated response according to the specific stimulus. With our growing understanding in the way CO exerts its effects, especially in the mitochondria and its intracellular pathways, it is tempting to speculate about a clinical application of this substance. Since HO-1 is not easy to induce, research focused on the application of the gaseous molecule CO by itself or the implementation of carbon monoxide releasing molecules (CO-RM) to deliver the molecule at a time- and dose dependently safe way to any target organ. After years of research in cellular systems and animal models, summing up data about safety issues as well as possible target to treat in various diseases, the first feasibility trials in humans were established. Up-to-date, safety issues have been cleared for low-dose carbon monoxide inhalation (up to 500 ppm), while there is no clinical data regarding the injection or intake of any kind of CO-RM so far. Current models of human research include sepsis, acute lung injury, and acute respiratory distress syndrome as well as acute kidney injury. Carbon monoxide is a most promising candidate in terms of a therapeutic agent to improve outbalanced organ conditions. In this paper, we summarized the current understanding of carbon monoxide’s biology and its possible organ targets to treating the critically ill patients in tomorrow’s ICU.
Collapse
Affiliation(s)
- Ulrich Goebel
- Department of Anaesthesiology and Critical Care, St. Franziskus-Hospital, Hohenzollernring 70, 48145, Münster, Germany.
| | - Jakob Wollborn
- Department of Anaesthesiology and Critical Care, Medical Centre - University of Freiburg, Faculty of Medicine, Freiburg im Breisgau, Germany
| |
Collapse
|
10
|
Inhibition of Heme Oxygenase-1 Activity Enhances Wilms Tumor-1-Specific T-Cell Responses in Cancer Immunotherapy. Int J Mol Sci 2019; 20:ijms20030482. [PMID: 30678050 PMCID: PMC6387130 DOI: 10.3390/ijms20030482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023] Open
Abstract
Wilms tumor protein-1 (WT1) is an attractive target for adoptive T-cell therapy due to its expression in solid tumors and hematologic malignancies. However, T cells recognizing WT1 occur in low frequencies in the peripheral blood of healthy donors, limiting potential therapeutic possibilities. Tin mesoporphyrin (SnMP) is known to inhibit heme oxygenase-1 (HO-1), which has been shown to boost the activation and proliferation of human virus-specific T cells. We analyzed the influence of this effect on the generation of WT1-specific T cells and developed strategies for generating quantities of these cells from healthy donors, sufficient for adoptive T-cell therapies. HO-1 inhibition with SnMP increased WT1-specific T-cell frequencies in 13 (26%) of 50 healthy donors. To assess clinical applicability, we measured the enrichment efficiency of SnMP-treated WT1-specific T cells in response to a WT1-specific peptide pool and a HLA-A*02:01-restricted WT1 peptide by cytokine secretion assay. SnMP treatment resulted in a 28-fold higher enrichment efficacy with equal functionality. In conclusion, pharmacological inhibition of HO-1 activity with SnMP results in more efficient generation of functionally active WT1-specific T cells. This study demonstrates the therapeutic potentials of inhibiting HO-1 with SnMP to enhance antigen-specific T-cell responses in the treatment of cancer patients with WT1-positive disease.
Collapse
|
11
|
Naturally derived Heme-Oxygenase 1 inducers attenuate inflammatory responses in human dendritic cells and T cells: relevance for psoriasis treatment. Sci Rep 2018; 8:10287. [PMID: 29980703 PMCID: PMC6035209 DOI: 10.1038/s41598-018-28488-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/20/2018] [Indexed: 01/04/2023] Open
Abstract
Psoriasis is a chronic autoimmune disease mediated by dysregulated immune responses in dendritic cells (DC) and T cells. The stress-response enzyme heme oxygenase-1 (HO-1) has been described as protective in animal models of psoriasis, however, implementation of HO-1-based therapies is hindered by the lack of clinically-suitable HO-1 inducers. The plant-derived polyphenols, carnosol and curcumin, have been identified as candidate HO-1 inducers however there has been little investigation into their effects on human immune cells. We demonstrate that treatment of human DC with these polyphenols limits DC maturation, reduces pro-inflammatory cytokine production, and prevents induction of allospecific T cell responses, in a manner partially dependent on carbon monoxide (CO). We also characterised their effects in ex-vivo psoriasis PBMC and report that curcumin, but not carnosol, strongly reduces T cell proliferation and cytokine poly-functionality, with reduced expression of psoriatic cytokines IFNγ, IL-17, GM-CSF and IL-22. This study therefore supports reports highlighting the therapeutic potential of curcumin in psoriasis by providing insight into its immunological effects on healthy human DC and psoriasis PBMC. We also demonstrate, for the first time, the anti-inflammatory effects of carnosol in human immune cells.
Collapse
|
12
|
Pibiri M, Leoni VP, Atzori L. Heme oxygenase-1 inhibitor tin-protoporphyrin improves liver regeneration after partial hepatectomy. Life Sci 2018; 204:9-14. [PMID: 29738777 DOI: 10.1016/j.lfs.2018.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/24/2018] [Accepted: 05/04/2018] [Indexed: 12/22/2022]
Abstract
AIMS This study investigates the effects of the heme oxygenase-1 (HO-1) inhibitor tin protoporphyrin IX (SnPP), on rat liver regeneration following 2/3 partial hepatectomy (PH) in order to clarify the controversial role of HO-1 in the regulation of cellular growth. MAIN METHODS Male Wistar rats received a subcutaneous injection of either SnPP (10 μmoles/kg body weight) or saline 12 h before PH and 0, 12 and 24 h after surgery. Rats were killed from 0.5 to 36 h after PH. Bromodeoxyuridine (BrdU) incorporation was used to analyze cell proliferation. Immunohistochemistry, Western blot analysis and quantitative Real Time-PCR were used to assess molecular and cellular changes after PH. KEY FINDINGS Data obtained have shown that administration of SnPP caused an increased entry of hepatocytes into S phase after PH, as demonstrated by labeling (L.I.) and mitotic (M.I.) indexes. Furthermore, enhanced cell cycle entry in PH-animals pre-treated with SnPP was associated with an earlier activation of IL-6 and transcription factors involved in liver regeneration, such as phospho-JNK and phospho-STAT3. SIGNIFICANCE Summarizing, data here reported demonstrate that inhibition of HO-1 enhances rat liver regeneration after PH which is associated to a very rapid increase in the levels of inflammatory mediators such as IL-6, phopsho-JNK and phospho-STAT3, suggesting that HO-1 could act as a negative modulator of liver regeneration. Knowledge about the mechanisms of liver regeneration can be applied to clinical problems caused by delayed liver growth, and HO-1 repression may be a mechanism by which cells can faster proliferate in response to tissue damage.
Collapse
Affiliation(s)
- Monica Pibiri
- Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Via Porcell 4, 09124 Cagliari, Italy
| | - Vera Piera Leoni
- Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Via Porcell 4, 09124 Cagliari, Italy
| | - Luigi Atzori
- Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Via Porcell 4, 09124 Cagliari, Italy.
| |
Collapse
|
13
|
Abstract
Over the past century, solid organ transplantation has been improved both at a surgical and postoperative level. However, despite the improvement in efficiency, safety, and survival, we are still far from obtaining full acceptance of all kinds of allograft in the absence of concomitant treatments. Today, transplanted patients are treated with immunosuppressive drugs (IS) to minimize immunological response in order to prevent graft rejection. Nevertheless, the lack of specificity of IS leads to an increase in the risk of cancer and infections. At this point, cell therapies have been shown as a novel promising resource to minimize the use of IS in transplantation. The main strength of cell therapy is the opportunity to generate allograft-specific tolerance, promoting in this way long-term allograft survival. Among several other regulatory cell types, tolerogenic monocyte-derived dendritic cells (Tol-MoDCs) appear to be an interesting candidate for cell therapy due to their ability to perform specific antigen presentation and to polarize immune response to immunotolerance. In this review, we describe the characteristics and the mechanisms of action of both human Tol-MoDCs and rodent tolerogenic bone marrow-derived DCs (Tol-BMDCs). Furthermore, studies performed in transplantation models in rodents and non-human primates corroborate the potential of Tol-BMDCs for immunoregulation. In consequence, Tol-MoDCs have been recently evaluated in sundry clinical trials in autoimmune diseases and shown to be safe. In addition to autoimmune diseases clinical trials, Tol-MoDC is currently used in the first phase I/II clinical trials in transplantation. Translation of Tol-MoDCs to clinical application in transplantation will also be discussed in this review.
Collapse
Affiliation(s)
- Eros Marín
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| | - Maria Cristina Cuturi
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| | - Aurélie Moreau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
14
|
Dreyer-Andersen N, Almeida AS, Jensen P, Kamand M, Okarmus J, Rosenberg T, Friis SD, Martínez Serrano A, Blaabjerg M, Kristensen BW, Skrydstrup T, Gramsbergen JB, Vieira HLA, Meyer M. Intermittent, low dose carbon monoxide exposure enhances survival and dopaminergic differentiation of human neural stem cells. PLoS One 2018; 13:e0191207. [PMID: 29338033 PMCID: PMC5770048 DOI: 10.1371/journal.pone.0191207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/30/2017] [Indexed: 12/18/2022] Open
Abstract
Exploratory studies using human fetal tissue have suggested that intrastriatal transplantation of dopaminergic neurons may become a future treatment for patients with Parkinson's disease. However, the use of human fetal tissue is compromised by ethical, regulatory and practical concerns. Human stem cells constitute an alternative source of cells for transplantation in Parkinson's disease, but efficient protocols for controlled dopaminergic differentiation need to be developed. Short-term, low-level carbon monoxide (CO) exposure has been shown to affect signaling in several tissues, resulting in both protection and generation of reactive oxygen species. The present study investigated the effect of CO produced by a novel CO-releasing molecule on dopaminergic differentiation of human neural stem cells. Short-term exposure to 25 ppm CO at days 0 and 4 significantly increased the relative content of β-tubulin III-immunoreactive immature neurons and tyrosine hydroxylase expressing catecholaminergic neurons, as assessed 6 days after differentiation. Also the number of microtubule associated protein 2-positive mature neurons had increased significantly. Moreover, the content of apoptotic cells (Caspase3) was reduced, whereas the expression of a cell proliferation marker (Ki67) was left unchanged. Increased expression of hypoxia inducible factor-1α and production of reactive oxygen species (ROS) in cultures exposed to CO may suggest a mechanism involving mitochondrial alterations and generation of ROS. In conclusion, the present procedure using controlled, short-term CO exposure allows efficient dopaminergic differentiation of human neural stem cells at low cost and may as such be useful for derivation of cells for experimental studies and future development of donor cells for transplantation in Parkinson's disease.
Collapse
Affiliation(s)
- Nanna Dreyer-Andersen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ana Sofia Almeida
- Instituto de Biologia Experimental e Tecnológica (IBET), Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica (ITQB), Oeiras, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Pia Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Morad Kamand
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Tine Rosenberg
- Department of Pathology, Odense University Hospital, Denmark & Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Stig Düring Friis
- Center for Insoluble Protein Structures (inSPIN), Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Alberto Martínez Serrano
- Department of Molecular Biology and Center of Molecular Biology Severo Ochoa, University Autonoma Madrid-C.S.I.C Campus Cantoblanco, Madrid, Spain
| | - Morten Blaabjerg
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, Denmark & Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Troels Skrydstrup
- Center for Insoluble Protein Structures (inSPIN), Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Jan Bert Gramsbergen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Helena L. A. Vieira
- Instituto de Biologia Experimental e Tecnológica (IBET), Oeiras, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciência Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
15
|
Loboda A, Jozkowicz A, Dulak J. Carbon monoxide: pro- or anti-angiogenic agent? Comment on Ahmad et al. (Thromb Haemost 2015; 113: 329–337). Thromb Haemost 2017; 114:432-3. [DOI: 10.1160/th15-01-0082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/26/2015] [Indexed: 12/31/2022]
|
16
|
Gao M, Guo KM, Wei YM, Ma MM, Cai JR, Xia TT, Ye QJ. Aspirin inhibits the proliferation of human uterine leiomyoma cells by downregulation of K‑Ras‑p110α interaction. Oncol Rep 2017; 38:2507-2517. [PMID: 28849118 DOI: 10.3892/or.2017.5915] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/14/2017] [Indexed: 11/06/2022] Open
Abstract
Aspirin has been confirmed as an effective antitumor drug in various cancers. However, the relationship between aspirin and uterine leiomyoma is still underexplored. Here, we explored the effects of aspirin on human uterine leiomyoma cells and provide insights into the underlying mechanisms. Cell Counting Kit-8 (CCK-8) and flow cytometry analysis showed that aspirin treatment inhibited cell proliferation and promoted cell cycle arrest at G0/G1 phase in a dose- and time‑dependent manner of human uterine leiomyoma cells. Further studies revealed that aspirin blocked the interaction between K-Ras and p110α by co-immunoprecipitation and immunofluorescence. Western blotting demonstrated K‑Ras‑p110α interaction was required for the effects of aspirin‑induced inhibition on cell growth and cell cycle transition via cell cycle regulators, including cyclin D1 and cyclin-dependent kinase 2 (CDK2). PI3K/Akt/caspase signaling pathway was involved in human uterine leiomyoma cell growth under aspirin treatment. Taken together, these results suggest that aspirin inhibited human uterine leiomyoma cell growth by regulating K‑Ras‑p110α interaction. Aspirin which targeting on interaction between K-Ras and p110α may serve as a new therapeutic drug for uterine leiomyoma treatment.
Collapse
Affiliation(s)
- Min Gao
- Department of Pharmacy, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Kai-Min Guo
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Ying-Mei Wei
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Ming-Ming Ma
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jia-Rong Cai
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Ting-Ting Xia
- Department of Infertility, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Qing-Jian Ye
- Department of Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
17
|
Bolisetty S, Zarjou A, Agarwal A. Heme Oxygenase 1 as a Therapeutic Target in Acute Kidney Injury. Am J Kidney Dis 2017; 69:531-545. [PMID: 28139396 DOI: 10.1053/j.ajkd.2016.10.037] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/22/2016] [Indexed: 01/06/2023]
Abstract
A common clinical condition, acute kidney injury (AKI) significantly influences morbidity and mortality, particularly in critically ill patients. The pathophysiology of AKI is complex and involves multiple pathways, including inflammation, autophagy, cell-cycle progression, and oxidative stress. Recent evidence suggests that a single insult to the kidney significantly enhances the propensity to develop chronic kidney disease. Therefore, the generation of effective therapies against AKI is timely. In this context, the cytoprotective effects of heme oxygenase 1 (HO-1) in animal models of AKI are well documented. HO-1 modulates oxidative stress, autophagy, and inflammation and regulates the progression of cell cycle via direct and indirect mechanisms. These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). This review highlights recent advances in the molecular mechanisms of HO-1-mediated cytoprotection and discusses the translational potential of HO-1 induction in AKI.
Collapse
Affiliation(s)
- Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL; Birmingham Veterans Administration Medical Center, Birmingham, AL.
| |
Collapse
|
18
|
Hussain MS, Qureshi AI, Kirmani JF, Divani AA, Hopkins LN. Development of Vascular Biology over the past 10 Years: Heme Oxygenase-1 in Cardiovascular Homeostasis. J Endovasc Ther 2016; 11 Suppl 2:II32-42. [PMID: 15760262 DOI: 10.1177/15266028040110s616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The study of vascular biology has provided strong evidence for the role that free radical attack plays in the pathogenesis of cardiovascular diseases. The endothelial cell (EC) dysfunction that results from exposure to oxidative stresses, such as oxidized LDL, influences vascular cell gene expression, promoting smooth muscle cell (SMC) mitogenesis and apoptosis. These factors also play an important role in atherogenesis, which is attenuated by antioxidants. Thus, antioxidants are important to understanding the pathophysiology of cardiovascular diseases and to constructing an effective treatment strategy for these patients. Over the last decade, there has been a tremendous interest in the biology of heme oxygenase-1 (HO-1), which exhibits antioxidant effects in various forms of tissue injury. Moreover, the reaction is also the major source of carbon dioxide (CO) in the body, which is a physiologically important gaseous vasodilator that inhibits SMC proliferation. Thus, HO-1–derived products provide various mechanisms to maintain cardiovascular homeostasis. We review recent work on the cellular and molecular biological aspects of the HO/CO system in vascular pathophysiology.
Collapse
Affiliation(s)
- M Shazam Hussain
- Zeenat Qureshi Stroke Research Center, Department of Neurology and Neurosciences, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07101, USA
| | | | | | | | | |
Collapse
|
19
|
Bunse CE, Fortmeier V, Tischer S, Zilian E, Figueiredo C, Witte T, Blasczyk R, Immenschuh S, Eiz-Vesper B. Modulation of heme oxygenase-1 by metalloporphyrins increases anti-viral T cell responses. Clin Exp Immunol 2015; 179:265-76. [PMID: 25196646 DOI: 10.1111/cei.12451] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2014] [Indexed: 12/14/2022] Open
Abstract
Heme oxygenase (HO)-1, the inducible isoform of HO, has immunomodulatory functions and is considered a target for therapeutic interventions. In the present study, we investigated whether modulation of HO-1 might have regulatory effects on in-vitro T cell activation. The study examined whether: (i) HO-1 induction by cobalt-protoporphyrin (CoPP) or inhibition by tin-mesoporphyrin (SnMP) can affect expansion and function of virus-specific T cells, (ii) HO-1 modulation might have a functional effect on other cell populations mediating effects on proliferating T cells [e.g. dendritic cells (DCs), regulatory T cells (T(regs)) and natural killer cells] and (iii) HO-1-modulated anti-viral T cells might be suitable for adoptive immunotherapy. Inhibition of HO-1 via SnMP in cytomegalovirus (CMV)pp65-peptide-pulsed peripheral blood mononuclear cells (PBMCs) led to increased anti-viral T cell activation and the generation of a higher proportion of effector memory T cells (CD45RA(-) CD62L(-)) with increased capability to secrete interferon (IFN)-γ and granzyme B. T(reg) depletion and SnMP exposure increased the number of anti-viral T cells 15-fold. To test the possibility that HO-1 modulation might be clinically applicable in conformity with good manufacturing practice (GMP), SnMP was tested in isolated anti-viral T cells using the cytokine secretion assay. Compared to control, SnMP treatment resulted in higher cell counts and purity without negative impact on quality and effector function [CD107a, IFN-γ and tumour necrosis factor (TNF)-α levels were stable]. These results suggest an important role of HO-1 in the modulation of adaptive immune responses. HO-1 inhibition resulted in markedly more effective generation of functionally active T cells suitable for adoptive T cell therapy.
Collapse
Affiliation(s)
- C E Bunse
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany; Integrated Research and Treatment Centre Transplantation - IFB-Tx, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhao H, Ozen M, Wong RJ, Stevenson DK. Heme oxygenase-1 in pregnancy and cancer: similarities in cellular invasion, cytoprotection, angiogenesis, and immunomodulation. Front Pharmacol 2015; 5:295. [PMID: 25642189 PMCID: PMC4294126 DOI: 10.3389/fphar.2014.00295] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/18/2014] [Indexed: 01/28/2023] Open
Abstract
Pregnancy can be defined as a “permissible” process, where a semi-allogeneic fetus and placenta are allowed to grow and survive within the mother. Similarly, in tumor growth, antigen-specific malignant cells proliferate and evade into normal tissues of the host. The microenvironments of the placenta and tumors are amazingly comparable, sharing similar mechanisms exploited by fetal or cancer cells with regard to surviving in a hypoxic microenvironment, invading tissues via degradation and vasculogenesis, and escaping host attack through immune privilege. Heme oxygease-1 (HO-1) is a stress-response protein that has antioxidative, anti-apoptotic, pro-angiogenic, and anti-inflammatory properties. Although a large volume of research has been published in recent years investigating the possible role(s) of HO-1 in pregnancy and in cancer development, the molecular mechanisms that regulate these “yin-yang” processes have still not been fully elucidated. Here, we summarize and compare pregnancy and cancer development, focusing primarily on the function of HO-1 in cellular invasion, cytoprotection, angiogenesis, and immunomodulation. Due to the similarities of both processes, a thorough understanding of the molecular mechanisms of each process may reveal and guide the development of new approaches to prevent not only pregnancy disorders; but also, to study cancer.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| | - Maide Ozen
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| | - Ronald J Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine Stanford, CA, USA
| |
Collapse
|
21
|
Nikolic I, Vujicic M, Stojanovic I, Stosic-Grujicic S, Saksida T. Carbon Monoxide-Releasing Molecule-A1 Inhibits Th1/Th17 and Stimulates Th2 DifferentiationIn vitro. Scand J Immunol 2014; 80:95-100. [DOI: 10.1111/sji.12189] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/21/2014] [Indexed: 12/29/2022]
Affiliation(s)
- I. Nikolic
- Institute for Biological Research ‘Sinisa Stankovic’; University of Belgrade; Belgrade Serbia
| | - M. Vujicic
- Institute for Biological Research ‘Sinisa Stankovic’; University of Belgrade; Belgrade Serbia
| | - I. Stojanovic
- Institute for Biological Research ‘Sinisa Stankovic’; University of Belgrade; Belgrade Serbia
| | - S. Stosic-Grujicic
- Institute for Biological Research ‘Sinisa Stankovic’; University of Belgrade; Belgrade Serbia
| | - T. Saksida
- Institute for Biological Research ‘Sinisa Stankovic’; University of Belgrade; Belgrade Serbia
| |
Collapse
|
22
|
Zhong H, Bao W, Friedman D, Yazdanbakhsh K. Hemin controls T cell polarization in sickle cell alloimmunization. THE JOURNAL OF IMMUNOLOGY 2014; 193:102-10. [PMID: 24879794 DOI: 10.4049/jimmunol.1400105] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Patients with sickle cell disease (SCD) often require transfusions to treat and prevent worsening anemia and other SCD complications. However, transfusions can trigger alloimmunization against transfused RBCs with serious clinical sequelae. Risk factors for alloimmunization in SCD remain poorly understood. We recently reported altered regulatory T cell (Treg) and Th responses with higher circulating Th1 (IFN-γ(+)) cytokines in chronically transfused SCD patients with alloantibodies as compared with those without alloantibodies. Because monocytes play a critical role in polarization of T cell subsets and participate in clearance of transfused RBCs, we tested the hypothesis that in response to the RBC breakdown product hemin, monocyte control of T cell polarization will differ between alloimmunized and non-alloimmunized SCD patients. Exogenous hemin induced Treg polarization in purified T cell/monocyte cocultures from healthy volunteers through the monocyte anti-inflammatory heme-degrading enzyme heme oxygenase-1. Importantly, hemin primarily through its effect on CD16+ monocytes induced an anti-inflammatory (higher Treg/lower Th1) polarization state in the non-alloimmunized SCD group, whereas it had little effect in the alloimmunized group. Non-alloimmunized SCD CD16+ monocytes expressed higher basal levels of heme oxygenase-1. Furthermore, IL-12, which contributed to a proinflammatory polarization state (low Treg/high Th1) in SCD, was dampened in hemin-treated stimulated monocytes from non-alloimmunized SCD patients, but not in the alloimmunized group. These data suggest that unlike alloimmunized patients, non-alloimmunized SCD CD16+ monocytes in response to transfused RBC breakdown products promote an anti-inflammatory state that is less conducive to alloimmunization.
Collapse
Affiliation(s)
- Hui Zhong
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065
| | - Weili Bao
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065
| | - David Friedman
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Karina Yazdanbakhsh
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065;
| |
Collapse
|
23
|
Hull TD, Agarwal A, George JF. The mononuclear phagocyte system in homeostasis and disease: a role for heme oxygenase-1. Antioxid Redox Signal 2014; 20:1770-88. [PMID: 24147608 PMCID: PMC3961794 DOI: 10.1089/ars.2013.5673] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 10/22/2013] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Heme oxygenase-1 (HO-1) is a potential therapeutic target in many diseases, especially those mediated by oxidative stress and inflammation. HO-1 expression appears to regulate the homeostatic activity and distribution of mononuclear phagocytes (MP) in lymphoid tissue under physiological conditions. It also regulates the ability of MP to modulate the inflammatory response to tissue injury. RECENT ADVANCES The induction of HO-1 within MP-particularly macrophages and dendritic cells-modulates the effector functions that they acquire after activation. These effector functions include cytokine production, surface receptor expression, maturation state, and polarization toward a pro- or anti-inflammatory phenotype. The importance of HO-1 in MP is emphasized by their expression of specific receptors that primarily function to ingest heme-containing substrate and deliver it to HO-1. CRITICAL ISSUES MP are the first immunological responders to tissue damage. They critically affect the outcome of injury to many organ systems, yet few therapies are currently available to specifically target MP during disease pathogenesis. Elucidation of the role of HO-1 expression in MP may help to direct broadly applicable therapies to clinical use that are based on the immunomodulatory capabilities of HO-1. FUTURE DIRECTIONS Unraveling the complexities of HO-1 expression specifically within MP will more completely define how HO-1 provides cytoprotection in vivo. The use of models in which HO-1 expression is specifically modulated in bone marrow-derived cells will allow for a more complete characterization of its immunoregulatory properties.
Collapse
Affiliation(s)
- Travis D. Hull
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| | - James F. George
- Division of Cardiothoracic Surgery, Department of Surgery, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
24
|
Gonzales MA, Mascharak PK. Photoactive metal carbonyl complexes as potential agents for targeted CO delivery. J Inorg Biochem 2014; 133:127-35. [DOI: 10.1016/j.jinorgbio.2013.10.015] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/18/2013] [Accepted: 10/19/2013] [Indexed: 01/06/2023]
|
25
|
Abid S, Houssaïni A, Mouraret N, Marcos E, Amsellem V, Wan F, Dubois-Randé JL, Derumeaux G, Boczkowski J, Motterlini R, Adnot S. P21-dependent protective effects of a carbon monoxide-releasing molecule-3 in pulmonary hypertension. Arterioscler Thromb Vasc Biol 2013; 34:304-12. [PMID: 24334871 DOI: 10.1161/atvbaha.113.302302] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Carbon monoxide-releasing molecules (CORMs) represent a pharmacological alternative to CO gas inhalation. Here, we questioned whether CORM-3, a well-characterized water-soluble CORM, could prevent and reverse pulmonary hypertension (PH) in chronically hypoxic mice and in smooth muscle promoter 22 serotonin transporter mice overexpressing the serotonin transporter in smooth muscle cells (SMCs). APPROACH AND RESULTS Treatment with CORM-3 (50 mg/kg per day once daily) for 3 weeks prevented PH, right ventricular hypertrophy, and distal pulmonary artery muscularization in mice exposed to chronic hypoxia and partially reversed PH in smooth muscle promoter 22 serotonin transporter mice by reducing Ki67 dividing pulmonary artery SMCs (PA-SMCs). In these models, CORM-3 markedly increased lung p21 mRNA and protein levels and p21-stained PA-SMCs. These effects contrasted with the transient pulmonary vasodilatation and rise in lung cGMP levels induced by a single injection of CORM-3 in mice exposed to acute hypoxia. Studies in cultured rat PA-SMCs revealed that the inhibitory effects of CORM-3 on cell growth were independent of cGMP formation but associated with increased p21 mRNA and protein levels. Protection against PH by CORM-3 required increased lung expression of p21, as indicated by the inability of CORM-3 to prevent chronic hypoxia-induced PH in p21-deficient mice and to alter the growth of PA-SMCs derived from p21-deficient mice. CORM-3-induced p21 overexpression was linked to p53 activation as assessed by the inability of CORM-3 to prevent PH and induce p21 expression in p53-deficient mice and in PA-SMCs derived from p53-deficient mice. CONCLUSIONS CORM-3 inhibits pulmonary vascular remodeling via p21, which may represent a useful approach for treating PH.
Collapse
Affiliation(s)
- Shariq Abid
- From the INSERM U955, Département de Physiologie (S.A., A.H., N.M., E.M., V.A., F.W., G.D., J.B., R.M., S.A.) and Service de Cardiologie (J.L.D.-R.), Hôpital Henri Mondor, AP-HP, Université Paris-Est Creteil (UPEC), Créteil, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wegiel B, Gallo D, Csizmadia E, Harris C, Belcher J, Vercellotti GM, Penacho N, Seth P, Sukhatme V, Ahmed A, Pandolfi PP, Helczynski L, Bjartell A, Persson JL, Otterbein LE. Carbon monoxide expedites metabolic exhaustion to inhibit tumor growth. Cancer Res 2013; 73:7009-21. [PMID: 24121491 DOI: 10.1158/0008-5472.can-13-1075] [Citation(s) in RCA: 270] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One classical feature of cancer cells is their metabolic acquisition of a highly glycolytic phenotype. Carbon monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, has been implicated in carcinogenesis and therapeutic resistance. However, the functional contributions of CO and HO-1 to these processes are poorly defined. In human prostate cancers, we found that HO-1 was nuclear localized in malignant cells, with low enzymatic activity in moderately differentiated tumors correlating with relatively worse clinical outcomes. Exposure to CO sensitized prostate cancer cells but not normal cells to chemotherapy, with growth arrest and apoptosis induced in vivo in part through mitotic catastrophe. CO targeted mitochondria activity in cancer cells as evidenced by higher oxygen consumption, free radical generation, and mitochondrial collapse. Collectively, our findings indicated that CO transiently induces an anti-Warburg effect by rapidly fueling cancer cell bioenergetics, ultimately resulting in metabolic exhaustion.
Collapse
Affiliation(s)
- Barbara Wegiel
- Authors' Affiliations: Department of Surgery, Transplant Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; Department of Clinical Sciences, Section of Urological Cancers, School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, United Kingdom; Department of Laboratory Medicine, University Hospital Malmö, Lund University, Malmö, Sweden; and Department of Medicine and Vascular Biology Center, University of Minnesota, Minneapolis, Alfama Inc., Oeiras, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhang Y, Zhang L, Wu J, Di C, Xia Z. Heme oxygenase-1 exerts a protective role in ovalbumin-induced neutrophilic airway inflammation by inhibiting Th17 cell-mediated immune response. J Biol Chem 2013; 288:34612-26. [PMID: 24097973 DOI: 10.1074/jbc.m113.494369] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Allergic asthma is conventionally considered as a Th2 immune response characterized by eosinophilic inflammation. Recent investigations revealed that Th17 cells play an important role in the pathogenesis of non-eosinophilic asthma (NEA), resulting in steroid-resistant neutrophilic airway inflammation. Heme oxygenase-1 (HO-1) has anti-inflammation, anti-oxidation, and anti-apoptosis functions. However, its role in NEA is still unclear. Here, we explore the role of HO-1 in a mouse model of NEA. HO-1 inducer hemin or HO-1 inhibitor tin protoporphyrin IX was injected intraperitoneally into ovalbumin-challenged DO11.10 mice. Small interfering RNA (siRNA) was delivered into mice to knock down HO-1 expression. The results show that induction of HO-1 by hemin attenuated airway inflammation and decreased neutrophil infiltration in bronchial alveolar lavage fluid and was accompanied by a lower proportion of Th17 cells in mediastinal lymph nodes and spleen. More importantly, induction of HO-1 down-regulated Th17-related transcription factor retinoic acid-related orphan receptor γt (RORγt) expression and decreased IL-17A levels, all of which correlated with a decrease in phosphorylated STAT3 (p-STAT3) level and inhibition of Th17 cell differentiation. Consistently, the above events could be reversed by tin protoporphyrin IX. Also, HO-1 siRNA transfection abolished the effect of hemin induced HO-1 in vivo. Meanwhile, the hemin treatment promoted the level of Foxp3 expression and enhanced the proportion of regulatory T cells (Tregs). Collectively, our findings indicate that HO-1 exhibits anti-inflammatory activity in the mouse model of NEA via inhibition of the p-STAT3-RORγt pathway, regulating kinetics of RORγt and Foxp3 expression, thus providing a possible novel therapeutic target in asthmatic patients.
Collapse
Affiliation(s)
- Yanjie Zhang
- From the Department of Pediatrics, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | |
Collapse
|
28
|
Bonnamain V, Mathieux E, Thinard R, Thébault P, Nerrière-Daguin V, Lévêque X, Anegon I, Vanhove B, Neveu I, Naveilhan P. Expression of heme oxygenase-1 in neural stem/progenitor cells as a potential mechanism to evade host immune response. Stem Cells 2013; 30:2342-53. [PMID: 22888011 DOI: 10.1002/stem.1199] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Besides their therapeutic benefit as cell source, neural stem/progenitor cells (NSPCs) exhibit immunosuppressive properties of great interest for modulating immune response in the central nervous system. To decipher the mechanisms of NSPC-mediated immunosuppression, activated T cells were exposed to NSPCs isolated from fetal rat brains. Analyses revealed that NSPCs inhibited T-cell proliferation and interferon-gamma production in a dose-dependent manner. A higher proportion of helper T cells (CD4+ T cells) was found in the presence of NSPCs, but analyses of FoxP3 population indicated that T-cell suppression was not secondary to an induction of suppressive regulatory T cells (FoxP3+ CD4+ CD25+). Conversely, induction of the high affinity interleukin-2 (IL-2) receptor (CD25) and the inability of IL-2 to rescue T-cell proliferation suggest that NSPCs display immunosuppressive activity without affecting T-cell activation. Cultures in Transwell chambers or addition of NSPC-conditioned medium to activated T cells indicated that part of the suppressive activity was not contact dependent. We therefore searched for soluble factors that mediate NSPC immunosuppression. We found that NSPCs express several immunosuppressive molecules, but the ability of these cells to inhibit T-cell proliferation was only counteracted by heme oxygenase (HO) inhibitors in association or not with nitric oxide synthase inhibitors. Taken together, our findings highlight a dynamic crosstalk between NSPCs and T lymphocytes and provide the first evidence of an implication of HO-1 in mediating the immunosuppressive effects of the NSPCs.
Collapse
|
29
|
Sener A, Tran KC, Deng JP, Garcia B, Lan Z, Liu W, Sun T, Arp J, Salna M, Acott P, Cepinskas G, Jevnikar AM, Luke PPW. Carbon monoxide releasing molecules inhibit cell death resulting from renal transplantation related stress. J Urol 2012; 190:772-8. [PMID: 23246477 DOI: 10.1016/j.juro.2012.12.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2012] [Indexed: 11/15/2022]
Abstract
PURPOSE Organ cold storage and subsequent transplantation are associated with significant ischemia-reperfusion injury, leading to cell death, graft inflammation and decreased graft function. MATERIALS AND METHODS CORM-3s reduce oxidative stress and prevent inflammation in kidneys stored at 4C and subsequently transplanted. Graft survival and function are markedly improved compared to kidneys preserved and stored in University of Wisconsin solution alone. We determined whether CORM-3 has direct antiapoptotic effects on in vitro preparations of human HUVECs exposed to anoxic conditions. We also determined whether direct administration of CORM-3 to renal grafts before and/or after cold storage would prevent renal damage during the transplantation process. RESULTS CORM-3 supplementation led to a significantly increased frequency of live cells (mean ± SD 72.3% ± 1.9%, p <0.01), reduced apoptosis (14.9% ± 6.1%, p <0.01) and decreased mitochondrial transmembrane potential (40.2% ± 7.2%, p <0.05) in HUVECs exposed to 20 hours of cold storage compared to controls (11.6% ± 3.5%, 82.2% ± 2.3% and 78.2% ± 3.2%, respectively). In keeping with this antiapoptotic effect CORM-3 supplementation led to a mean 7.4 ± 2.1-fold up-regulation in Bcl-2 gene expression. CORM-3 supplementation in standard preservation solution was most beneficial at initial ischemic injury and before cold storage exposure. However, additional reflushing before vascular reperfusion showed an additive benefit to graft survival and function after transplantation. This was confirmed by decreased glomerular and tubular necrosis, and apoptosis in double flushed grafts. CONCLUSIONS CORM-3 supplementation in standard University of Wisconsin solution has a significant impact on decreasing cellular and graft injury, and improving survival through its antiapoptotic effects, which are likely mediated through mitochondrial membrane stabilization.
Collapse
Affiliation(s)
- Alp Sener
- Division of Urology, Department of Surgery, London Health Sciences Center, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim HJ, Zheng M, Kim SK, Cho JJ, Shin CH, Joe Y, Chung HT. CO/HO-1 Induces NQO-1 Expression via Nrf2 Activation. Immune Netw 2011; 11:376-82. [PMID: 22346778 PMCID: PMC3275707 DOI: 10.4110/in.2011.11.6.376] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 10/21/2011] [Accepted: 11/07/2011] [Indexed: 12/15/2022] Open
Abstract
Background Carbon monoxide (CO) is a cytoprotective and homeostatic molecule with important signaling capabilities in physiological and pathophysiological situations. CO protects cells/tissues from damage by free radicals or oxidative stress. NAD(P)H:quinone oxidoreductase (NQO1) is a highly inducible enzyme that is regulated by the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway, which is central to efficient detoxification of reactive metabolites and reactive oxygen species (ROS). Methods We generated NQO1 promoter construct. HepG2 cells were treated with CO Releasing Molecules-2 (CORM-2) or CO gas and the gene expressions were measured by RT-PCR, immunoblot, and luciferase assays. Results CO induced expression of NQO1 in human hepatocarcinoma cell lines by activation of Nrf2. Exposure of HepG2 cells to CO resulted in significant induction of NQO1 in dose- and time-dependent manners. Analysis of the NQO1 promoter indicated that an antioxidant responsible element (ARE)-containing region was critical for the CO-induced Nrf2-dependent increase of NQO1 gene expression in HepG2 cells. Conclusion Our results suggest that CO-induced Nrf2 increases the expression of NQO1 which is well known to detoxify reactive metabolites and ROS.
Collapse
Affiliation(s)
- Hyo Jeong Kim
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
HO-1 (haem oxygenase-1) is a ubiquitously expressed inducible enzyme degrading haem to CO, biliverdin and Fe2+. Its activation reduces oxidative stress in cells and inhibits inflammation, both due to removal of haem and because of the biological activity of HO-1 products. CO may act similarly to NO, activating soluble guanylate cyclase and elevating cGMP production. It inhibits platelet aggregation, reduces leucocyte adhesion, decreases apoptosis and lowers the production of some pro-inflammatory cytokines. Biliverdin is converted into bilirubin by biliverdin reductase, and both compounds are potent antioxidants, free radical scavengers and inhibitors of the complement cascade. Iron ions can be potentially toxic, increasing the generation of hydroxyl radicals, but simultaneous induction of ferritin and activation of the Fe-ATPase iron transporter protects cells from oxidative stress. Importantly, basal and induced expression of HO-1 is very variable in the human population because of the highly polymorphic (GT)n fragment in the promoter, which may have clinical relevance. The recognized roles of HO-1 are far beyond cytoprotection. The enzyme is important in the regulation of cell proliferation, differentiation and apoptosis. Its activity improves neovascularization, attenuates inflammation and modulates the immune response, thereby influencing carcinogenesis, wound healing, transplant survival and the progression of cardiovascular diseases. Recent results indicate that HO-1 may also act through the regulation of microRNAs, which suggests a much broader involvement of HO-1 in the modulation of cell functions and offers a potential explanation for some well-known activities whose mechanism has hitherto been unclear.
Collapse
|
32
|
Beneficial effects of perioperative low-dose inhaled carbon monoxide on pulmonary allograft survival in MHC-inbred CLAWN miniature swine. Transplantation 2011; 90:1336-43. [PMID: 21076382 DOI: 10.1097/tp.0b013e3181ff8730] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND We have recently reported that perioperative low-dose carbon monoxide (CO) inhalation decreases lung ischemia-reperfusion injury in miniature swine. The aims of this study were to establish a large animal model of pulmonary allograft rejection using polymerase chain reaction-typed major histocompatibility complex (MHC)-inbred CLAWN miniature swine and to examine the effects of CO on allograft survival. METHODS Eleven CLAWN miniature swines received fully MHC-mismatched lungs followed by 12 days of tacrolimus (days 0-11; blood level 35-45 ng/mL). Six recipients received tacrolimus alone (control group). Five recipients were additionally treated with inhaled CO (180 min for donors until graft harvest; 390 min for recipients until 2 hr after reperfusion). RESULTS All recipients treated with tacrolimus alone uniformly rejected their grafts by postoperative day 63 with development of cytotoxic antidonor antibodies. CO treatment was effective in prolonging allograft survival from a mean of 47±7 to 82±13 days (P=0.017), with one CO-treated animal maintaining function until postoperative day 120. Development of antidonor antibodies and donor-specific responsiveness by cell-mediated lympholysis and mixed lymphocyte reaction assays was delayed in animals that received CO therapy. Furthermore, serum concentrations of proinflammatory cytokines (interleukin-1β and -6) 1 day after transplant were significantly decreased in the CO-treated group. CONCLUSIONS Fully MHC-mismatched lungs in CLAWN miniature swine were consistently rejected within 63 days, suggesting that this is a robust large animal model ideal for investigating mechanisms and treatment of lung rejection. Perioperative low-dose CO inhalation prolonged graft survival and inhibited antidonor antibody production and was associated with decreased proinflammatory mediators in this model.
Collapse
|
33
|
Blancou P, Tardif V, Simon T, Rémy S, Carreño L, Kalergis A, Anegon I. Immunoregulatory properties of heme oxygenase-1. Methods Mol Biol 2011; 677:247-268. [PMID: 20941616 DOI: 10.1007/978-1-60761-869-0_18] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. A growing body of evidence points toward the capacity of this molecule to inhibit immune reactions and the pivotal role of HO-1 in inflammatory diseases. We will first review the physiological role of HO-1 as determined by the analysis of HO-1-deficient individuals. This will be followed by an examination of the effect of HO-1 within immunopathological contexts such as immune disorders (autoimmunity and allergy) or infections. A section will be devoted to the use of an HO-1 inducer as an immunosuppressive molecule in transplantation. Finally, we will review the molecular basis of HO-1 actions on different immune cells.
Collapse
|
34
|
Abstract
Shortly after the identification of nitric oxide (NO) as a product of macrophages, it was discovered that NO generated by inducible NO synthase (iNOS) inhibits the proliferation of T lymphocytes. Since then, it has become clear that iNOS activity also regulates the development, differentiation, and/or function of various types of T cells and B cells and also affects NK cells. The three key mechanisms underlying the iNOS-dependent immunoregulation are (a) the modulation of signaling processes by NO, (b) the depletion of arginine, and (c) the alteration of accessory cell functions. This chapter highlights important principles of iNOS-dependent immunoregulation of lymphocytes and also reviews more recent evidence for an effect of endothelial or neuronal NO synthase in lymphocytes.
Collapse
Affiliation(s)
- Christian Bogdan
- Medical Microbiology and Immunology of Infectious Diseases, Microbiology Institute - Clinical Microbiology, Immunology and Hygiene, Friedrich-Alexander-University Erlangen-Nuremberg and University Clinic of Erlangen, Erlangen, Germany
| |
Collapse
|
35
|
Hydrogen sulfide as an effective and specific novel therapy for acute carbon monoxide poisoning. Biochem Biophys Res Commun 2011; 404:6-9. [DOI: 10.1016/j.bbrc.2010.11.113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Accepted: 11/24/2010] [Indexed: 01/23/2023]
|
36
|
Burt TD, Seu L, Mold JE, Kappas A, McCune JM. Naive human T cells are activated and proliferate in response to the heme oxygenase-1 inhibitor tin mesoporphyrin. THE JOURNAL OF IMMUNOLOGY 2010; 185:5279-88. [PMID: 20921523 DOI: 10.4049/jimmunol.0903127] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heme oxygenase-1 (HO-1) and its catabolic by-products have potent anti-inflammatory activity in many models of disease. It is not known, however, if HO-1 also plays a role in the homeostatic control of T cell activation and proliferation. We demonstrate here that the HO-1 inhibitor tin mesoporphyrin (SnMP) induces activation, proliferation, and maturation of naive CD4(+) and CD8(+) T cells via interactions with CD14(+) monocytes in vitro. This response is dependent upon interactions of T cells with MHC class I and II on the surface of CD14(+) monocytes. Furthermore, CD4(+)CD25(+)FoxP3(+) regulatory T cells were able to suppress this proliferation, even though their suppressive activity was itself impaired by SnMP. Given the magnitude of the Ag-independent T cell response induced by SnMP, we speculate that HO-1 plays an important role in dampening nonspecific T cell activation. Based on these findings, we propose a potential role for HO-1 in the control of naive T cell homeostatic proliferation.
Collapse
Affiliation(s)
- Trevor D Burt
- Division of Neonatology, Department of Pediatrics, University of California San Francisco, San Francisco, CA 94110, USA
| | | | | | | | | |
Collapse
|
37
|
|
38
|
Li M, Li Z, Sun X, Yang L, Fang P, Liu Y, Li W, Xu J, Lu J, Xie M, Zhang D. Heme oxygenase-1/p21WAF1 mediates peroxisome proliferator-activated receptor-gamma signaling inhibition of proliferation of rat pulmonary artery smooth muscle cells. FEBS J 2010; 277:1543-50. [PMID: 20163460 DOI: 10.1111/j.1742-4658.2010.07581.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Activation of peroxisome proliferator-activated receptor (PPAR)-gamma suppresses proliferation of rat pulmonary artery smooth muscle cells (PASMCs), and therefore ameliorates the development of pulmonary hypertension in animal models. However, the molecular mechanisms underlying this effect remain largely unknown. This study addressed this issue. The PPARgamma agonist rosiglitazone dose-dependently stimulated heme oxygenase (HO)-1 expression in PASMCs, 5 microm rosiglitazone inducing a 12.1-fold increase in the HO-1 protein level. Cells pre-exposed to rosiglitazone showed a dose-dependent reduction in proliferation in response to serotonin; this was abolished by pretransfection of cells with sequence-specific small interfering RNA against HO-1. In addition, rosiglitazone stimulated p21(WAF1) expression in PASMCs, a 2.34-fold increase in the p21(WAF1) protein level being achieved with 5 microm rosiglitazone; again, this effect was blocked by knockdown of HO-1. Like loss of HO-1, loss of p21(WAF1) through siRNA transfection also reversed the inhibitory effect of rosiglitazone on PASMC proliferation triggered by serotonin. Taken together, our findings suggest that activation of PPARgamma induces HO-1 expression, and that this in turn stimulates p21(WAF1) expression to suppress PASMC proliferation. Our study also indicates that rosiglitazone, a medicine widely used in the treatment of type 2 diabetes mellitus, has potential benefits for patients with pulmonary hypertension.
Collapse
Affiliation(s)
- Manxiang Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Rubio V, Valverde M, Rojas E. Effects of atmospheric pollutants on the Nrf2 survival pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2010; 17:369-82. [PMID: 19367423 DOI: 10.1007/s11356-009-0140-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Accepted: 02/16/2009] [Indexed: 04/15/2023]
Abstract
BACKGROUND, AIM, AND SCOPE Atmospheric pollution is a worldwide problem. Exposure to atmospheric pollutants causes toxic cellular effects. One of the mechanisms of toxicity by these pollutants is the promotion of oxidative stress. Several signaling pathways control cellular redox homeostasis. In this respect, nuclear factor erythroid 2-related factor 2 (Nrf2) is a crucial transcription factor in the cell's response to oxidative stress. MAIN FEATURES In cellular animal models, exposure to atmospheric pollutants activates Nrf2, attenuating its toxic and even its carcinogenic effects. Therefore, we have reviewed the scientific literature in order to indicate that air pollutants, such as particulate matter, polycyclic aromatic hydrocarbons, and gaseous matter, are Nrf2 pathway inductors, triggering self-defense through the establishment of proinflammatory and antioxidant responses. RESULTS AND DISCUSSION Exposure to reactive molecules as atmospheric pollutants causes the activation of Nrf2 and the subsequent regulation of the expression of cytoprotective and detoxifying enzymes, as well as antioxidants. Moreover, induction of Nrf2 prior to exposure reduces the harmful effects of pollutants. The present article discusses the protective role of the Nrf2 pathway against different atmospheric pollutant insults. CONCLUSIONS Nrf2 regulates the expression of numerous cytoprotective genes that function to detoxify reactive species produced during atmospheric pollutant metabolic reactions. From the papers highlighted in this review, we conclude that Nrf2 has an important role in the defense against atmospheric pollutant-induced toxicity. PERSPECTIVES Further studies are needed to understand the signaling events that turn on the system in response to atmospheric pollutant stress. This could allow for the possibility of targeting the pathway for prevention benefits in the near future.
Collapse
Affiliation(s)
- Valentina Rubio
- Instituto de Investigaciones Biomédicas, Departamento de Medicina Genómica y Toxicología Ambiental, Universidad Nacional Autónoma de México, México D.F., 04510, Mexico
| | | | | |
Collapse
|
40
|
Schwer CI, Mutschler M, Stoll P, Goebel U, Humar M, Hoetzel A, Schmidt R. Carbon monoxide releasing molecule-2 inhibits pancreatic stellate cell proliferation by activating p38 mitogen-activated protein kinase/heme oxygenase-1 signaling. Mol Pharmacol 2010; 77:660-9. [PMID: 20053955 DOI: 10.1124/mol.109.059519] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Proliferation of pancreatic stellate cells (PSCs) plays a cardinal role during fibrosis development. Therefore, the suppression of PSC growth represents a therapeutic option for the treatment of pancreatic fibrosis. It has been shown that up-regulation of the enzyme heme oxygenase-1 (HO-1) could exert antiproliferative effects on PSCs, but no information is available on the possible role of carbon monoxide (CO), a catalytic byproduct of the HO metabolism, in this process. In the present study, we have examined the effect of CO releasing molecule-2 (CORM-2) liberated CO on PSC proliferation and have elucidated the mechanisms involved. Using primary rat PSCs, we found that CORM-2 inhibited PSC proliferation at nontoxic concentrations by arresting cells at the G(0)/G(1) phase of the cell cycle. This effect was associated with activation of p38 mitogen-activated protein kinase (MAPK) signaling, induction of HO-1 protein, and up-regulation of the cell cycle inhibitor p21(Waf1/Cip1). The p38 MAPK inhibitor 4-(4-flurophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)imidazole (SB203580) abolished the inhibitory effect of CORM-2 on PSC proliferation and prevented both CORM-2-induced HO-1 and p21(Waf1/Cip1) up-regulation. Treatment with tin protoporphyrin IX, an HO inhibitor, or transfection of HO-1 small interfering RNA abolished the inductive effect of CORM-2 on p21(Waf1/Cip1) and reversed the suppressive effect of CORM-2 on PSC growth. The ability of CORM-2 to induce cell cycle arrest was abrogated in p21(Waf1/Cip1)-silenced cells. Taken together, our results suggest that CORM-2 inhibits PSC proliferation by activation of the p38/HO-1 pathway. These findings may indicate a therapeutic potential of CO carriers in the treatment of pancreatic fibrosis.
Collapse
Affiliation(s)
- Christian I Schwer
- Department of Anesthesiology, University Medical Center, Hugstetter Strasse 55, D-79106 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Donor HO-1 Expression Inhibits Intimal Hyperplasia in Unmanipulated Graft Recipients: A Potential Role for CD8+ T-Cell Modulation by Carbon Monoxide. Transplantation 2009; 88:653-61. [DOI: 10.1097/tp.0b013e3181b2fd83] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Bannenberg GL, Vieira HLA. Therapeutic applications of the gaseous mediators carbon monoxide and hydrogen sulfide. Expert Opin Ther Pat 2009; 19:663-82. [PMID: 19441940 DOI: 10.1517/13543770902858824] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hydrogen sulfide (H(2)S) and carbon monoxide (CO) are endogenously produced gaseous autacoids that regulate a number of physiological processes, including the inflammatory response, cell death and proliferation, neural transmission and smooth muscle tone. OBJECTIVE/METHODS The current review aims to provide a comprehensive overview of all recent patent applications that address the potential therapeutic applications of CO and H(2)S. RESULTS/CONCLUSION Beyond the direct administration of CO and H(2)S, this review highlights the therapeutic applications of a variety of gas-releasing molecules that are being developed to deliver CO and H(2)S to diseased tissues at therapeutic doses. The term autacoid, which, in addition to its pharmacological use to describe a locally-acting hormone, literally translates from Greek as 'self-drug', seems to particularly well describe the current approach to capture the potential therapeutic use of these two gasotransmitters. In summary, we can conclude that there is a markedly growing interest in harnessing the tissue-protective actions of CO and H(2)S.
Collapse
Affiliation(s)
- Gerard L Bannenberg
- Campus de la Universidad Autónoma, Centro Nacional de Biotecnología / CSIC, Department of Plant Molecular Genetics, Madrid, Spain.
| | | |
Collapse
|
43
|
Induction of heme oxygenase-1 with hemin attenuates hippocampal injury in rats after acute carbon monoxide poisoning. Toxicology 2009; 262:146-52. [PMID: 19520142 DOI: 10.1016/j.tox.2009.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 05/19/2009] [Accepted: 06/01/2009] [Indexed: 11/20/2022]
Abstract
Carbon monoxide (CO) poisoning is a major cause of brain injury and mortality; delayed neurological syndrome (DNS) is encountered in survivors of acute CO exposure. The toxic effects of CO have been attributed to oxidative stress induced by hypoxia. Heme oxygenase-1 (HO-1) is the inducible heme oxygenase isoform, and its induction acts as an important cellular defense mechanism against oxidative stress, cellular injury and disease. In this study, we examined the functional roles of HO-1 induction in a rat model of CO-exposured hippocampal injury. We report that acute CO exposure produces severe hippocampal injury in rats. However, hemin pretreatment reduced both the CO-induced rise in hippocampal water content and levels of neuronal damage in the hippocampus; survival rates at 24 h were significantly improved. Upregulation of HO-1 by hemin pretreatment resulted in a significant decrease in hippocampal levels of malondialdehyde (MDA), a marker of oxidative stress; levels of pro-apoptotic caspase-3 were also reduced. In contrast, inhibition of HO activity by administration of tin protoporphyrin IX (SnPP, a specific inhibitor of HO) abolished the neuroprotective effects of HO-1 induction. These data suggested that the upregulation of endogenous HO-1 expression therefore plays a pivotal protective role in CO neurotoxicity. Though the precise mechanisms underlying hemin-mediated HO-1 induction and neuroprotection are not known, these may involve the anti-oxidant and anti-apoptotic effects of HO-1 enzyme activity.
Collapse
|
44
|
Vadori M, Seveso M, Besenzon F, Bosio E, Tognato E, Fante F, Boldrin M, Gavasso S, Ravarotto L, Mann BE, Simioni P, Ancona E, Motterlini R, Cozzi E. In vitro and in vivo effects of the carbon monoxide-releasing molecule, CORM-3, in the xenogeneic pig-to-primate context. Xenotransplantation 2009; 16:99-114. [PMID: 19392725 DOI: 10.1111/j.1399-3089.2009.00521.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Carbon monoxide (CO) interferes with inflammatory and apoptotic processes associated with ischemia-reperfusion injury and graft rejection. Here, the in vitro effects of carbon monoxide releasing molecule-3 (CORM-3), a novel water-soluble carbonyl CO carrier, have been investigated on porcine aortic endothelial cells (PAEC) and primate peripheral blood mononuclear cells (PBMC). Furthermore, the pharmacodynamics and pharmacotolerance of CORM-3 after administration of single and multiple doses in the primate have been assessed in view of its potential application in pig-to-primate xenotransplantation models. METHODS For in vitro studies, PAEC and primate PBMC were exposed for 24, 48 and 72 h to CORM-3 (20 to 1000 microm) and viability was measured using an MTS assay. PAEC and primate PBMC proliferation after exposure to CORM-3 was assessed by CFSE labelling. Proliferation of primate PBMC against irradiated pig lymphocytes was also assessed. Tumor necrosis factor alpha (TNF-alpha) production and Caspase-3 and -7 activity in Concanavalin A (conA)-stimulated primate PBMC were measured following treatment with CORM-3. In vivo, CORM-3 was administered i.v. to cynomolgus monkeys at 4 mg/kg, as single or multiple doses for up to 30 days. The effect of CORM-3 was evaluated by the assessment of production of TNF-alpha and interleukin 1beta following PBMC stimulation with LPS by species-specific ELISA. Complete hematologic and biochemical analyses were routinely performed in treated primates. RESULTS At concentrations <500 microm, CORM-3 did not alter the viability of PAEC or primate PBMC cultures in vitro, nor did it induce significant levels of apoptosis or necrosis. Interestingly, at concentrations of 300 and 500 microm, significant PAEC proliferation was observed, whilst concentrations > or =50 microm inhibited conA-activated primate lymphocyte proliferation (IC(50) of 345.8 +/- 51.9 microm) and the primate xenogeneic response against pig PBMC. Such responses were demonstrated to be CO-dependent. In addition, CORM-3 significantly inhibited caspase-3 and -7 activity at concentrations between 200 and 500 microm and caused a significant reduction in TNF-alpha production (IC(50) 332.8 +/- 33.9 microm). In vivo, following the administration of multiple doses, TNF-alpha production was significantly reduced in comparison to pre-treatment responses, with decreased levels maintained throughout the study. Moreover, a slight and transient increase in transaminases and bilirubin was observed in animals exposed to multiple doses of CORM-3. CONCLUSIONS These studies suggest that CORM-3 has anti-inflammatory and immunomodulatory properties in primates that may result in clinical benefit to allo- and xenografted organs.
Collapse
Affiliation(s)
- Marta Vadori
- Consorzio per la Ricerca sul Trapianto d'Organi, University of Padua, Via dei Giustiniani 2, Padua, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Zaki MH, Fujii S, Okamoto T, Islam S, Khan S, Ahmed KA, Sawa T, Akaike T. Cytoprotective function of heme oxygenase 1 induced by a nitrated cyclic nucleotide formed during murine salmonellosis. THE JOURNAL OF IMMUNOLOGY 2009; 182:3746-56. [PMID: 19265153 DOI: 10.4049/jimmunol.0803363] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Signaling mechanisms of NO-mediated host defense are yet to be elucidated. In this study, we report a unique signal pathway for cytoprotection during Salmonella infection that involves heme oxygenase 1 (HO-1) induced by a nitrated cyclic nucleotide, 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP). Wild-type C57BL/6 mice and C57BL/6 mice lacking inducible NO synthase (iNOS) were infected with Salmonella enterica serovar Typhimurium LT2. HO-1 was markedly up-regulated during the infection, the level being significantly higher in wild-type mice than in iNOS-deficient mice. HO-1 up-regulation was associated with 8-nitro-cGMP formation detected immunohistochemically in Salmonella-infected mouse liver and peritoneal macrophages. 8-Nitro-cGMP either exogenously added or formed endogenously induced HO-1 in cultured macrophages infected with Salmonella. HO-1 inhibition by polyethylene glycol-conjugated zinc-protoporphyrin IX impaired intracellular killing of bacteria in mouse liver and in both RAW 264 cells and peritoneal macrophages. Infection-associated apoptosis was also markedly increased in polyethylene glycol-conjugated zinc-protoporphyrin IX-treated mouse liver cells and cultured macrophages. This effect of HO-1 inhibition was further confirmed by using HO-1 short interfering RNA in peritoneal macrophages. Our results suggest that HO-1 induced by NO-mediated 8-nitro-cGMP formation contributes, via its potent cytoprotective function, to host defense during murine salmonellosis.
Collapse
Affiliation(s)
- Mohammad Hasan Zaki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Moreau A, Hill M, Thébault P, Deschamps JY, Chiffoleau E, Chauveau C, Moullier P, Anegon I, Alliot-Licht B, Cuturi MC. Tolerogenic dendritic cells actively inhibit T cells through heme oxygenase‐1 in rodents and in nonhuman primates. FASEB J 2009; 23:3070-7. [DOI: 10.1096/fj.08-128173] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- A. Moreau
- INSERM U649 CHU Hotel-Dieu Nantes France
| | - M. Hill
- INSERM U643 Nantes France
- CHU Nantes Institut de Transplantation et de Recherche en Transplantation Nantes France
- Université de Nantes Faculté de Médecine Nantes France
| | - P. Thébault
- INSERM U643 Nantes France
- CHU Nantes Institut de Transplantation et de Recherche en Transplantation Nantes France
- Université de Nantes Faculté de Médecine Nantes France
| | - J. Y. Deschamps
- Ecole Nationale Vétérinaire de Nantes Service d'Urgence Nantes France
| | - E. Chiffoleau
- INSERM U643 Nantes France
- CHU Nantes Institut de Transplantation et de Recherche en Transplantation Nantes France
- Université de Nantes Faculté de Médecine Nantes France
| | - C. Chauveau
- INSERM U643 Nantes France
- CHU Nantes Institut de Transplantation et de Recherche en Transplantation Nantes France
- Université de Nantes Faculté de Médecine Nantes France
| | | | - I. Anegon
- INSERM U643 Nantes France
- CHU Nantes Institut de Transplantation et de Recherche en Transplantation Nantes France
- Université de Nantes Faculté de Médecine Nantes France
| | - B. Alliot-Licht
- INSERM U643 Nantes France
- CHU Nantes Institut de Transplantation et de Recherche en Transplantation Nantes France
- Université de Nantes Faculté de Médecine Nantes France
| | - M. C. Cuturi
- INSERM U643 Nantes France
- CHU Nantes Institut de Transplantation et de Recherche en Transplantation Nantes France
- Université de Nantes Faculté de Médecine Nantes France
| |
Collapse
|
47
|
Therapeutic effects of heme oxygenase-1 on psoriasiform skin lesions in guinea pigs. Arch Dermatol Res 2009; 301:459-66. [DOI: 10.1007/s00403-009-0956-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 04/09/2009] [Accepted: 04/19/2009] [Indexed: 12/22/2022]
|
48
|
Abstract
Ischemia reperfusion injury (IRI) is a common and important clinical problem in many different organ systems, including kidney, brain, heart, liver, lung, and intestine. IRI occurs during all deceased donor organ transplants. IRI is a highly complex cascade of events that includes interactions between vascular endothelium, interstitial compartments, circulating cells, and numerous biochemical entities. It is well established that the innate immune system, such as complement, neutrophils, cytokines, chemokines, and macrophages participate in IRI. Recent data demonstrates an important role for lymphocytes, particularly T cells but also B cells in IRI. Lymphocytes not only participate in augmenting injury responses after IRI, but could also be playing a protective role depending on the cell type and stage of injury. Furthermore, lymphocytes appear to be participating in the healing response from IRI. These new data open the possibility for lymphocyte targeted therapeutics to improve the short and long term outcomes from IRI.
Collapse
Affiliation(s)
- Douglas Linfert
- Nephrology Division, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
49
|
Franco R, Sánchez-Olea R, Reyes-Reyes EM, Panayiotidis MI. Environmental toxicity, oxidative stress and apoptosis: ménage à trois. Mutat Res 2008; 674:3-22. [PMID: 19114126 DOI: 10.1016/j.mrgentox.2008.11.012] [Citation(s) in RCA: 352] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 11/27/2008] [Indexed: 12/21/2022]
Abstract
Apoptosis is an evolutionary conserved homeostatic process involved in distinct physiological processes including organ and tissue morphogenesis, development and senescence. Its deregulation is also known to participate in the etiology of several human diseases including cancer, neurodegenerative and autoimmune disorders. Environmental stressors (cytotoxic agents, pollutants or toxicants) are well known to induce apoptotic cell death and to contribute to a variety of pathological conditions. Oxidative stress seems to be the central element in the regulation of the apoptotic pathways triggered by environmental stressors. In this work, we review the established mechanisms by which oxidative stress and environmental stressors regulate the apoptotic machinery with the aim to underscore the relevance of apoptosis as a component in environmental toxicity and human disease progression.
Collapse
Affiliation(s)
- Rodrigo Franco
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, P. O. Box 12233, 111. T.W. Alexander Drive, Research Triangle Park, NC 27709, United States.
| | | | | | | |
Collapse
|
50
|
Cao YA, Wagers AJ, Karsunky H, Zhao H, Reeves R, Wong RJ, Stevenson DK, Weissman IL, Contag CH. Heme oxygenase-1 deficiency leads to disrupted response to acute stress in stem cells and progenitors. Blood 2008; 112:4494-502. [PMID: 18509090 PMCID: PMC2597124 DOI: 10.1182/blood-2007-12-127621] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Accepted: 04/18/2008] [Indexed: 12/15/2022] Open
Abstract
An effective response to extreme hematopoietic stress requires an extreme elevation in hematopoiesis and preservation of hematopoietic stem cells (HSCs). These diametrically opposed processes are likely to be regulated by genes that mediate cellular adaptation to physiologic stress. Herein, we show that heme oxygenase-1 (HO-1), the inducible isozyme of heme degradation, is a key regulator of these processes. Mice lacking one allele of HO-1 (HO-1(+/-)) showed accelerated hematopoietic recovery from myelotoxic injury, and HO-1(+/-) HSCs repopulated lethally irradiated recipients with more rapid kinetics. However, HO-1(+/-) HSCs were ineffective in radioprotection and serial repopulation of myeloablated recipients. Perturbations in key stem cell regulators were observed in HO-1(+/-) HSCs and hematopoietic progenitors (HPCs), which may explain the disrupted response of HO-1(+/-) HPCs and HPCs to acute stress. Control of stem cell stress response by HO-1 presents opportunities for metabolic manipulation of stem cell-based therapies.
Collapse
Affiliation(s)
- Yu-An Cao
- Departments of Pediatrics, Stanford University School of Medicine, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|