1
|
Su M, Ji X, Liu F, Li Z, Yan D. Chemical Strategies Toward Prodrugs and Fluorescent Probes for Gasotransmitters. Mini Rev Med Chem 2024; 24:300-329. [PMID: 37102481 DOI: 10.2174/1389557523666230427152234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/03/2023] [Accepted: 02/20/2023] [Indexed: 04/28/2023]
Abstract
Three gaseous molecules are widely accepted as important gasotransmitters in mammalian cells, namely NO, CO and H2S. Due to the pharmacological effects observed in preclinical studies, these three gasotransmitters represent promising drug candidates for clinical translation. Fluorescent probes of the gasotransmitters are also in high demand; however, the mechanisms of actions or the roles played by gasotransmitters under both physiological and pathological conditions remain to be answered. In order to bring these challenges to the attention of both chemists and biologists working in this field, we herein summarize the chemical strategies used for the design of both probes and prodrugs of these three gasotransmitters.
Collapse
Affiliation(s)
- Ma Su
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Suzhou University, China
| | - Xingyue Ji
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Suzhou University, China
| | - Feng Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Suzhou University, China
| | - Zhang Li
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Suzhou University, China
| | - Duanyang Yan
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Suzhou University, China
| |
Collapse
|
2
|
Niu Q, Du F, Yang X, Yang X, Wang X. Carbon monoxide-releasing molecule 2 inhibits inflammation associated with intestinal ischemia-reperfusion injury in a rat model of hemorrhagic shock. Int Immunopharmacol 2022; 113:109441. [DOI: 10.1016/j.intimp.2022.109441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/24/2022]
|
3
|
Silva RCMC, Vasconcelos LR, Travassos LH. The different facets of heme-oxygenase 1 in innate and adaptive immunity. Cell Biochem Biophys 2022; 80:609-631. [PMID: 36018440 DOI: 10.1007/s12013-022-01087-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 07/20/2022] [Indexed: 11/26/2022]
Abstract
Heme oxygenase (HO) enzymes are responsible for the main oxidative step in heme degradation, generating equimolar amounts of free iron, biliverdin and carbon monoxide. HO-1 is induced as a crucial stress response protein, playing protective roles in physiologic and pathological conditions, due to its antioxidant, anti-apoptotic and anti-inflammatory effects. The mechanisms behind HO-1-mediated protection are being explored by different studies, affecting cell fate through multiple ways, such as reduction in intracellular levels of heme and ROS, transcriptional regulation, and through its byproducts generation. In this review we focus on the interplay between HO-1 and immune-related signaling pathways, which culminate in the activation of transcription factors important in immune responses and inflammation. We also discuss the dual interaction of HO-1 and inflammatory mediators that govern resolution and tissue damage. We highlight the dichotomy of HO-1 in innate and adaptive immune cells development and activation in different disease contexts. Finally, we address different known anti-inflammatory pharmaceuticals that are now being described to modulate HO-1, and the possible contribution of HO-1 in their anti-inflammatory effects.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Luiz Ricardo Vasconcelos
- Cellular Signaling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Leonardo Holanda Travassos
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Lin H, Peng S, Guo S, Ma B, Lucherelli MA, Royer C, Ippolito S, Samorì P, Bianco A. 2D Materials and Primary Human Dendritic Cells: A Comparative Cytotoxicity Study. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107652. [PMID: 35451183 DOI: 10.1002/smll.202107652] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/24/2022] [Indexed: 06/14/2023]
Abstract
Human health can be affected by materials indirectly through exposure to the environment or directly through close contact and uptake. With the ever-growing use of 2D materials in many applications such as electronics, medical therapeutics, molecular sensing, and energy storage, it has become more pertinent to investigate their impact on the immune system. Dendritic cells (DCs) are highly important, considering their role as the main link between the innate and the adaptive immune system. By using primary human DCs, it is shown that hexagonal boron nitride (hBN), graphene oxide (GO) and molybdenum disulphide have minimal effects on viability. In particular, it is evidenced that hBN and GO increase DC maturation, while GO leads to the release of reactive oxygen species and pro-inflammatory cytokines. hBN and MoS2 increase T cell proliferation with and without the presence of DCs. hBN in particular does not show any sign of downstream T cell polarization. The study allows ranking of the three materials in terms of inherent toxicity, providing the following trend: GO > hBN ≈ MoS2 , with GO the most cytotoxic.
Collapse
Affiliation(s)
- Hazel Lin
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Shiyuan Peng
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Shi Guo
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Baojin Ma
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Matteo Andrea Lucherelli
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Cathy Royer
- Plateforme Imagerie In Vitro de l'ITI Neurostra, CNRS UAR 3156, University of Strasbourg, Strasbourg, 67000, France
| | | | - Paolo Samorì
- CNRS, ISIS, Université de Strasbourg, Strasbourg, 67000, France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry UPR 3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| |
Collapse
|
5
|
Watabe Y, Taguchi K, Sakai H, Enoki Y, Maruyama T, Otagiri M, Kohno M, Matsumoto K. Bioinspired carbon monoxide delivery using artificial blood attenuates the progression of obliterative bronchiolitis via suppression of macrophage activation by IL-17A. Eur J Pharm Biopharm 2021; 170:43-51. [PMID: 34864198 DOI: 10.1016/j.ejpb.2021.11.011] [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] [Received: 07/19/2021] [Revised: 10/28/2021] [Accepted: 11/28/2021] [Indexed: 12/18/2022]
Abstract
Carbon monoxide (CO) is expected to attenuate the progression of obliterative bronchiolitis (OB), which is a serious complication after lung transplantation. However, issues in terms of feasible exogenous CO supply, such as continuousness and safety, remain unsolved. Here, we applied nano red blood cells, namely hemoglobin vesicles (Hb-V), as a CO cargo based on the biomimetic concept and investigated the therapeutic potential of CO-loaded Hb-V on OB in orthotopic tracheal transplant model mice. The CO-loaded Hb-V was comprised of negatively charged liposomes encapsulating carbonylhemoglobin with a size of ca. 220 nm. The results of histological evaluation showed that allograft luminal occlusion and fibrosis were significantly ameliorated by treatment with CO-loaded Hb-V compared to treatment with saline, cyclosporine, and Hb-V. The therapeutic effects of CO-loaded Hb-V on OB were due to the suppression of M1 macrophage activation in tracheal allografts, resulting from decreased IL-17A production. Furthermore, the expression of TNF-α and TGF-β in tracheal allografts was decreased by CO-loaded Hb-V treatment but not saline and Hb-V treatment, indicating that CO liberated from CO-loaded Hb-V inhibits epithelial-mesenchymal transition. These findings suggest that CO-loaded Hb-V exerts strong therapeutic efficacy against OB via the regulation of macrophage activation by IL-17A and TGF-β-driven epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Yuki Watabe
- Faculty of Pharmacy, Keio University, Tokyo, Japan
| | | | - Hiromi Sakai
- Department of Chemistry, Nara Medical University, Kashihara, Japan
| | - Yuki Enoki
- Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Toru Maruyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan; DDS Research Institute, Sojo University, Kumamoto, Japan
| | - Mitsutomo Kohno
- Department of General Thoracic Surgery, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | | |
Collapse
|
6
|
Yang X, Lu W, Hopper CP, Ke B, Wang B. Nature's marvels endowed in gaseous molecules I: Carbon monoxide and its physiological and therapeutic roles. Acta Pharm Sin B 2021; 11:1434-1445. [PMID: 34221861 PMCID: PMC8245769 DOI: 10.1016/j.apsb.2020.10.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 08/03/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
Nature has endowed gaseous molecules such as O2, CO2, CO, NO, H2S, and N2 with critical and diverse roles in sustaining life, from supplying energy needed to power life and building blocks for life's physical structure to mediating and coordinating cellular functions. In this article, we give a brief introduction of the complex functions of the various gaseous molecules in life and then focus on carbon monoxide as a specific example of an endogenously produced signaling molecule to highlight the importance of this class of molecules. The past twenty years have seen much progress in understanding CO's mechanism(s) of action and pharmacological effects as well as in developing delivery methods for easy administration. One remarkable trait of CO is its pleiotropic effects that have few parallels, except perhaps its sister gaseous signaling molecules such as nitric oxide and hydrogen sulfide. This review will delve into the sophistication of CO-mediated signaling as well as its validated pharmacological functions and possible therapeutic applications.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Wen Lu
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| | - Christopher P. Hopper
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
- Institut für Experimentelle Biomedizin, Universitätsklinikum Würzburg, Würzburg, Bavaria 97080, Germany
| | - Bowen Ke
- Department of Anesthesiology, West China Hospital, Chengdu 610041, China
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
7
|
P2X7 Receptors Mediate CO-Induced Alterations in Gene Expression in Cultured Cortical Astrocytes—Transcriptomic Study. Mol Neurobiol 2018; 56:3159-3174. [DOI: 10.1007/s12035-018-1302-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 08/06/2018] [Indexed: 01/31/2023]
|
8
|
Takagi T, Naito Y, Tanaka M, Mizushima K, Ushiroda C, Toyokawa Y, Uchiyama K, Hamaguchi M, Handa O, Itoh Y. Carbon monoxide ameliorates murine T-cell-dependent colitis through the inhibition of Th17 differentiation. Free Radic Res 2018; 52:1328-1335. [PMID: 29695203 DOI: 10.1080/10715762.2018.1470327] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent studies have identified carbon monoxide (CO) as a potential therapeutic molecule for the treatment of inflammatory diseases including intestinal inflammation. In the present study, we explored the efficacy and the mechanisms of action of CO-releasing molecule (CORM)-A1 in T-cell transfer induced colitis model in mice. In addition, the impact of CORM-A1 on the T helper (Th) cell differentiation was evaluated using naïve CD4+ T cells isolated from the spleens in Balb/c mice. The results showed that CORM-A1 conferred protection against the development of intestinal inflammation and attenuated Th17 cell differentiation. Hence, the observed immunomodulatory effects of CORM-A1 could be useful for developing novel therapeutic approaches for managing intestinal inflammation through the regulation of Th17 differentiation.
Collapse
Affiliation(s)
- Tomohisa Takagi
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan.,b Department of Medical Innovation and Translational Medical Science, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Yuji Naito
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Makoto Tanaka
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Katsura Mizushima
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Chihiro Ushiroda
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Yuki Toyokawa
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Kazuhiko Uchiyama
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Masahide Hamaguchi
- c Department of Endocrinology and Metabolism, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Osamu Handa
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Yoshito Itoh
- a Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science , Kyoto Prefectural University of Medicine , Kyoto , Japan
| |
Collapse
|
9
|
Chen X, Liu Q, Huang W, Cai C, Xia W, Peng Y, Zheng S, Li G, Xu Y, Wang J, Liu C, Zhang X, Huang L, Xiang AP, Zhang Q. Stanniocalcin-2 contributes to mesenchymal stromal cells attenuating murine contact hypersensitivity mainly via reducing CD8 + Tc1 cells. Cell Death Dis 2018; 9:548. [PMID: 29748538 PMCID: PMC5945630 DOI: 10.1038/s41419-018-0614-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/19/2018] [Accepted: 04/18/2018] [Indexed: 12/22/2022]
Abstract
Mesenchymal stromal cells (MSCs) have been demonstrated to ameliorate allergic contact dermatitis (ACD), a typical T-cell-mediated disorder. However, the underlying mechanisms behind the MSC-based treatment for ACD have not yet been fully elucidated. The stanniocalcins (STCs) comprise a family of secreted glycoprotein hormones that act as important anti-inflammatory proteins. Here, we investigated the roles of STCs in MSC-mediated T-cell suppression and their potential role in the MSC-based treatment for ACD. Gene expression profiling revealed that STC2, but not STC1, was highly expressed in MSCs. STC2 knockdown in MSCs significantly impaired their effects in reducing TNF-α- and IFN-γ-producing CD8+ T cells. Importantly, silencing the STC2 expression in MSCs abated their therapeutic effect on contact hypersensitivity (CHS) in mice, mainly restoring the generation and infiltration of IFN-γ-producing CD8+ T cells (Tc1 cells). Mechanistically, STC2 co-localized with heme oxygenase 1 (HO-1) in MSCs, and contributed to MSC-mediated reduction of CD8+ Tc1 cells via regulating HO-1 activity. Together, these findings newly identify STC2 as the first stanniocalcin responsible for mediating the immunomodulatory effects of MSCs on allogeneic T cells and STC2 contribute to MSC-based treatment for ACD mainly via reducing the CD8+ Tc1 cells.
Collapse
Affiliation(s)
- Xiaoyong Chen
- The Biotherapy Center, the Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, 510630, Guangzhou, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Qiuli Liu
- The Biotherapy Center, the Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, 510630, Guangzhou, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Chuang Cai
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Wenjie Xia
- Institute of Blood Transfusion, Guangzhou Blood Centre, 510095, Guangzhou, China
| | - Yanwen Peng
- The Biotherapy Center, the Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, 510630, Guangzhou, China
| | - Shuwei Zheng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Gang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Yan Xu
- The Biotherapy Center, the Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, 510630, Guangzhou, China
| | - Jiancheng Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Chang Liu
- The Biotherapy Center, the Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, 510630, Guangzhou, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Li Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Andy Peng Xiang
- The Biotherapy Center, the Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, 510630, Guangzhou, China.
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, 510080, Guangzhou, China.
- Department of Biochemistry, Zhongshan Medical School, Sun Yat-Sen University, 510080, Guangzhou, China.
| | - Qi Zhang
- The Biotherapy Center, the Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, 510630, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, China.
| |
Collapse
|
10
|
Yoon SJ, Kim SJ, Lee SM. Overexpression of HO-1 Contributes to Sepsis-Induced Immunosuppression by Modulating the Th1/Th2 Balance and Regulatory T-Cell Function. J Infect Dis 2017; 215:1608-1618. [DOI: 10.1093/infdis/jix142] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/17/2017] [Indexed: 01/07/2023] Open
|
11
|
Maharshak N, Ryu HS, Fan TJ, Onyiah JC, Schulz S, Otterbein SL, Wong R, Hansen JJ, Otterbein LE, Carroll IM, Plevy SE. Escherichia coli heme oxygenase modulates host innate immune responses. Microbiol Immunol 2016; 59:452-65. [PMID: 26146866 DOI: 10.1111/1348-0421.12282] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 06/24/2015] [Accepted: 06/28/2015] [Indexed: 12/13/2022]
Abstract
Induction of mammalian heme oxygenase (HO)-1 and exposure of animals to carbon monoxide (CO) ameliorates experimental colitis. When enteric bacteria, including Escherichia coli, are exposed to low iron conditions, they express an HO-like enzyme, chuS, and metabolize heme into iron, biliverdin and CO. Given the abundance of enteric bacteria residing in the intestinal lumen, our postulate was that commensal intestinal bacteria may be a significant source of CO and those that express chuS and other Ho-like molecules suppress inflammatory immune responses through release of CO. According to real-time PCR, exposure of mice to CO results in changes in enteric bacterial composition and increases E. coli 16S and chuS DNA. Moreover, the severity of experimental colitis correlates positively with E. coli chuS expression in IL-10 deficient mice. To explore functional roles, E. coli were genetically modified to overexpress chuS or the chuS gene was deleted. Co-culture of chuS-overexpressing E. coli with bone marrow-derived macrophages resulted in less IL-12p40 and greater IL-10 secretion than in wild-type or chuS-deficient E. coli. Mice infected with chuS-overexpressing E. coli have more hepatic CO and less serum IL-12 p40 than mice infected with chuS-deficient E. coli. Thus, CO alters the composition of the commensal intestinal microbiota and expands populations of E. coli that harbor the chuS gene. These bacteria are capable of attenuating innate immune responses through expression of chuS. Bacterial HO-like molecules and bacteria-derived CO may represent novel targets for therapeutic intervention in inflammatory conditions.
Collapse
Affiliation(s)
- Nitsan Maharshak
- Department of Medicine and Center for GI Biology and Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599.,Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Hyungjin Sally Ryu
- Department of Medicine and Center for GI Biology and Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Ting-Jia Fan
- Department of Medicine and Center for GI Biology and Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Joseph C Onyiah
- Department of Medicine and Center for GI Biology and Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599.,Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, Aurora, CO and Denver VA Medical Center, Denver, Colorado, 80220
| | - Stephanie Schulz
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, California, 94305
| | - Sherrie L Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Ron Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, California, 94305
| | - Jonathan J Hansen
- Department of Medicine and Center for GI Biology and Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Leo E Otterbein
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Palo Alto, California, 94305
| | - Ian M Carroll
- Department of Medicine and Center for GI Biology and Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| | - Scott E Plevy
- Department of Medicine and Center for GI Biology and Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599
| |
Collapse
|
12
|
Almeida AS, Soares NL, Vieira M, Gramsbergen JB, Vieira HLA. Carbon Monoxide Releasing Molecule-A1 (CORM-A1) Improves Neurogenesis: Increase of Neuronal Differentiation Yield by Preventing Cell Death. PLoS One 2016; 11:e0154781. [PMID: 27144388 PMCID: PMC4856303 DOI: 10.1371/journal.pone.0154781] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 04/19/2016] [Indexed: 11/19/2022] Open
Abstract
Cerebral ischemia and neurodegenerative diseases lead to impairment or death of neurons in the central nervous system. Stem cell based therapies are promising strategies currently under investigation. Carbon monoxide (CO) is an endogenous product of heme degradation by heme oxygenase (HO) activity. Administration of CO at low concentrations produces several beneficial effects in distinct tissues, namely anti-apoptotic and anti-inflammatory. Herein the CO role on modulation of neuronal differentiation was assessed. Three different models with increasing complexity were used: human neuroblastoma SH-S5Y5 cell line, human teratocarcinoma NT2 cell line and organotypic hippocampal slice cultures (OHSC). Cell lines were differentiated into post-mitotic neurons by treatment with retinoic acid (RA) supplemented with CO-releasing molecule A1 (CORM-A1). CORM-A1 positively modulated neuronal differentiation, since it increased final neuronal production and enhanced the expression of specific neuronal genes: Nestin, Tuj1 and MAP2. Furthermore, during neuronal differentiation process, there was an increase in proliferative cell number (ki67 mRNA expressing cells) and a decrease in cell death (lower propidium iodide (PI) uptake, limitation of caspase-3 activation and higher Bcl-2 expressing cells). CO supplementation did not increase the expression of RA receptors. In the case of SH-S5Y5 model, small amounts of reactive oxygen species (ROS) generation emerges as important signaling molecules during CO-promoted neuronal differentiation. CO's improvement of neuronal differentiation yield was validated using OHSC as ex vivo model. CORM-A1 treatment of OHSC promoted higher levels of cells expressing the neuronal marker Tuj1. Still, CORM-A1 increased cell proliferation assessed by ki67 expression and also prevented cell death, which was followed by increased Bcl-2 expression, decreased levels of active caspase-3 and PI uptake. Likewise, ROS signaling emerged as key factors in CO's increasing number of differentiated neurons in OHSC. In conclusion, CO's increasing number of differentiated neurons is a novel biological role disclosed herein. CO improves neuronal yield due to its capacity to reduce cell death, promoting an increase in proliferative population. However, one cannot disregard a direct CO's effect on specific cellular processes of neuronal differentiation. Further studies are needed to evaluate how CO can potentially modulate cell mechanisms involved in neuronal differentiation. In summary, CO appears as a promising therapeutic molecule to stimulate endogenous neurogenesis or to improve in vitro neuronal production for cell therapy strategies.
Collapse
Affiliation(s)
- Ana S. Almeida
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
- Instituto de Tecnologia Química e Biológica (ITQB), Universidade Nova de Lisboa, Apartado 127, 2781-901 Oeiras, Portugal
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal
| | - Nuno L. Soares
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - Melissa Vieira
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - Jan Bert Gramsbergen
- Institute of Molecular Medicine, University of Southern Denmark, Winsløwparken 21, DK-5000 Odense C, Denmark
| | - Helena L. A. Vieira
- CEDOC, Faculdade de Ciência Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal
| |
Collapse
|
13
|
Schisandrin B inhibits Th1/Th17 differentiation and promotes regulatory T cell expansion in mouse lymphocytes. Int Immunopharmacol 2016; 35:257-264. [PMID: 27085037 DOI: 10.1016/j.intimp.2016.03.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 03/25/2016] [Accepted: 03/27/2016] [Indexed: 01/25/2023]
Abstract
Schisandrin B (Sch-B), the most abundant active ingredient of the fruit of Schisandra chinensis, has been proposed to have antioxidant, anti-tumor and anti-inflammatory effects. The present study was undertaken to investigate the effect of Sch-B on differentiation of T helper cells (Th). Using mouse splenic lymphocytes stimulated with concanavalin A (Con A) in vitro and ex vivo as inflammation models, we found that Sch-B significantly inhibited secretion of Th1 and Th17 related cytokines, such as IFN-γ and IL-17. In addition, we found that Sch-B suppressed the differentiation of naive CD4+ T cells into Th1 and Th17 cells, while promoted their differentiation into the regulatory T cells (Treg) in vitro. We further found that Sch-B suppressed transcription of Th1-related T-box transcription factor, T-bet, and Th17-related transcription factor, retinoid related orphan receptor gamma t (RORγt), while enhanced transcription of Treg-related transcription factor forkhead box protein 3 (Foxp3) in naive CD4+ T cells under Th cell polarization conditions. Furthermore, the effect of Sch-B on the T cell differentiation was abrogated by heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin. Taken together, we conclude that Sch-B can modulate differentiation of naïve CD4+ T cells into specific lineages of effector cells, which may have potential benefits for treatment of autoimmune diseases.
Collapse
|
14
|
Naito Y, Takagi T, Uchiyama K, Katada K, Yoshikawa T. Multiple targets of carbon monoxide gas in the intestinal inflammation. Arch Biochem Biophys 2016; 595:147-52. [DOI: 10.1016/j.abb.2015.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 06/07/2015] [Accepted: 06/26/2015] [Indexed: 01/06/2023]
|
15
|
Oliveira SR, Vieira HLA, Duarte CB. Effect of carbon monoxide on gene expression in cerebrocortical astrocytes: Validation of reference genes for quantitative real-time PCR. Nitric Oxide 2015. [PMID: 26196856 DOI: 10.1016/j.niox.2015.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) is a widely used technique to characterize changes in gene expression in complex cellular and tissue processes, such as cytoprotection or inflammation. The accurate assessment of changes in gene expression depends on the selection of adequate internal reference gene(s). Carbon monoxide (CO) affects several metabolic pathways and de novo protein synthesis is crucial in the cellular responses to this gasotransmitter. Herein a selection of commonly used reference genes was analyzed to identify the most suitable internal control genes to evaluate the effect of CO on gene expression in cultured cerebrocortical astrocytes. The cells were exposed to CO by treatment with CORM-A1 (CO releasing molecule A1) and four different algorithms (geNorm, NormFinder, Delta Ct and BestKeeper) were applied to evaluate the stability of eight putative reference genes. Our results indicate that Gapdh (glyceraldehyde-3-phosphate dehydrogenase) together with Ppia (peptidylpropyl isomerase A) is the most suitable gene pair for normalization of qRT-PCR results under the experimental conditions used. Pgk1 (phosphoglycerate kinase 1), Hprt1 (hypoxanthine guanine phosphoribosyl transferase I), Sdha (Succinate Dehydrogenase Complex, Subunit A), Tbp (TATA box binding protein), Actg1 (actin gamma 1) and Rn18s (18S rRNA) genes presented less stable expression profiles in cultured cortical astrocytes exposed to CORM-A1 for up to 60 min. For validation, we analyzed the effect of CO on the expression of Bdnf and bcl-2. Different results were obtained, depending on the reference genes used. A significant increase in the expression of both genes was found when the results were normalized with Gapdh and Ppia, in contrast with the results obtained when the other genes were used as reference. These findings highlight the need for a proper and accurate selection of the reference genes used in the quantification of qRT-PCR results in studies on the effect of CO in gene expression.
Collapse
Affiliation(s)
- Sara R Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Helena L A Vieira
- Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Lisbon, Portugal; Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal
| | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
16
|
Carbon monoxide-releasing molecule-A1 (CORM-A1) improves clinical signs of experimental autoimmune uveoretinitis (EAU) in rats. Clin Immunol 2015; 157:198-204. [PMID: 25701800 DOI: 10.1016/j.clim.2015.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 11/20/2022]
Abstract
Uveitis is a sight-threatening inflammatory disease of the eye which represents the third leading cause of blindness in the developed countries. The conventional pharmacological treatment includes corticosteroids and immunosuppressive agents, which are limited by their side effects. New therapeutic strategies are thus strongly needed. Exogenously-administered carbon monoxide (CO) may represent an effective treatment for conditions characterized by a dysregulated inflammatory response. Carbon monoxide-releasing molecules (CORMs) are a novel group of compounds capable of carrying and liberating controlled quantities of CO. Among CORMs, CORM-A1 represents the first example of water soluble CO releaser. We show here that CORM-A1 under a late prophylactic regime is able to significantly ameliorate the natural course of experimental autoimmune uveoretinitis, a rodent model of immunoinflammatory posterior uveitis. The present study strongly supports the development of CORM-A1 as a potential new drug for treatment of patients with non-infectious posterior uveitis.
Collapse
|