51
|
Adeniran SO, Zheng P, Feng R, Adegoke EO, Huang F, Ma M, Wang Z, Ifarajimi OO, Li X, Zhang G. The Antioxidant Role of Selenium via GPx1 and GPx4 in LPS-Induced Oxidative Stress in Bovine Endometrial Cells. Biol Trace Elem Res 2022; 200:1140-1155. [PMID: 33895964 DOI: 10.1007/s12011-021-02731-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/18/2021] [Indexed: 01/15/2023]
Abstract
This study investigated the antioxidant role of selenium (Se) in the form of selenomethionine (SLM) in LPS-induced oxidative stress via the glutathione peroxidase (GPx) enzymes and the Nrf2/HO-1 transcription factor. The impact of serum supplementation in culture media on GPxs was also studied. The bovine uterus is constantly exposed to exogenous pathogens postpartum, and the endometrium is the first contact against bacteria invasion. Endometritis is an inflammation of the endometrium and is brought about by bacterial lipopolysaccharide capable of inducing oxidative stress. The BEND cells were supplemented at the point of seeding with the following SLM concentrations 0, 100, 500, and 1000 nM for 48 h. BEND cells, cultured with or without SLM (100 nM), were initially incubated for 48 h, and then, we serum starved the SLM group for 24, 48, and 72 h. Similarly, an assay involving serum volume (0, 2, 5, and 10%) supplementation in culture media (v/v) with or without SLM (100 nM) was performed for 48 h. The BEND cells were also seeded into four experimental groups and cultured for an initial 48 h as follows: control, LPS (20 μg/mL), SLM (100 nM), and SLM + LPS groups followed by 6-h LPS treatment. The role of SLM in modulating the expressions of GPx1 and GPx4 and the Nrf2 transcription factor-related genes was assessed using qRT-PCR and Western blot techniques. The results showed serum starvation in the presence of SLM supplementation decreased the expression of GPx1 enzyme but increased GPx4 compared to the control. The addition of SLM to cell culture media in an FBS limiting condition improved the expressions of both GPx1 and GPx4. SLM supplementation promoted GPx enzymes' expressions in a serum-free media (0%) and at 2% FBS in media. However, it did not improve their expressions at 10% FBS in media than the untreated groups. Together, our data show the protective role of Se by regulating the expressions of GPx1 and GPx4 enzymes in BEND cells. It also shows that SLM promoted the expression of Nrf2 transcription factor-related genes at both the mRNA and protein levels in BEND cells during LPS stimulation.
Collapse
Affiliation(s)
- Samson O Adeniran
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Peng Zheng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Rui Feng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Elikanah O Adegoke
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
- Department of Animal Science and Technology and BET Research Institute, Chung-Ang University, Anseong, South Korea
| | - Fushuo Huang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Mingjun Ma
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Ziming Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Olamigoke O Ifarajimi
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Xiaoyu Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China
| | - Guixue Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Northeast Agricultural University, Harbin, People's Republic of China.
| |
Collapse
|
52
|
Ren B, Wu Q, Muskhelishvili L, Davis K, Wang Y, Rua D, Cao X. Evaluating the Sub-Acute Toxicity of Formaldehyde Fumes in an In Vitro Human Airway Epithelial Tissue Model. Int J Mol Sci 2022; 23:2593. [PMID: 35269734 PMCID: PMC8910234 DOI: 10.3390/ijms23052593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/22/2022] [Indexed: 11/22/2022] Open
Abstract
Formaldehyde (FA) is an irritating, highly reactive aldehyde that is widely regarded as an asthmagen. In addition to its use in industrial applications and being a product of combustion reaction and endogenous metabolism, FDA-regulated products may contain FA or release FA fumes that present toxicity risks for both patients and healthcare workers. Exposure to airborne FA is associated with nasal neoplastic lesions in both animals and humans. It is classified as a Group 1 carcinogen by International Agency for Research on Cancer (IARC) based on the increased incidence of cancer in animals and a known human carcinogen in the Report on Carcinogens by National Toxicology Program (NTP). Herein, we systematically evaluated the tissue responses to FA fumes in an in vitro human air-liquid-interface (ALI) airway tissue model. Cultures were exposed at the air interface to 7.5, 15, and 30 ppm of FA fumes 4 h per day for 5 consecutive days. Exposure to 30 ppm of FA induced sustained oxidative stress, along with functional changes in ciliated and goblet cells as well as possible squamous differentiation. Furthermore, secretion of the proinflammatory cytokines, IL-1β, IL-2, IL-8, GM-CSF, TNF-a and IFN-γ, was induced by repeated exposures to FA fumes. Expression of MMP-1, MMP-3, MMP-7, MMP-10, MMP-12, and MMP-13 was downregulated at the end of the 5-day exposure. Although DNA-damage was not detected by the comet assay, FA exposures downregulated the DNA repair enzymes MGMT and FANCD2, suggesting its possible interference in the DNA repair capacity. Overall, a general concordance was observed between our in vitro responses to FA fume exposures and the reported in vivo toxicity of FA. Our findings provide further evidence supporting the application of the ALI airway system as a potential in vitro alternative for screening and evaluating the respiratory toxicity of inhaled substances.
Collapse
Affiliation(s)
- Baiping Ren
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (B.R.); (Y.W.)
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA;
| | | | - Kelly Davis
- Toxicologic Pathology Associates, Jefferson, AR 72079, USA; (L.M.); (K.D.)
| | - Yiying Wang
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (B.R.); (Y.W.)
| | - Diego Rua
- Division of Biology, Chemistry, and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD 20993, USA;
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (B.R.); (Y.W.)
| |
Collapse
|
53
|
Gall Trošelj K, Tomljanović M, Jaganjac M, Matijević Glavan T, Čipak Gašparović A, Milković L, Borović Šunjić S, Buttari B, Profumo E, Saha S, Saso L, Žarković N. Oxidative Stress and Cancer Heterogeneity Orchestrate NRF2 Roles Relevant for Therapy Response. Molecules 2022; 27:1468. [PMID: 35268568 PMCID: PMC8912061 DOI: 10.3390/molecules27051468] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/19/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress and its end-products, such as 4-hydroxynonenal (HNE), initiate activation of the Nuclear Factor Erythroid 2-Related Factor 2 (NRF2)/Kelch Like ECH Associated Protein 1 (KEAP1) signaling pathway that plays a crucial role in the maintenance of cellular redox homeostasis. However, an involvement of 4-HNE and NRF2 in processes associated with the initiation of cancer, its progression, and response to therapy includes numerous, highly complex events. They occur through interactions between cancer and stromal cells. These events are dependent on many cell-type specific features. They start with the extent of NRF2 binding to its cytoplasmic repressor, KEAP1, and extend to the permissiveness of chromatin for transcription of Antioxidant Response Element (ARE)-containing genes that are NRF2 targets. This review will explore epigenetic molecular mechanisms of NRF2 transcription through the specific molecular anatomy of its promoter. It will explain the role of NRF2 in cancer stem cells, with respect to cancer therapy resistance. Additionally, it also discusses NRF2 involvement at the cross-roads of communication between tumor associated inflammatory and stromal cells, which is also an important factor involved in the response to therapy.
Collapse
Affiliation(s)
- Koraljka Gall Trošelj
- Laboratory for Epigenomics, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia;
| | - Marko Tomljanović
- Laboratory for Epigenomics, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia;
| | - Morana Jaganjac
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Tanja Matijević Glavan
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia;
| | - Ana Čipak Gašparović
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Lidija Milković
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Suzana Borović Šunjić
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.); (S.S.)
| | - Elisabetta Profumo
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.); (S.S.)
| | - Sarmistha Saha
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.); (S.S.)
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00161 Rome, Italy;
| | - Neven Žarković
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| |
Collapse
|
54
|
The Cyclic Nitroxide TEMPOL Ameliorates Oxidative Stress but Not Inflammation in a Cell Model of Parkinson’s Disease. Antioxidants (Basel) 2022; 11:antiox11020257. [PMID: 35204139 PMCID: PMC8868255 DOI: 10.3390/antiox11020257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/25/2022] Open
Abstract
The cyclic nitroxide TEMPOL exerts anti-oxidative and anti-inflammatory effects, and thus may provide therapeutic benefit in Parkinson’s disease (PD), in which mitochondrial dysfunction, oxidative damage and inflammation have been implicated as pathophysiological mechanisms underlying the selective loss of dopaminergic neurons. Markers of oxidative stress and inflammation were investigated in a cell model of differentiated human neuroblastoma (SH-SY5Y) cells treated with the neurotoxin, 6-hydroxydopamine (6-OHDA). Treatment with TEMPOL ameliorated 6-OHDA-mediated cytotoxicity and attenuated biomarkers of oxidative stress including: mitochondrial superoxide anion free radical production, lipid peroxidation, induction of heme oxygenase 1 (HO-1) protein expression and NFκB activation. Treatment with TEMPOL abated decreased gene expression of DRD2S and DRD2L induced by 6-OHDA indicating that TEMPOL may prevent mitochondrial dysfunction and activation of pathways that result in receptor desensitization. 6-OHDA insult decreased gene expression of the antioxidant, SOD-1, and this diminution was also mitigated by TEMPOL. Activation of NFκB increased pro-inflammatory IFNy and decreased IL-6, however, TEMPOL had no effect on these inflammation mediators. Overall, this data suggests that cyclic nitroxides may preserve dopaminergic neuronal cell viability by attenuating oxidative stress and mitochondrial dysfunction, but are unable to affect inflammatory mediators that propagate cellular damage and neurodegeneration in PD.
Collapse
|
55
|
The Pressing Issue of Micro- and Nanoplastic Contamination: Profiling the Reproductive Alterations Mediated by Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11020193. [PMID: 35204076 PMCID: PMC8868557 DOI: 10.3390/antiox11020193] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 01/25/2023] Open
Abstract
Micro- and nanoplastics (MPs/NPs) are among the most widely distributed pollutants in the environment. It has been suggested that exposure to MPs/NPs can trigger toxicity pathways among which inflammation and oxidative stress (OS) play a pivotal role. Once absorbed, MPs/NPs may act locally or access the bloodstream and, following the translocation process, reach several organs and tissues, including the gonads. Notably, MPs/NPs can bioaccumulate in human and murine placenta, opening new scenarios for toxicological evaluations. We review recent studies on the effects of MPs/NPs on the reproductive health in aquatic and terrestrial organisms of both sexes, focusing on the role of OS and the antioxidant defence system failure as the main underlying mechanisms. Alterations in gametogenesis, embryonic and offspring development, and survival have been shown in most studies and often related to a broken redox balance. All these detrimental effects are inversely related to particle size, whereas they are closely linked to shape, plastic polymer type, superficial functionalization, concentration, and time of exposure. To date, the studies provide insights into the health impacts, but no conclusions can be drawn for reproduction toxicity. The main implication of the few studies on antioxidant substances reveals their potential role in mitigating MP-induced toxic effects.
Collapse
|
56
|
Søderstrøm S, Lie KK, Lundebye AK, Søfteland L. Beauvericin (BEA) and enniatin B (ENNB)-induced impairment of mitochondria and lysosomes - Potential sources of intracellular reactive iron triggering ferroptosis in Atlantic salmon primary hepatocytes. Food Chem Toxicol 2022; 161:112819. [PMID: 35038498 DOI: 10.1016/j.fct.2022.112819] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022]
Abstract
Beauvericin (BEA) and enniatin B (ENNB) are emerging mycotoxins frequently detected in plant-based fish feed. With ionophoric properties, they have shown cytotoxic potential in mammalian models. Sensitivity in fish is still largely unknown. Primary hepatocytes isolated from Atlantic salmon (Salmo salar) were used as a model and exposed to BEA and ENNB (0.05-10 μM) for 48 h. Microscopy, evaluation of cell viability, total ATP, total H2O2, total iron content, total Gpx enzyme activity, and RNA sequencing were used to characterize the toxicodynamics of BEA and ENNB. Both mycotoxins became cytotoxic at ≥ 5 μM, causing condensation of the hepatocytes followed by formation of blister-like protrusions on the cell's membrane. RNA sequencing analysis at sub-cytotoxic levels indicated BEA and ENNB exposed hepatocytes to experience increased energy expenditure, elevated oxidative stress, and iron homeostasis disturbances sensitizing the hepatocytes to ferroptosis. The present study provides valuable knowledge disclosing the toxic action of these mycotoxins in Atlantic salmon primary hepatocytes.
Collapse
Affiliation(s)
| | - Kai K Lie
- Institute of Marine Research (IMR), Bergen, Norway
| | | | | |
Collapse
|
57
|
Hara Y, Oshima Y, Tagami Y, Aoki A, Fujii H, Izawa A, Seki K, Kanai A, Yabe A, Watanabe K, Horita N, Kobayashi N, Kaneko T. Clinical importance of serum heme oxygenase-1 measurement in patients with acute exacerbation of idiopathic pulmonary fibrosis triggered by coronavirus disease 2019. Respir Med Case Rep 2022; 36:101615. [PMID: 35223424 PMCID: PMC8858429 DOI: 10.1016/j.rmcr.2022.101615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/15/2022] [Accepted: 02/17/2022] [Indexed: 11/26/2022] Open
|
58
|
Wang B, Liu P, Huang H, Wang X, Zhang M, Huang J, Lu F, Chen J, Liu Y, Kang Z. Carbon dots up-regulate heme oxygenase-1 expression towards acute lung injury therapy. J Mater Chem B 2021; 9:9005-9011. [PMID: 34617947 DOI: 10.1039/d1tb01283e] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pneumonia is a kind of inflammation, which can cause high morbidity and mortality, and the treatment of pneumonia has received widespread attention. Heme oxygenase-1 (HMOX1) is a cell protective enzyme and can generate an anti-inflammatory response. Here, we demonstrate that degradable carbon dots (from L-ascorbic acid, CDs-1) can up-regulate the expression of HMOX1 in animal cells and tissues, which has a therapeutic effect on LPS-induced acute lung injury in mice. It was confirmed from in vitro experiments that CDs-1 could significantly up-regulate the expression of mRNA and the protein of HMOX1, which can increase the expression of HMOX1 by 5 times in a short time, decreasing the reactive oxygen species level in a cellular inflammation model induced by LPS. Furthermore, a series of in vivo comparative experiments show that CDs-1 could effectively treat acute lung injury and improve the survival rate of mice to 80%. Our work provides a practical way for the treatment of acute inflammation and the promising application of CDs in anti-inflammation.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Peipei Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Hui Huang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Xiting Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Mengling Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Jian Huang
- School of Biology & Basic Medical Science, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China
| | - Fang Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Jian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China. .,Chinese Institute for Brain Research, Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing, 102206, China.
| | - Yang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Zhenhui Kang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China. .,Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa 999078, Macau SAR, China
| |
Collapse
|
59
|
Floresta G, Fallica AN, Salerno L, Sorrenti V, Pittalà V, Rescifina A. Growing the molecular architecture of imidazole-like ligands in HO-1 complexes. Bioorg Chem 2021; 117:105428. [PMID: 34710668 DOI: 10.1016/j.bioorg.2021.105428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/27/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022]
Abstract
Up-regulation of HO-1 had been frequently reported in different cases and types of human malignancies. Since poor clinical outcomes are reported in these cases, this enzyme's inhibition is considered a valuable and proven anticancer approach. To identify novel HO-1 inhibitors suitable for drug development, we report a structure-guided fragment-based approach to identify new lead compounds. Different parts of the selected molecules were analyzed, and the different series of novel compounds were virtually evaluated. The growing experiments of the classical HO-1 inhibitors structure led us to different hit-compounds. A synthetic pathway for six selected molecules was designed, and the compounds were synthesized. The biological activity revealed that molecules 10 and 12 inhibit the HO-1 activity with an IC50 of 1.01 and 0.90 μM, respectively. This study suggested that our growing approach was successful, and these results are ongoing for further development.
Collapse
Affiliation(s)
- Giuseppe Floresta
- Department of Analytical, Environmental and Forensic Sciences, King's College London, London, UK.
| | - Antonino N Fallica
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy.
| | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| |
Collapse
|
60
|
Snijders KE, Fehér A, Táncos Z, Bock I, Téglási A, van den Berk L, Niemeijer M, Bouwman P, Le Dévédec SE, Moné MJ, Van Rossom R, Kumar M, Wilmes A, Jennings P, Verfaillie CM, Kobolák J, Ter Braak B, Dinnyés A, van de Water B. Fluorescent tagging of endogenous Heme oxygenase-1 in human induced pluripotent stem cells for high content imaging of oxidative stress in various differentiated lineages. Arch Toxicol 2021; 95:3285-3302. [PMID: 34480604 PMCID: PMC8448683 DOI: 10.1007/s00204-021-03127-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/27/2021] [Indexed: 12/28/2022]
Abstract
Tagging of endogenous stress response genes can provide valuable in vitro models for chemical safety assessment. Here, we present the generation and application of a fluorescent human induced pluripotent stem cell (hiPSC) reporter line for Heme oxygenase-1 (HMOX1), which is considered a sensitive and reliable biomarker for the oxidative stress response. CRISPR/Cas9 technology was used to insert an enhanced green fluorescent protein (eGFP) at the C-terminal end of the endogenous HMOX1 gene. Individual clones were selected and extensively characterized to confirm precise editing and retained stem cell properties. Bardoxolone-methyl (CDDO-Me) induced oxidative stress caused similarly increased expression of both the wild-type and eGFP-tagged HMOX1 at the mRNA and protein level. Fluorescently tagged hiPSC-derived proximal tubule-like, hepatocyte-like, cardiomyocyte-like and neuron-like progenies were treated with CDDO-Me (5.62–1000 nM) or diethyl maleate (5.62–1000 µM) for 24 h and 72 h. Multi-lineage oxidative stress responses were assessed through transcriptomics analysis, and HMOX1-eGFP reporter expression was carefully monitored using live-cell confocal imaging. We found that eGFP intensity increased in a dose-dependent manner with dynamics varying amongst lineages and stressors. Point of departure modelling further captured the specific lineage sensitivities towards oxidative stress. We anticipate that the newly developed HMOX1 hiPSC reporter will become a valuable tool in understanding and quantifying critical target organ cell-specific oxidative stress responses induced by (newly developed) chemical entities.
Collapse
Affiliation(s)
- Kirsten E Snijders
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | | | | | | | | | - Linda van den Berk
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Marije Niemeijer
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Peter Bouwman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Sylvia E Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Martijn J Moné
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Rob Van Rossom
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Manoj Kumar
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Amsterdam, The Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Amsterdam, The Netherlands
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | | | - Bas Ter Braak
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - András Dinnyés
- BioTalentum Ltd., 2100, Gödöllő, Hungary. .,Department of Physiology and Animal Health, Institute of Physiology and Animal Health, Hungarian University of Agriculture and Life Sciences, 2100, Gödöllő, Hungary.
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
61
|
Ciaffaglione V, Modica MN, Pittalà V, Romeo G, Salerno L, Intagliata S. Mutual Prodrugs of 5-Fluorouracil: From a Classic Chemotherapeutic Agent to Novel Potential Anticancer Drugs. ChemMedChem 2021; 16:3496-3512. [PMID: 34415107 PMCID: PMC9290623 DOI: 10.1002/cmdc.202100473] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/18/2021] [Indexed: 12/18/2022]
Abstract
The development of potent antitumor agents with a low toxicological profile against healthy cells is still one of the greatest challenges facing medicinal chemistry. In this context, the “mutual prodrug” approach has emerged as a potential tool to overcome undesirable physicochemical features and mitigate the side effects of approved drugs. Among broad‐spectrum chemotherapeutics available for clinical use today, 5‐fluorouracil (5‐FU) is one of the most representative, also included in the World Health Organization model list of essential medicines. Unfortunately, severe side effects and drug resistance phenomena are still the primary limits and drawbacks in its clinical use. This review describes the progress made over the last ten years in developing 5‐FU‐based mutual prodrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues.
Collapse
Affiliation(s)
- Valeria Ciaffaglione
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Maria N Modica
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
62
|
Reyes-Ramos CA, Gaxiola-Robles R, Vázquez-Medina JP, Ramírez-Jirano LJ, Bitzer-Quintero OK, Zenteno-Savín T. In silico Characterization of the Heme Oxygenase 1 From Bottlenose Dolphin ( Tursiops truncatus): Evidence of Changes in the Active Site and Purifying Selection. Front Physiol 2021; 12:711645. [PMID: 34456750 PMCID: PMC8388933 DOI: 10.3389/fphys.2021.711645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/12/2021] [Indexed: 11/13/2022] Open
Abstract
Cetacea is a clade well-adapted to the aquatic lifestyle, with diverse adaptations and physiological responses, as well as a robust antioxidant defense system. Serious injuries caused by boats and fishing nets are common in bottlenose dolphins (Tursiops truncatus); however, these animals do not show signs of serious infections. Evidence suggests an adaptive response to tissue damage and associated infections in cetaceans. Heme oxygenase (HO) is a cytoprotective protein that participates in the anti-inflammatory response. HO catalyzes the first step in the oxidative degradation of the heme group. Various stimuli, including inflammatory mediators, regulate the inducible HO-1 isoform. This study aims to characterize HO-1 of the bottlenose dolphin in silico and compare its structure to the terrestrial mammal protein. Upstream HO-1 sequence of the bottlenose dolphin was obtained from NCBI and Ensemble databases, and the gene structure was determined using bioinformatics tools. Five exons and four introns were identified, and proximal regulatory elements were detected in the upstream region. The presence of 10 α-helices, three 310 helices, the heme group lodged between the proximal and distal helices, and a histidine-25 in the proximal helix serving as a ligand to the heme group were inferred for T. truncatus. Amino acid sequence alignment suggests HO-1 is a conserved protein. The HO-1 "fingerprint" and histidine-25 appear to be fully conserved among all species analyzed. Evidence of positive selection within an α-helix configuration without changes in protein configuration and evidence of purifying selection were found, indicating evolutionary conservation of the coding sequence structure.
Collapse
Affiliation(s)
- Carlos A. Reyes-Ramos
- Centro de Investigaciones Biológicas del Noroeste, S.C. Planeación Ambiental y Conservación, La Paz, Mexico
| | - Ramón Gaxiola-Robles
- Centro de Investigaciones Biológicas del Noroeste, S.C. Planeación Ambiental y Conservación, La Paz, Mexico
- Hospital General de Zona No. 1, Instituto Mexicano del Seguro Social, La Paz, Mexico
| | | | - Luis Javier Ramírez-Jirano
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | - Oscar Kurt Bitzer-Quintero
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | - Tania Zenteno-Savín
- Centro de Investigaciones Biológicas del Noroeste, S.C. Planeación Ambiental y Conservación, La Paz, Mexico
| |
Collapse
|
63
|
Bhurtel S, Bok E, Katila N, Kim J, Choi DY. Activation of Nrf2 by methylene blue is associated with the neuroprotection against MPP + induced toxicity via ameliorating oxidative stress and mitochondrial dysfunction. Biochem Pharmacol 2021; 192:114719. [PMID: 34352280 DOI: 10.1016/j.bcp.2021.114719] [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: 07/28/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 01/18/2023]
Abstract
The neuropathological hallmark of Parkinson's disease (PD) is the preferential loss of dopaminergic neurons in the substantia nigra and presence of Lewy bodies in the dying neurons. Though specific molecular mechanisms for the neurodegeneration remains to be clarified, mitochondrial dysfunction and increased oxidative stress are major players associated with PD pathogenesis and these pathogenic mechanisms can be reproduced in cells and animals by application of various neurotoxins such as MPP+. In this study, we attempted to determine the neuroprotective effects of methylene blue (MB) against 1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity, and to elucidate its action mechanism. We observed that MB attenuated MPP+-induced apoptotic cell death in SH-SY5Y cells and the mescencephalic dopaminergic neurons. In addition, MB protected the cells against MPP+-induced oxidative stress and mitochondrial dysfunction as evidenced by restoration of mitochondrial complex I activity and ATP levels, and attenuation of oxidative stress. Moreover, we demonstrated that MB induced antioxidant molecules, and activated Nrf2 pathway through AKT activation. These results indicate that MB protects the neurons from MPP+-induced toxicity through activation of antioxidant system, thereby reducing the oxidative stress and mitochondrial impairment, implying the potential use of MB in the treatment of neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
- Sunil Bhurtel
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Eugene Bok
- Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Nikita Katila
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Jaekwang Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu 41062, Republic of Korea.
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
64
|
Steimer M, Kaiser S, Ulbrich F, Kalbhenn J, Bürkle H, Schallner N. Expression of HO1 and PER2 can predict the incidence of delirium in trauma patients with concomitant brain injury. Sci Rep 2021; 11:15388. [PMID: 34321570 PMCID: PMC8319290 DOI: 10.1038/s41598-021-94773-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 07/16/2021] [Indexed: 11/09/2022] Open
Abstract
Intensive care unit (ICU)-acquired delirium is associated with adverse outcome in trauma patients with concomitant traumatic brain injury (TBI), but diagnosis remains challenging. Quantifying circadian disruption by analyzing expression of the circadian gene period circadian regulator 2 (PER2) and heme oxygenase 1 (HO1), which determines heme turnover, may prove to be potential diagnostic tools. Expression of PER2 and HO1 was quantified using qPCR from blood samples 1 day and 7 days after trauma. Association analysis was performed comparing mRNA expression levels with parameters of trauma (ISS—injury severity score), delirium, acute kidney injury (AKI) and length of ICU stay. 48 polytraumatized patients were included (equal distribution of TBI versus non-TBI) corrected for ISS, age and gender using a matched pairs approach. Expression levels of PER2 and HO1 were independent of age (PER2: P = 0.935; HO1: P = 0.988), while expression levels were significantly correlated with trauma severity (PER2: P = 0.009; HO1: P < 0.001) and longer ICU length of stay (PER2: P = 0.018; HO1: P < 0.001). High expression levels increased the odds of delirium occurrence (PER2: OR = 4.32 [1.14–13.87]; HO1: OR = 4.50 [1.23–14.42]). Patients with TBI showed a trend towards elevated PER2 (OR = 3.00 [0.84–9.33], P = 0.125), but not towards delirium occurrence (P = 0.556). TBI patients were less likely to develop AKI compared to non-TBI (P = 0.022). Expression levels of PER2 and HO1 correlate with the incidence of delirium in an age-independent manner and may potentially improve diagnostic algorithms when used as delirium biomarkers. Trial registration: German Clinical Trials Register (Trial-ID DRKS00008981; Universal Trial Number U1111-1172-6077; Jan. 18, 2018).
Collapse
Affiliation(s)
- Matti Steimer
- Department of Anesthesiology & Critical Care Medicine, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sandra Kaiser
- Department of Anesthesiology & Critical Care Medicine, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Ulbrich
- Department of Anesthesiology & Critical Care Medicine, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Johannes Kalbhenn
- Department of Anesthesiology & Critical Care Medicine, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Bürkle
- Department of Anesthesiology & Critical Care Medicine, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care Medicine, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany. .,Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
65
|
Takemoto M, Sakamoto H, Higashimoto Y, Taira J. Complex Formation of Heme Oxygenase-2 with Heme Is Competitively Inhibited by the Cytosolic Domain of Caveolin-1. Biochemistry 2021; 60:2300-2308. [PMID: 34223768 DOI: 10.1021/acs.biochem.1c00247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mechanism and physiological functions of heme oxygenase-2 (HO-2)-mediated carbon monoxide (CO) production, accompanied by heme metabolism, have been studied intensively in recent years. The enzymatic activity of constitutively expressed HO-2 must be strictly controlled in terms of the toxicity and chemical stability of CO. In this study, the molecular interaction between HO-2 and caveolin-1 and its effect on HO action were evaluated. An enzyme kinetics assay with residues 82-101 of caveolin-1, also called the caveolin scaffold domain, inhibited HO-2 activity in a competitive manner. Analytical ultracentrifugation and a hemin titration assay suggested that the inhibitory effect was generated by direct binding of caveolin-1 to aromatic residues, which were defined as components of the caveolin-binding motif in the HO-2 heme pocket. Herein, we developed a HO-2-based fluorescence bioprobe, namely EGFP-Δ19/D159H, which was capable of quantifying heme binding by HO-2 as the initial step in the CO production. The fluorescence of EGFP-Δ19/D159H decreased in accordance with 5-aminolevulinic acid-facilitated heme biosynthesis in COS-7 cells. In contrast, expression of the N-terminal cytosolic domain of caveolin-1 (residues 1-101) increased the probe fluorescence, suggesting that the cytosolic domain of caveolin-1 potently inhibits the binding of heme to the heme pocket of EGFP-Δ19/D159H. Taken together, our results suggest that caveolin-1 is a negative regulator of HO-2 enzymatic action. Moreover, our bioprobe EGFP-Δ19/D159H represents a powerful tool for use in future studies addressing HO-2-mediated CO production.
Collapse
Affiliation(s)
- Misaki Takemoto
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| | - Hiroshi Sakamoto
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan
| | - Yuichiro Higashimoto
- Department of Chemistry, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Junichi Taira
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Iizuka 820-8502, Japan.,Department of Chemistry, Kurume University School of Medicine, Kurume 830-0011, Japan
| |
Collapse
|
66
|
Salat APJ, Williams KL, Chiu S, Eickmeyer DC, Kimpe LE, Blais JM, Crump D. Extracts from Dated Lake Sediment Cores in the Athabasca Oil Sands Region Alter Ethoxyresorufin-O-deethylase Activity and Gene Expression in Avian Hepatocytes. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:1883-1893. [PMID: 33751657 DOI: 10.1002/etc.5040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/12/2021] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
Increases in oil sands mining operations in the Athabasca oil sands region have resulted in increased concentrations of polycyclic aromatic compounds (PACs) and heavy metals in aquatic systems located near surface mining operations. In the present study, sediment cores were collected from 3 lakes with varying proximity to surface mining operations to determine the differences in PAC concentrations. Sediment cores were separated into 2 sections-current mining (top; 2000-2017) and premining (bottom; pre-1945)-and extracts were prepared for in vitro screening using a well-established chicken embryonic hepatocyte (CEH) assay. Concentrations and composition of PACs varied between sites, with the highest ∑PACs in Saline Lake, 5 km from an active oil sands mine site. The proportion of alkylated PACs was greater than that of parent PACs in the top sediment sections compared with the bottom. Ethoxyresorufin-O-deethylase activity in CEH permitted the ranking of lake sites/core sections based on an aryl hydrocarbon receptor-mediated end point; mean median effect concentration values were lowest for the top cores from Saline Lake and another near-mining operations lake, referred to as WF1. A ToxChip polymerase chain reaction (PCR) array was used to evaluate gene expression changes across 43 target genes associated with numerous toxicological pathways following exposure to top and bottom sediment core extracts. The 2 study sites with the greatest ∑PAC concentrations (Saline Lake and WF1) had the highest gene expression alterations on the ToxChip PCR array (19 [top] and 17 [bottom]/43), compared with a reference site (13 [top] and 7 [bottom]/43). The avian in vitro bioassay was useful for identifying the toxicity of complex PAC extracts associated with variably contaminated sediment cores, supporting its potential use for hotspot identification and complex mixture screening. EnvironToxicol Chem 2021;40:1883-1893. © 2021 SETAC.
Collapse
Affiliation(s)
| | - Kim L Williams
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University, Ottawa, Ontario, Canada
| | - Suzanne Chiu
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University, Ottawa, Ontario, Canada
| | - David C Eickmeyer
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Linda E Kimpe
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jules M Blais
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Doug Crump
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, National Wildlife Research Centre, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
67
|
Zhang W, Chen L, Xiong Y, Panayi AC, Abududilibaier A, Hu Y, Yu C, Zhou W, Sun Y, Liu M, Xue H, Hu L, Yan C, Xie X, Lin Z, Cao F, Mi B, Liu G. Antioxidant Therapy and Antioxidant-Related Bionanomaterials in Diabetic Wound Healing. Front Bioeng Biotechnol 2021; 9:707479. [PMID: 34249895 PMCID: PMC8264455 DOI: 10.3389/fbioe.2021.707479] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/03/2021] [Indexed: 12/23/2022] Open
Abstract
Ulcers are a lower-extremity complication of diabetes with high recurrence rates. Oxidative stress has been identified as a key factor in impaired diabetic wound healing. Hyperglycemia induces an accumulation of intracellular reactive oxygen species (ROS) and advanced glycation end products, activation of intracellular metabolic pathways, such as the polyol pathway, and PKC signaling leading to suppression of antioxidant enzymes and compounds. Excessive and uncontrolled oxidative stress impairs the function of cells involved in the wound healing process, resulting in chronic non-healing wounds. Given the central role of oxidative stress in the pathology of diabetic ulcers, we performed a comprehensive review on the mechanism of oxidative stress in diabetic wound healing, focusing on the progress of antioxidant therapeutics. We summarize the antioxidant therapies proposed in the past 5 years for use in diabetic wound healing, including Nrf2- and NFκB-pathway-related antioxidant therapy, vitamins, enzymes, hormones, medicinal plants, and biological materials.
Collapse
Affiliation(s)
- Wenqian Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Lang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Abudula Abududilibaier
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yiqiang Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Chenyan Yu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Wu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yun Sun
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China.,Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Hang Xue
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Liangcong Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Chenchen Yan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xuedong Xie
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ze Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Faqi Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
68
|
Carbon Monoxide Releasing Molecule-3 Enhances Heme Oxygenase-1 Induction via ROS-Dependent FoxO1 and Nrf2 in Brain Astrocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5521196. [PMID: 34194603 PMCID: PMC8214505 DOI: 10.1155/2021/5521196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022]
Abstract
Carbon monoxide releasing molecule-3 (CORM-3) has been shown to protect inflammatory diseases via the upregulation of heme oxygenases-1 (HO-1). However, in rat brain astrocytes (RBA-1), the mechanisms underlying CORM-3-induced HO-1 remain poorly defined. This study used western blot, real-time PCR, and promoter activity assays to determine the levels of HO-1 expression and 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) and dihydroethidium (DHE) to measure reactive oxygen species (ROS). We found that CORM-3-induced HO-1 expression was mediated through ROS generation by Nox or mitochondria. The signaling components were differentiated by pharmacological inhibitors and small interfering RNA (siRNA). Subcellular fractions, immunofluorescent staining, and chromatin immunoprecipitation assay were used to evaluate the nuclear translocation and promoter binding activity of Nrf2 induced by CORM-3. The roles of mTOR and FoxO1 in CORM-3-stimulated responses are still unknown in RBA-1 cells. Our results demonstrated that transfection with siRNAs or pretreatment with pharmacological inhibitors attenuated the levels of HO-1 and phosphorylation of signaling components including Akt, mTOR, FoxO1, and Nrf2 stimulated by CORM-3. Moreover, pretreatment with N-acetyl-L-cysteine, diphenyleneiodonium chloride, apocynin, or rotenone blocked nuclear translocation and promoter binding activity of Nrf2 induced by CORM-3. The present study concluded that in RBA-1 cells, CORM-3-induced HO-1 expression is, at least partially, mediated through Nox and mitochondria/ROS-dependent PI3K/Akt/mTOR cascade to activate FoxO1 or ROS leading to activation of Nrf2 activity.
Collapse
|
69
|
Akagi R, Kubo T, Hatori Y, Miyamoto T, Inouye S. Heme oxygenase-1 induction by heat shock in rat hepatoma cell line is regulated by the coordinated function of HSF1, NRF2, AND BACH1. J Biochem 2021; 170:501-510. [PMID: 34061198 DOI: 10.1093/jb/mvab065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/25/2021] [Indexed: 11/12/2022] Open
Abstract
The mechanism of heme oxygenase-1 (HO-1) induction by heat shock (HS) loading remains unclear. Here, we investigated the contribution of transcription factors to HS-induced HO-1 expression, using a rat hepatoma cell line (H-4-II-E). Our results demonstrated that HS treatment resulted in a marked induction of HO-1. Immunohistochemical analysis showed a slight mismatch in the expression levels of HO-1 and HSP70 by HS among cells, suggesting a conflict between multiple induction mechanisms. We observed HS-induced nuclear localization of, not only phosphorylated HSF1, but also NRF2, which is a typical transcription factor activated by oxidative stress. HSF1 knockdown in H-4-II-E markedly reduced HO-1 induction by HS, while NRF2 knockdown resulted in a partial effect. The chromatin immunoprecipitation assay demonstrated that HS loading resulted in significant binding of HSF1 to the HSE in the promoter proximal region of HO-1 gene and another HSE located close to the MARE in the -4 kb upstream enhancer region 1, where NRF2 also bound, together with BACH1, a negative transcription factor of HO-1. These observations indicate that HO-1 induction by HS is mainly mediated by HSF1 binding to the proximal HSE. NRF2 binding to MARE by HS is predominantly suppressed by an increased binding of BACH1.
Collapse
Affiliation(s)
- Reiko Akagi
- Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan
| | - Takanori Kubo
- Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan
| | - Yuta Hatori
- Department of Pharmacy, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima 731-0153, Japan
| | - Takafumi Miyamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba-City, Ibaraki 305-8575, Japan
| | - Sachiye Inouye
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University 1-1-1 Daigakudohri, Sanyo-onoda-shi 756-0884, Japan
| |
Collapse
|
70
|
Belperain S, Kang ZY, Dunphy A, Priebe B, Chiu NHL, Jia Z. Anti-Inflammatory Effect and Cellular Uptake Mechanism of Carbon Nanodots in in Human Microvascular Endothelial Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1247. [PMID: 34068511 PMCID: PMC8151002 DOI: 10.3390/nano11051247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 11/25/2022]
Abstract
Cardiovascular disease (CVD) has become an increasingly important topic in the field of medical research due to the steadily increasing rates of mortality caused by this disease. With recent advancements in nanotechnology, a push for new, novel treatments for CVD utilizing these new materials has begun. Carbon Nanodots (CNDs), are a new form of nanoparticles that have been coveted due to the green synthesis method, biocompatibility, fluorescent capabilities and potential anti-antioxidant properties. With much research pouring into CNDs being used as bioimaging and drug delivery tools, few studies have been completed on their anti-inflammatory potential, especially in the cardiovascular system. CVD begins initially by endothelial cell inflammation. The cause of this inflammation can come from many sources; one being tumor necrosis factor (TNF-α), which can not only trigger inflammation but prolong its existence by causing a storm of pro-inflammatory cytokines. This study investigated the ability of CNDs to attenuate TNF-α induced inflammation in human microvascular endothelial cells (HMEC-1). Results show that CNDs at non-cytotoxic concentrations reduce the expression of pro-inflammatory genes, mainly Interleukin-8 (IL-8), and interleukin 1 beta (IL-1β). The uptake of CNDs by HMEC-1s was examined. Results from the studies involving channel blockers and endocytosis disruptors suggest that uptake takes place by endocytosis. These findings provide insights on the interaction CNDs and endothelial cells undergoing TNF-α induced cellular inflammation.
Collapse
Affiliation(s)
- Sarah Belperain
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Zi Yae Kang
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Andrew Dunphy
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Brandon Priebe
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| | - Norman H. L. Chiu
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27412, USA;
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC 27401, USA
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27412, USA; (S.B.); (Z.Y.K.); (A.D.); (B.P.)
| |
Collapse
|
71
|
Xiong R, Wu Y, Wu Q, Muskhelishvili L, Davis K, Tripathi P, Chen Y, Chen T, Bryant M, Rosenfeldt H, Healy SM, Cao X. Integration of transcriptome analysis with pathophysiological endpoints to evaluate cigarette smoke toxicity in an in vitro human airway tissue model. Arch Toxicol 2021; 95:1739-1761. [PMID: 33660061 PMCID: PMC8113308 DOI: 10.1007/s00204-021-03008-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/16/2021] [Indexed: 01/04/2023]
Abstract
Exposure to cigarette smoke (CS) is a known risk factor in the pathogenesis of smoking-caused diseases, such as chronic obstructive pulmonary diseases (COPD) and lung cancer. To assess the effects of CS on the function and phenotype of airway epithelial cells, we developed a novel repeated treatment protocol and comprehensively evaluated the progression of key molecular, functional, and structural abnormalities induced by CS in a human in vitro air-liquid-interface (ALI) airway tissue model. Cultures were exposed to CS (diluted with 0.5 L/min, 1.0 L/min, and 4.0 L/min clean air) generated from smoking five 3R4F University of Kentucky reference cigarettes under the International Organization for Standardization (ISO) machine smoking regimen, every other day for 4 weeks (3 days per week, 40 min/day). By integrating the transcriptomics-based approach with the in vitro pathophysiological measurements, we demonstrated CS-mediated effects on oxidative stress, pro-inflammatory cytokines and matrix metalloproteinases (MMPs), ciliary function, expression and secretion of mucins, and squamous cell differentiation that are highly consistent with abnormalities observed in airways of smokers. Enrichment analysis on the transcriptomic profiles of the ALI cultures revealed key molecular pathways, such as xenobiotic metabolism, oxidative stress, and inflammatory responses that were perturbed in response to CS exposure. These responses, in turn, may trigger aberrant tissue remodeling, eventually leading to the onset of respiratory diseases. Furthermore, changes of a panel of genes known to be disturbed in smokers with COPD were successfully reproduced in the ALI cultures exposed to CS. In summary, findings from this study suggest that such an integrative approach may be a useful tool for identifying genes and adverse cellular events caused by inhaled toxicants, like CS.
Collapse
Affiliation(s)
- Rui Xiong
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Yue Wu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | | | - Kelly Davis
- Toxicologic Pathology Associates, Jefferson, AR, 72079, USA
| | - Priya Tripathi
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Ying Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Matthew Bryant
- Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Hans Rosenfeldt
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Sheila M Healy
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
72
|
Danielak A, Wallace JL, Brzozowski T, Magierowski M. Gaseous Mediators as a Key Molecular Targets for the Development of Gastrointestinal-Safe Anti-Inflammatory Pharmacology. Front Pharmacol 2021; 12:657457. [PMID: 33995080 PMCID: PMC8116801 DOI: 10.3389/fphar.2021.657457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) represent one of the most widely used classes of drugs and play a pivotal role in the therapy of numerous inflammatory diseases. However, the adverse effects of these drugs, especially when applied chronically, frequently affect gastrointestinal (GI) tract, resulting in ulceration and bleeding, which constitutes a significant limitation in clinical practice. On the other hand, it has been recently discovered that gaseous mediators nitric oxide (NO), hydrogen sulfide (H2S) and carbon monoxide (CO) contribute to many physiological processes in the GI tract, including the maintenance of GI mucosal barrier integrity. Therefore, based on the possible therapeutic properties of NO, H2S and CO, a novel NSAIDs with ability to release one or more of those gaseous messengers have been synthesized. Until now, both preclinical and clinical studies have shown promising effects with respect to the anti-inflammatory potency as well as GI-safety of these novel NSAIDs. This review provides an overview of the gaseous mediators-based NSAIDs along with their mechanisms of action, with special emphasis on possible implications for GI mucosal defense mechanisms.
Collapse
Affiliation(s)
- Aleksandra Danielak
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - John L Wallace
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
73
|
Cai J, Zang X, Wu Z, Liu J, Wang D. Altered protein S-glutathionylation depicts redox imbalance triggered by transition metal oxide nanoparticles in a breastfeeding system. NANOIMPACT 2021; 22:100305. [PMID: 35559962 DOI: 10.1016/j.impact.2021.100305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/25/2021] [Accepted: 02/19/2021] [Indexed: 06/15/2023]
Abstract
Nanosafety has become a public concern following nanotechnology development. By now, attention has seldom been paid to breastfeeding system, which is constructed by mammary physiological structure and derived substances (endogenous or exogenous), cells, tissues, organs, and individuals (mother and child), connecting environment and organism, and spans across mother-child dyad. Thus, breastfeeding system is a center of nutrients transport and a unique window of toxic susceptibility in the mother-child dyad. We applied metabolomics combined with redox proteomics to depict how nanoparticles cause metabolic burden via their spontaneous redox cycling in lactating mammary glands. Two widely used nanoparticles [titanium dioxide (nTiO2) and zinc oxide (nZnO)] were exposed to lactating mice via intranasal administration. Biodistribution and biopersistence of nTiO2 and nZnO in mammary glands destroyed its structure, reflective of significantly reduced claudin-3 protein level by 32.1% (P < 0.01) and 47.8% (P < 0.01), and significantly increased apoptosis index by 85.7 (P < 0.01) and 100.3 (P < 0.01) fold change, respectively. Airway exposure of nTiO2 trended to reduced milk production by 22.7% (P = 0.06), while nZnO significantly reduced milk production by 33.0% (P < 0.01). Metabolomics analysis revealed a metabolic shift by nTiO2 or nZnO, such as increased glycolysis (nTiO2: fold enrichment = 3.31, P < 0.05; nZnO: fold enrichment = 3.68, P < 0.05), glutathione metabolism (nTiO2: fold enrichment = 5.57, P < 0.01; nZnO: fold enrichment = 4.43, P < 0.05), and fatty acid biosynthesis (nTiO2: fold enrichment = 3.52, P < 0.05; nZnO: fold enrichment = 3.51, P < 0.05) for tissue repair at expense of lower milk fat synthesis (35.7% reduction by nTiO2; 51.8% reduction by nZnO), and finally led to oxidative stress of mammary glands. The increased GSSG/GSH ratio (57.5% increase by nTiO2; 105% increase by nZnO) with nanoparticle exposure confirmed an alteration in the redox state and a metabolic shift in mammary glands. Redox proteomics showed that nanoparticles induced S-glutathionylation (SSG) modification at Cys sites of proteins in a nanoparticle type-dependent manner. The nTiO2 induced more protein SSG modification sites (nTiO2: 21; nZnO:16), whereas nZnO induced fewer protein SSG modification sites but at deeper SSG levels (26.6% higher in average of nZnO than that of nTiO2). In detail, SSG modification by nTiO2 was characterized by Ltf at Cys423 (25.3% increase), and Trf at Cys386;395;583 (42.3%, 42.3%, 22.8% increase) compared with control group. While, SSG modification by nZnO was characterized by Trfc at Cys365 (71.3% increase) and Fasn at Cys1010 (41.0% increase). The discovery of SSG-modified proteins under airway nanoparticle exposure further supplemented the oxidative stress index and mammary injury index, and deciphered precise mechanisms of nanotoxicity into a molecular level. The unique quantitative site-specific redox proteomics and metabolomics can serve as a new technique to identify nanotoxicity and provide deep insights into nanoparticle-triggered oxidative stress, contributing to a healthy breastfeeding environment.
Collapse
Affiliation(s)
- Jie Cai
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Xinwei Zang
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Zezhong Wu
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China
| | - Jianxin Liu
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Diming Wang
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| |
Collapse
|
74
|
Okuma Y, Becker LB, Hayashida K, Aoki T, Saeki K, Nishikimi M, Shoaib M, Miyara SJ, Yin T, Shinozaki K. Effects of Post-Resuscitation Normoxic Therapy on Oxygen-Sensitive Oxidative Stress in a Rat Model of Cardiac Arrest. J Am Heart Assoc 2021; 10:e018773. [PMID: 33775109 PMCID: PMC8174361 DOI: 10.1161/jaha.120.018773] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Cardiac arrest (CA) can induce oxidative stress after resuscitation, which causes cellular and organ damage. We hypothesized that post‐resuscitation normoxic therapy would protect organs against oxidative stress and improve oxygen metabolism and survival. We tested the oxygen‐sensitive reactive oxygen species from mitochondria to determine the association with hyperoxia‐induced oxidative stress. Methods and Results Sprague–Dawley rats were subjected to 10‐minute asphyxia‐induced CA with a fraction of inspired O2 of 0.3 or 1.0 (normoxia versus hyperoxia, respectively) after resuscitation. The survival rate at 48 hours was higher in the normoxia group than in the hyperoxia group (77% versus 28%, P<0.01), and normoxia gave a lower neurological deficit score (359±140 versus 452±85, P<0.05) and wet to dry weight ratio (4.6±0.4 versus 5.6±0.5, P<0.01). Oxidative stress was correlated with increased oxygen levels: normoxia resulted in a significant decrease in oxidative stress across multiple organs and lower oxygen consumption resulting in normalized respiratory quotient (0.81±0.05 versus 0.58±0.03, P<0.01). After CA, mitochondrial reactive oxygen species increased by ≈2‐fold under hyperoxia. Heme oxygenase expression was also oxygen‐sensitive, but it was paradoxically low in the lung after CA. In contrast, the HMGB‐1 (high mobility group box‐1) protein was not oxygen‐sensitive and was induced by CA. Conclusions Post‐resuscitation normoxic therapy attenuated the oxidative stress in multiple organs and improved post‐CA organ injury, oxygen metabolism, and survival. Additionally, post‐CA hyperoxia increased the mitochondrial reactive oxygen species and activated the antioxidation system.
Collapse
Affiliation(s)
- Yu Okuma
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY
| | - Lance B Becker
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY.,Department of Emergency Medicine Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Hempstead NY
| | - Kei Hayashida
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY
| | - Tomoaki Aoki
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY
| | - Kota Saeki
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY.,Nihon Kohden Innovation Center Cambridge MA
| | | | - Muhammad Shoaib
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY
| | - Santiago J Miyara
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY.,Elmezzi Graduate School of Molecular Medicine Manhasset NY
| | - Tai Yin
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY
| | - Koichiro Shinozaki
- The Feinstein Institutes for Medical ResearchNorthwell Manhasset NY.,Department of Emergency Medicine Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Hempstead NY
| |
Collapse
|
75
|
Protective Effect of Lactobacillus rhamnosus GG on TiO 2 Nanoparticles-Induced Oxidative Stress Damage in the Liver of Young Rats. NANOMATERIALS 2021; 11:nano11030803. [PMID: 33801059 PMCID: PMC8004042 DOI: 10.3390/nano11030803] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Abstract
The potential toxicity of titanium dioxide nanoparticles (TiO2 NPs) to mammals has become a widespread concern. Young individuals exposed to TiO2 NPs have a higher risk than adults. In this study, the protective effects of Lactobacillus rhamnosus GG (LGG) on liver toxicity in young rats induced by TiO2 NPs were explored. Results show that the four-week-old rats that underwent LGG after the oral intake of TiO2 NPs could prevent weight loss, reduce hematological indicators (WBC and NEUT) and serum biochemical indicators (AST, ALT, AST/ALT, and ALP). Moreover, it alleviated the pathological damage of the liver (as indicated by the disordered hepatocytes, more eosinophilic, ballooning degeneration, and accompany with blood cells), but it did not reduce the Ti contents in the liver. In addition, RT-qPCR results indicated that LGG restored the expression of anti-oxidative stress-related genes, such as SOD1, SOD2, CAT, HO-1, GSH, GCLC, and GCLM in the liver. In summary, the hepatotoxicity of TiO2 NPs in young rats is closely related to oxidative stress, and the antioxidant effect of LGG might protect the harmful effects caused by TiO2 NPs.
Collapse
|
76
|
El-Shahat RA, El-Demerdash RS, El Sherbini ES, Saad EA. HCl-induced acute lung injury: a study of the curative role of mesenchymal stem/stromal cells and cobalt protoporphyrin. J Genet Eng Biotechnol 2021; 19:41. [PMID: 33721136 PMCID: PMC7958097 DOI: 10.1186/s43141-021-00139-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Background This study was designed to investigate bone marrow mesenchymal stem/stromal cells (BM-MSCs) and cobalt protoporphyrin (CoPP) curable effects on HCl-induced acute lung injury (ALI) and its underlying mechanisms hoping this might aid to offer a therapeutic opportunity for ALI. Results Forty male Sprague Dawley rats were randomly allocated into four groups; normal (normal rats), ALI (rats injected with 2 ml hydrochloric acid (HCl)/kg via trachea), ALI + BM-MSCs (ALI rats intravenously injected twice with 1 × 106 BM-MSCs/rat/week), and ALI + CoPP (ALI rats intraperitoneally injected twice with CoPP (0.5 mg/100 g/week)). White blood cells (WBCs), red blood cells (RBCs), hemoglobin (Hb), serum tumor necrosis factor-alpha (TNF-α), lung histopathology, apoptosis markers (caspase-3 and Bcl2), and oxidative stress markers (malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT)) were measured. ALI caused increases in WBCs, TNF-α, caspase-3, and MDA, and morphological damage score of lungs with decreases in RBCs, Hb, Bcl2, SOD, and CAT (p < 0.05). BM-MSCs or CoPP treatment reversed these ALI-induced changes (p < 0.05) towards normal. Conclusions BM-MSCs and CoPP could attenuate ALI by modulation of inflammation, oxidative stress, and apoptosis. Curative roles of BM-MSCs were more effective than those of CoPP. This highlights BM-MSCs as a potent therapy for HCl-associated ALI.
Collapse
Affiliation(s)
- Reham A El-Shahat
- Chemistry Department, Faculty of Science, Damietta University, Damietta, Egypt
| | - Reda S El-Demerdash
- Urology & Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - El Said El Sherbini
- Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Entsar A Saad
- Chemistry Department, Faculty of Science, Damietta University, Damietta, Egypt.
| |
Collapse
|
77
|
Zhao J, Que W, Du X, Fujino M, Ichimaru N, Ueta H, Tokuda N, Guo WZ, Zabrocki P, de Haard H, Nonomura N, Li XK. Monotherapy With Anti-CD70 Antibody Causes Long-Term Mouse Cardiac Allograft Acceptance With Induction of Tolerogenic Dendritic Cells. Front Immunol 2021; 11:555996. [PMID: 33737923 PMCID: PMC7961176 DOI: 10.3389/fimmu.2020.555996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/31/2020] [Indexed: 01/02/2023] Open
Abstract
Allograft rejection has been an obstacle for the long-term survival of patients. CD70, a tumor necrosis factor (TNF) family member critically expressed on antigen-presenting cells and strongly but transiently up-regulated during lymphocyte activation, represents an important co-stimulatory molecule that induces effective T cell responses. We used a mouse heterotopic cardiac transplantation model to evaluate the effects of monotherapy with the antibody targeting mouse CD70 (FR70) on transplantation tolerance and its immunoregulatory activity. FR70-treated C3H recipient mice permanently accepted B6 fully mismatched cardiac allografts. Consistent with the graft survival, the infiltration of CD8+ T cells in the graft was reduced, dendritic cells were differentiated into a tolerogenic status, and the number of regulatory T cells was elevated both in the graft and the recipient’s spleen. In addition, naïve C3H given an adoptive transfer of spleen cells from the primary recipients with FR70 treatment accepted a heart graft from a matching B6 donor but not third-party BALB/c mice. Our findings show that treatment with FR70 induced regulatory cells and inhibited cytotoxic T cell proliferation, which led to long-term acceptance of mouse cardiac allografts. These findings highlight the potential role of anti-CD70 antibodies as a clinically effective treatment for allograft rejection.
Collapse
Affiliation(s)
- Jing Zhao
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Osaka, Japan.,Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weitao Que
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Xiaoxiao Du
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Naotsugu Ichimaru
- Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hisashi Ueta
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Nobuko Tokuda
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Wen-Zhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | - Norio Nonomura
- Department of Specific Organ Regulation (Urology), Osaka University Graduate School of Medicine, Osaka, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
78
|
Haga J, Sato N, Anazawa T, Kimura T, Kenjo A, Gotoh M, Marubashi S. Comprehensive analysis of gene expression of isolated pancreatic islets during pretransplant culture. Fukushima J Med Sci 2021; 67:17-26. [PMID: 33597316 PMCID: PMC8075558 DOI: 10.5387/fms.2020-25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The aim of this study was to investigate the effect of pretransplant culture on the survival of pancreatic islet grafts, and to determine the biological characteristics of isolated islets during pretransplant culture. Methods: The survival of islets from Wistar rats, transplanted to diabetic C57BL/B6 mice, was compared between fresh islets and cultured islets. A comprehensive gene expression analysis was employed to investigate biological processes during pretransplant culture, and in vitro validation studies were performed. Results: Survival of cultured xenografts was significantly prolonged as compared to that of fresh islets (fresh: 12.5 ± 1.9 days, 1-day cultured: 16.0 ± 1.3 days (p= 0.017), 3-day cultured: 17.0 ± 2.6 days (p= 0.014)). Comprehensive gene expression analysis identified significant upregulation of annotated functions associated with inflammation in cultured groups. Six proinflammatory genes, including heme oxygenase 1 (HO-1) and IL-6, were significantly upregulated during culture. Validation studies revealed significantly higher levels of IL-6 in the supernatant of cultured islets and HO-1 in the cultured islets when compared with fresh islets. Conclusion: Transplantation of cultured islets induced significant but minimal prolongation of graft survival in xenogeneic combinations. Comprehensive analysis of gene expression in cultured islets showed biological processes associated with proinflammation during culture.
Collapse
Affiliation(s)
- Junichiro Haga
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Naoya Sato
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Takayuki Anazawa
- Department of Surgery, Graduate School of Medicine, Kyoto University
| | - Takashi Kimura
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Akira Kenjo
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Mitsukazu Gotoh
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Shigeru Marubashi
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| |
Collapse
|
79
|
Hara Y, Nakashima K, Nagasawa R, Murohashi K, Tagami Y, Aoki A, Okudela K, Kaneko T. Heme Oxygenase-1 in Patients With Interstitial Lung Disease: A Review of the Clinical Evidence. Am J Med Sci 2021; 362:122-129. [PMID: 33587911 DOI: 10.1016/j.amjms.2021.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/09/2021] [Indexed: 11/17/2022]
Abstract
The clinical course and rate of progression of interstitial lung disease (ILD) are extremely variable among patients. For the purpose of monitoring disease activity, ILD diagnosis, and predicting disease prognosis, there are various biomarkers, including symptoms, physiological, radiological, and pathological findings, and peripheral blood and bronchoalveolar lavage fluid results. Of these, blood biomarkers such as sialylated carbohydrate antigen, surfactant proteins-A and -D, CC-chemokine ligand 18, matrix metalloprotease-1 and -7, CA19-9, and CA125 have been previously proposed. In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Recent research suggests that HO-1 can increase in lung tissues of patients with ILD, reflecting anti-inflammatory M2 macrophage activation, and the measurement of HO-1 levels in peripheral blood can be useful for evaluating the severity of lung damage in ILD and for predicting subsequent fibrosis formation.
Collapse
Affiliation(s)
- Yu Hara
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 4-57 Fukuura, Kanazawa-ku, Yokohama City, 236-0024, Japan.
| | - Kentaro Nakashima
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 4-57 Fukuura, Kanazawa-ku, Yokohama City, 236-0024, Japan
| | - Ryo Nagasawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 4-57 Fukuura, Kanazawa-ku, Yokohama City, 236-0024, Japan
| | - Kota Murohashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 4-57 Fukuura, Kanazawa-ku, Yokohama City, 236-0024, Japan
| | - Yoichi Tagami
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 4-57 Fukuura, Kanazawa-ku, Yokohama City, 236-0024, Japan
| | - Ayako Aoki
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 4-57 Fukuura, Kanazawa-ku, Yokohama City, 236-0024, Japan
| | - Koji Okudela
- Department of Pathology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama City, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, 4-57 Fukuura, Kanazawa-ku, Yokohama City, 236-0024, Japan
| |
Collapse
|
80
|
Heme Oxygenase-1 Deficiency and Oxidative Stress: A Review of 9 Independent Human Cases and Animal Models. Int J Mol Sci 2021; 22:ijms22041514. [PMID: 33546372 PMCID: PMC7913498 DOI: 10.3390/ijms22041514] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 12/28/2022] Open
Abstract
Since Yachie et al. reported the first description of human heme oxygenase (HO)-1 deficiency more than 20 years ago, few additional human cases have been reported in the literature. A detailed analysis of the first human case of HO-1 deficiency revealed that HO-1 is involved in the protection of multiple tissues and organs from oxidative stress and excessive inflammatory reactions, through the release of multiple molecules with anti-oxidative stress and anti-inflammatory functions. HO-1 production is induced in vivo within selected cell types, including renal tubular epithelium, hepatic Kupffer cells, vascular endothelium, and monocytes/macrophages, suggesting that HO-1 plays critical roles in these cells. In vivo and in vitro studies have indicated that impaired HO-1 production results in progressive monocyte dysfunction, unregulated macrophage activation and endothelial cell dysfunction, leading to catastrophic systemic inflammatory response syndrome. Data from reported human cases of HO-1 deficiency and numerous studies using animal models suggest that HO-1 plays critical roles in various clinical settings involving excessive oxidative stress and inflammation. In this regard, therapy to induce HO-1 production by pharmacological intervention represents a promising novel strategy to control inflammatory diseases.
Collapse
|
81
|
Lignelli E, Palumbo F, Bayindir SG, Nagahara N, Vadász I, Herold S, Seeger W, Morty RE. The H 2S-generating enzyme 3-mercaptopyruvate sulfurtransferase regulates pulmonary vascular smooth muscle cell migration and proliferation but does not impact normal or aberrant lung development. Nitric Oxide 2021; 107:31-45. [PMID: 33338600 DOI: 10.1016/j.niox.2020.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/07/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023]
Abstract
Along with nitric oxide (NO), the gasotransmitters carbon monoxide (CO) and hydrogen sulfide (H2S) are emerging as potentially important players in newborn physiology, as mediators of newborn disease, and as new therapeutic modalities. Several recent studies have addressed H2S in particular in animal models of bronchopulmonary dysplasia (BPD), a common complication of preterm birth where oxygen toxicity stunts lung development. In those studies, exogenous H2S attenuated the impact of oxygen toxicity on lung development, and two H2S-generating enzymes were documented to affect pulmonary vascular development. H2S is directly generated endogenously by three enzymes, one of which, 3-mercaptopyruvate sulfurtransferase (MPST), has not been studied in the lung. In a hyperoxia-based animal model of BPD, oxygen exposure deregulated MPST expression during post-natal lung development, where MPST was localized to the smooth muscle layer of the pulmonary vessels in developing lungs. siRNA-mediated abrogation of MPST expression in human pulmonary artery smooth muscle cells in vitro limited baseline cell migration and cell proliferation, without affecting apoptosis or cell viability. In vivo, MPST was dispensable for normal lung development in Mpst-/-mice, and MPST did not contribute to stunted lung development driven by hyperoxia exposure, assessed by design-based stereology. These data demonstrate novel roles for MPST in pulmonary vascular smooth muscle cell physiology. The potential caveats of using Mpst-/- mice to study normal and aberrant lung development are also discussed, highlighting the possible confounding, compensatory effects of other H2S-generating enzymes that are present alongside MPST in the smooth muscle compartment of developing pulmonary vessels.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Selahattin Görkem Bayindir
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany
| | - Noriyuki Nagahara
- Isotope Research Laboratory, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; Institute for Lung Health (ILH), Justus Liebig University Giessen, Aulweg 130, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Member of the German Center for Lung Research (DZL), Parkstrasse 1, 60231, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Aulweg 123, 35392, Giessen, Germany; CardioPulmonary Institute, Justus Liebig University Giessen, Klinikstrasse 33, Giessen, Germany.
| |
Collapse
|
82
|
Gauthier AG, Wu J, Lin M, Sitapara R, Kulkarni A, Thakur GA, Schmidt EE, Perron JC, Ashby CR, Mantell LL. The Positive Allosteric Modulation of alpha7-Nicotinic Cholinergic Receptors by GAT107 Increases Bacterial Lung Clearance in Hyperoxic Mice by Decreasing Oxidative Stress in Macrophages. Antioxidants (Basel) 2021; 10:135. [PMID: 33477969 PMCID: PMC7835977 DOI: 10.3390/antiox10010135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 01/15/2021] [Indexed: 12/20/2022] Open
Abstract
Supplemental oxygen therapy with supraphysiological concentrations of oxygen (hyperoxia; >21% O2) is a life-saving intervention for patients experiencing respiratory distress. However, prolonged exposure to hyperoxia can compromise bacterial clearance processes, due to oxidative stress-mediated impairment of macrophages, contributing to the increased susceptibility to pulmonary infections. This study reports that the activation of the α7 nicotinic acetylcholine receptor (α7nAChR) with the delete allosteric agonistic-positive allosteric modulator, GAT107, decreases the bacterial burden in mouse lungs by improving hyperoxia-induced lung redox imbalance. The incubation of RAW 264.7 cells with GAT107 (3.3 µM) rescues hyperoxia-compromised phagocytic functions in cultured macrophages, RAW 264.7 cells, and primary bone marrow-derived macrophages. Similarly, GAT107 (3.3 µM) also attenuated oxidative stress in hyperoxia-exposed macrophages, which prevents oxidation and hyper-polymerization of phagosome filamentous actin (F-actin) from oxidation. Furthermore, GAT107 (3.3 µM) increases the (1) activity of superoxide dismutase 1; (2) activation of Nrf2 and (3) the expression of heme oxygenase-1 (HO-1) in macrophages exposed to hyperoxia. Overall, these data suggest that the novel α7nAChR compound, GAT107, could be used to improve host defense functions in patients, such as those with COVID-19, who are exposed to prolonged periods of hyperoxia.
Collapse
Affiliation(s)
- Alex G. Gauthier
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Jiaqi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Mosi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Ravikumar Sitapara
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (A.K.); (G.A.T.)
| | - Ganesh A. Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (A.K.); (G.A.T.)
| | - Edward E. Schmidt
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA;
| | - Jeanette C. Perron
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| |
Collapse
|
83
|
Lu D, Le Y, Ding J, Dou X, Mao W, Zhu J. CLIC1 Inhibition Protects Against Cellular Senescence and Endothelial Dysfunction Via the Nrf2/HO-1 Pathway. Cell Biochem Biophys 2021; 79:239-252. [PMID: 33432550 DOI: 10.1007/s12013-020-00959-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 02/02/2023]
Abstract
Chloride intracellular channel 1 (CLIC1) is a sensor of oxidative stress in endothelial cells (EC). However, the mechanism by which CLIC1 mediate the regulation of endothelial dysfunction has not been established. In this study, overexpressed CLIC1 impaired the ability of the vascular cells to resist oxidative damage and promoted cellular senescence. Besides, suppressed CLIC1 protected against cellular senescence and dysfunction in Human Umbilical Vein Endothelial Cells (HUVECs) through the Nrf2/HO-1 pathway. We also found that ROS-activated CLIC1-induced oxidative stress in HUVECs. Nrf2 nuclear translocation was inhibited by CLIC1 overexpression, but was enhanced by IAA94 (CLICs inhibitor) treatment or knockdown of CLIC1. The Nrf2/HO-1 pathway plays a critical role in the anti-oxidative effect of suppressing CLIC1. And inhibition of CLIC1 decreases oxidative stress injury by downregulating the levels of ROS, MDA, and the expression of EC effectors (ICAM1 and VCAM1) protein expression and promotes the activity of superoxide dismutase (SOD). The AMPK-mediated signaling pathway activates Nrf2 through Nrf2 phosphorylation and nuclear translocation, which is also regulated by CLIC1. Moreover, the activation of CLIC1 contributes to H2O2-induced mitochondrial dysfunction and activation of mitochondrial fission. Therefore, elucidation of the mechanisms by which CLIC1 is involved in these pivotal pathways may uncover its therapeutic potential in alleviating ECs oxidative stress and age-related cardiovascular disease development.
Collapse
Affiliation(s)
- Dezhao Lu
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Yifei Le
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Jiali Ding
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Xiaobing Dou
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Wei Mao
- Cardiovascular department, The First Affiliated Hospital of Zhejiang Chinese Medicine University, 310006, Hangzhou, China.
| | - Ji Zhu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
84
|
Connick JP, Reed JR, Cawley GF, Backes WL. Heme oxygenase-1 affects cytochrome P450 function through the formation of heteromeric complexes: Interactions between CYP1A2 and heme oxygenase-1. J Biol Chem 2021; 296:100030. [PMID: 33148696 PMCID: PMC7948974 DOI: 10.1074/jbc.ra120.015911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/27/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022] Open
Abstract
Heme oxygenase 1 (HO-1) and the cytochromes P450 (P450s) are endoplasmic reticulum-bound enzymes that rely on the same protein, NADPH-cytochrome P450 reductase (POR), to provide the electrons necessary for substrate metabolism. Although the HO-1 and P450 systems are interconnected owing to their common electron donor, they generally have been studied separately. As the expressions of both HO-1 and P450s are affected by xenobiotic exposure, changes in HO-1 expression can potentially affect P450 function and, conversely, changes in P450 expression can influence HO-1. The goal of this study was to examine interactions between the P450 and HO-1 systems. Using bioluminescence resonance energy transfer (BRET), HO-1 formed HO-1•P450 complexes with CYP1A2, CYP1A1, and CYP2D6, but not all P450s. Studies then focused on the HO-1-CYP1A2 interaction. CYP1A2 formed a physical complex with HO-1 that was stable in the presence of POR. As expected, both HO-1 and CYP1A2 formed BRET-detectable complexes with POR. The POR•CYP1A2 complex was readily disrupted by the addition of HO-1, whereas the POR•HO-1 complex was not significantly affected by the addition of CYP1A2. Interestingly, enzyme activities did not follow this pattern. BRET data suggested substantial inhibition of CYP1A2-mediated 7-ethoxyresorufin de-ethylation in the presence of HO-1, whereas its activity was actually stimulated at subsaturating POR. In contrast, HO-1-mediated heme metabolism was inhibited at subsaturating POR. These results indicate that HO-1 and CYP1A2 form a stable complex and have mutual effects on the catalytic behavior of both proteins that cannot be explained by a simple competition for POR.
Collapse
Affiliation(s)
- J Patrick Connick
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - James R Reed
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - George F Cawley
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Wayne L Backes
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
| |
Collapse
|
85
|
Hahn D, Shin SH, Bae JS. Natural Antioxidant and Anti-Inflammatory Compounds in Foodstuff or Medicinal Herbs Inducing Heme Oxygenase-1 Expression. Antioxidants (Basel) 2020; 9:E1191. [PMID: 33260980 PMCID: PMC7761319 DOI: 10.3390/antiox9121191] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that catalyzes heme group degradation. Decreased level of HO-1 is correlated with disease progression, and HO-1 induction suppresses development of metabolic and neurological disorders. Natural compounds with antioxidant activities have emerged as a rich source of HO-1 inducers with marginal toxicity. Here we discuss the therapeutic role of HO-1 in obesity, hypertension, atherosclerosis, Parkinson's disease and hepatic fibrosis, and present important signaling pathway components that lead to HO-1 expression. We provide an updated, comprehensive list of natural HO-1 inducers in foodstuff and medicinal herbs categorized by their chemical structures. Based on the continued research in HO-1 signaling pathways and rapid development of their natural inducers, HO-1 may serve as a preventive and therapeutic target for metabolic and neurological disorders.
Collapse
Affiliation(s)
- Dongyup Hahn
- School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Korea;
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
86
|
Merk R, Heßelbach K, Osipova A, Popadić D, Schmidt-Heck W, Kim GJ, Günther S, Piñeres AG, Merfort I, Humar M. Particulate Matter (PM 2.5) from Biomass Combustion Induces an Anti-Oxidative Response and Cancer Drug Resistance in Human Bronchial Epithelial BEAS-2B Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E8193. [PMID: 33171923 PMCID: PMC7664250 DOI: 10.3390/ijerph17218193] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Nearly half of the world's population relies on combustion of solid biofuels to cover fundamental energy demands. Epidemiologic data demonstrate that particularly long-term emissions adversely affect human health. However, pathological molecular mechanisms are insufficiently characterized. Here we demonstrate that long-term exposure to fine particulate matter (PM2.5) from biomass combustion had no impact on cellular viability and proliferation but increased intracellular reactive oxygen species (ROS) levels in bronchial epithelial BEAS-2B cells. Exposure to PM2.5 induced the nuclear factor erythroid 2-related factor 2 (Nrf2) and mediated an anti-oxidative response, including enhanced levels of intracellular glutathione (GSH) and nuclear accumulation of heme oxygenase-1 (HO-1). Activation of Nrf2 was promoted by the c-Jun N-terminal kinase JNK1/2, but not p38 or Akt, which were also induced by PM2.5. Furthermore, cells exposed to PM2.5 acquired chemoresistance to doxorubicin, which was associated with inhibition of apoptosis and elevated levels of GSH in these cells. Our findings propose that exposure to PM2.5 induces molecular defense mechanisms, which prevent cellular damage and may thus explain the initially relative rare complications associated with PM2.5. However, consistent induction of pro-survival pathways may also promote the progression of diseases. Environmental conditions inducing anti-oxidative responses may have the potential to promote a chemoresistant cellular phenotype.
Collapse
Affiliation(s)
- Regina Merk
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Katharina Heßelbach
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Anastasiya Osipova
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Désirée Popadić
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| | - Wolfgang Schmidt-Heck
- Department of Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knöll Institute (HKI), 07745 Jena, Germany;
| | - Gwang-Jin Kim
- Department of Pharmaceutical Bioinformatics, Institute of Pharmaceutical Sciences, Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany; (G.-J.K.); (S.G.)
| | - Stefan Günther
- Department of Pharmaceutical Bioinformatics, Institute of Pharmaceutical Sciences, Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany; (G.-J.K.); (S.G.)
| | - Alfonso García Piñeres
- Centro de Investigación en Biología Celular y Molecular (CIBCM), Universidad de Costa Rica, 11501-2060 San José, Costa Rica;
- Escuela de Química, Universidad de Costa Rica, 11501-2060 San José, Costa Rica
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany
| | - Matjaz Humar
- Department of Pharmaceutical Biology and Biotechnology, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, 79104 Freiburg, Germany; (R.M.); (K.H.); (A.O.); (D.P.)
| |
Collapse
|
87
|
Cao X, Tang L, Zeng Z, Wang B, Zhou Y, Wang Q, Zou P, Li W. Effects of Probiotics BaSC06 on Intestinal Digestion and Absorption, Antioxidant Capacity, Microbiota Composition, and Macrophage Polarization in Pigs for Fattening. Front Vet Sci 2020; 7:570593. [PMID: 33240950 PMCID: PMC7677304 DOI: 10.3389/fvets.2020.570593] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/29/2020] [Indexed: 01/24/2023] Open
Abstract
This study aimed to compare the effects of BaSC06 and antibiotics on growth, digestive functions, antioxidant capacity, macrophage polarization, and intestinal microbiota of pigs for fattening. A total of 117 pigs for fattening with similar weight and genetic basis were divided into 3 groups: Anti group (containing 40 g/t Kitasamycin in the diet), Anti+Ba group (containing 20 g/t Kitasamycin and 0.5 × 108 CFU/kg BaSC06 in the diet) and Ba group (containing 1 × 108 cfu/Kg BaSC06 in the diet without any antibiotics). Each treatment was performed in three replicates with 13 pigs per replicate. Results showed that BaSC06 replacement significantly improved the ADG (P < 0.05), intestinal digestion and absorption function by increasing the activity of intestinal digestive enzymes and the expression of glucose transporters SGLT1 (P < 0.05) and small peptide transporters PEPT1 (P < 0.05). Besides, BaSC06 supplementation enhanced intestinal and body antioxidant capacity by activating the Nrf2/Keap1 antioxidant signaling pathway due to the increased expression of p-Nrf2 (P < 0.05). Notably, BaSC06 alleviated intestinal inflammation by inhibiting the production of pro-inflammatory cytokines, IL-8, IL-6, and MCP1 (P < 0.05), and simultaneously increasing the expression of M1 macrophage marker protein iNOS (P < 0.05) and M2 macrophage marker protein Arg (P < 0.05) in the intestinal mucosa. Moreover, BaSC06 promoted the polarization of macrophages to M2 phenotype by stimulating the STAT3 signaling pathway. It was also noted that BaSC06 improved microbiota composition by enhancing the proportion of Firmicutes, and reducing that of Bacteroidetes and Proteobacteria. Taken together, our results indicate that dietary supplementation of BaSC06 in pigs for fattening improves the growth, mucosal structure, antioxidative capacity, immune functions (including increasing M1 and M2 polarization of macrophage) and composition of intestinal microbiota, which is much better than antibiotics, suggesting that it is an effective alternative to antibiotics in the preparation of pig feed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University, and Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
| |
Collapse
|
88
|
Chau AS, Cole BL, Debley JS, Nanda K, Rosen ABI, Bamshad MJ, Nickerson DA, Torgerson TR, Allenspach EJ. Heme oxygenase-1 deficiency presenting with interstitial lung disease and hemophagocytic flares. Pediatr Rheumatol Online J 2020; 18:80. [PMID: 33066778 PMCID: PMC7565350 DOI: 10.1186/s12969-020-00474-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/06/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Through biliverdin reductase, biliverdin becomes bilirubin. HMOX1-deficiency is a rare autosomal recessive disorder with hallmark features of direct antibody negative hemolytic anemia with normal bilirubin, hyperinflammation and features similar to macrophage activation syndrome. Clinical findings have included asplenia, nephritis, hepatitis, and vasculitis. Pulmonary features and evaluation of the immune response have been limited. CASE PRESENTATION We present a young boy who presented with chronic respiratory failure due to nonspecific interstitial pneumonia following a chronic history of infection-triggered recurrent hyperinflammatory flares. Episodes included hemolysis without hyperbilirubinemia, immunodeficiency, hepatomegaly with mild transaminitis, asplenia, leukocytosis, thrombocytosis, joint pain and features of macrophage activation with negative autoimmune serologies. Lung biopsy revealed cholesterol granulomas. He was found post-mortem by whole exome sequencing to have a compound heterozygous paternal frame shift a paternal frame shift HMOX1 c.264_269delCTGG (p.L89Sfs*24) and maternal splice donor HMOX1 (c.636 + 2 T > A) consistent with HMOX1 deficiency. Western blot analysis confirmed lack of HMOX1 protein upon oxidant stimulation of the patient cells. CONCLUSIONS Here, we describe a phenotype expansion for HMOX1-deficiency to include not only asplenia and hepatomegaly, but also interstitial lung disease with cholesterol granulomas and inflammatory flares with hemophagocytosis present in the bone marrow.
Collapse
Affiliation(s)
- Alice S. Chau
- grid.34477.330000000122986657Division of Allergy & Infectious Disease, Department of Medicine, University of Washington, Seattle, Washington USA ,grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA
| | - Bonnie L. Cole
- grid.34477.330000000122986657Department of Pathology and Laboratory Medicine, University of Washington, Seattle, Washington USA ,grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA
| | - Jason S. Debley
- grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA ,grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA
| | - Kabita Nanda
- grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA
| | - Aaron B. I. Rosen
- grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA
| | - Michael J. Bamshad
- grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA ,grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA ,grid.34477.330000000122986657Genome Sciences, University of Washington, Seattle, Washington USA
| | - Deborah A. Nickerson
- grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA ,grid.34477.330000000122986657Genome Sciences, University of Washington, Seattle, Washington USA
| | - Troy R. Torgerson
- grid.507729.eExperimental Immunology, Allen Institute, Seattle, Washington USA
| | - Eric J. Allenspach
- grid.240741.40000 0000 9026 4165Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Jack MacDonald Building – 6th floor, 1900 9th Avenue, Seattle, Washington 98101 USA ,grid.507913.9Brotman Baty Institute for Precision Medicine, Seattle, Washington USA ,grid.34477.330000000122986657Department of Pediatrics, University of Washington, Seattle, Washington USA
| |
Collapse
|
89
|
Possible Susceptibility Genes for Intervention against Chemotherapy-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4894625. [PMID: 33110473 PMCID: PMC7578723 DOI: 10.1155/2020/4894625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/07/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Recent therapeutic advances have significantly improved the short- and long-term survival rates in patients with heart disease and cancer. Survival in cancer patients may, however, be accompanied by disadvantages, namely, increased rates of cardiovascular events. Chemotherapy-related cardiac dysfunction is an important side effect of anticancer therapy. While advances in cancer treatment have increased patient survival, treatments are associated with cardiovascular complications, including heart failure (HF), arrhythmias, cardiac ischemia, valve disease, pericarditis, and fibrosis of the pericardium and myocardium. The molecular mechanisms of cardiotoxicity caused by cancer treatment have not yet been elucidated, and they may be both varied and complex. By identifying the functional genetic variations responsible for this toxicity, we may be able to improve our understanding of the potential mechanisms and pathways of treatment, paving the way for the development of new therapies to target these toxicities. Data from studies on genetic defects and pharmacological interventions have suggested that many molecules, primarily those regulating oxidative stress, inflammation, autophagy, apoptosis, and metabolism, contribute to the pathogenesis of cardiotoxicity induced by cancer treatment. Here, we review the progress of genetic research in illuminating the molecular mechanisms of cancer treatment-mediated cardiotoxicity and provide insights for the research and development of new therapies to treat or even prevent cardiotoxicity in patients undergoing cancer treatment. The current evidence is not clear about the role of pharmacogenomic screening of susceptible genes. Further studies need to done in chemotherapy-induced cardiotoxicity.
Collapse
|
90
|
Systematic Surveys of Iron Homeostasis Mechanisms Reveal Ferritin Superfamily and Nucleotide Surveillance Regulation to be Modified by PINK1 Absence. Cells 2020; 9:cells9102229. [PMID: 33023155 PMCID: PMC7650593 DOI: 10.3390/cells9102229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022] Open
Abstract
Iron deprivation activates mitophagy and extends lifespan in nematodes. In patients suffering from Parkinson’s disease (PD), PINK1-PRKN mutations via deficient mitophagy trigger iron accumulation and reduce lifespan. To evaluate molecular effects of iron chelator drugs as a potential PD therapy, we assessed fibroblasts by global proteome profiles and targeted transcript analyses. In mouse cells, iron shortage decreased protein abundance for iron-binding nucleotide metabolism enzymes (prominently XDH and ferritin homolog RRM2). It also decreased the expression of factors with a role for nucleotide surveillance, which associate with iron-sulfur-clusters (ISC), and are important for growth and survival. This widespread effect included prominently Nthl1-Ppat-Bdh2, but also mitochondrial Glrx5-Nfu1-Bola1, cytosolic Aco1-Abce1-Tyw5, and nuclear Dna2-Elp3-Pold1-Prim2. Incidentally, upregulated Pink1-Prkn levels explained mitophagy induction, the downregulated expression of Slc25a28 suggested it to function in iron export. The impact of PINK1 mutations in mouse and patient cells was pronounced only after iron overload, causing hyperreactive expression of ribosomal surveillance factor Abce1 and of ferritin, despite ferritin translation being repressed by IRP1. This misregulation might be explained by the deficiency of the ISC-biogenesis factor GLRX5. Our systematic survey suggests mitochondrial ISC-biogenesis and post-transcriptional iron regulation to be important in the decision, whether organisms undergo PD pathogenesis or healthy aging.
Collapse
|
91
|
Vodošek Hojs N, Bevc S, Ekart R, Hojs R. Oxidative Stress Markers in Chronic Kidney Disease with Emphasis on Diabetic Nephropathy. Antioxidants (Basel) 2020; 9:925. [PMID: 32992565 PMCID: PMC7600946 DOI: 10.3390/antiox9100925] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/20/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetes prevalence is increasing worldwide, especially through the increase of type 2 diabetes. Diabetic nephropathy occurs in up to 40% of diabetic patients and is the leading cause of end-stage renal disease. Various factors affect the development and progression of diabetic nephropathy. Hyperglycaemia increases free radical production, resulting in oxidative stress, which plays an important role in the pathogenesis of diabetic nephropathy. Free radicals have a short half-life and are difficult to measure. In contrast, oxidation products, including lipid peroxidation, protein oxidation, and nucleic acid oxidation, have longer lifetimes and are used to evaluate oxidative stress. In recent years, different oxidative stress biomarkers associated with diabetic nephropathy have been found. This review summarises current evidence of oxidative stress biomarkers in patients with diabetic nephropathy. Although some of them are promising, they cannot replace currently used clinical biomarkers (eGFR, proteinuria) in the development and progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Nina Vodošek Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia; (N.V.H.); (S.B.)
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia; (N.V.H.); (S.B.)
- Faculty of Medicine, University of Maribor, Taborska 8, 2000 Maribor, Slovenia;
| | - Robert Ekart
- Faculty of Medicine, University of Maribor, Taborska 8, 2000 Maribor, Slovenia;
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska 5, 2000 Maribor, Slovenia; (N.V.H.); (S.B.)
- Faculty of Medicine, University of Maribor, Taborska 8, 2000 Maribor, Slovenia;
| |
Collapse
|
92
|
Hirao H, Dery KJ, Kageyama S, Nakamura K, Kupiec-Weglinski JW. Heme Oxygenase-1 in liver transplant ischemia-reperfusion injury: From bench-to-bedside. Free Radic Biol Med 2020; 157:75-82. [PMID: 32084514 PMCID: PMC7434658 DOI: 10.1016/j.freeradbiomed.2020.02.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/02/2020] [Accepted: 02/17/2020] [Indexed: 12/16/2022]
Abstract
Hepatic ischemia-reperfusion injury (IRI), a major risk factor for early allograft dysfunction (EAD) and acute or chronic graft rejection, contributes to donor organ shortage for life-saving orthotopic liver transplantation (OLT). The graft injury caused by local ischemia (warm and/or cold) leads to parenchymal cell death and release of danger-associated molecular patterns (DAMPs), followed by reperfusion-triggered production of reactive oxygen species (ROS), activation of inflammatory cells, hepatocellular damage and ultimate organ failure. Heme oxygenase 1 (HO-1), a heat shock protein-32 induced under IR-stress, is an essential component of the cytoprotective mechanism in stressed livers. HO-1 regulates anti-inflammatory responses and may be crucial in the pathogenesis of chronic diseases, such as arteriosclerosis, hypertension, diabetes and steatosis. An emerging area of study is macrophage-derived HO-1 and its pivotal intrahepatic homeostatic function played in IRI-OLT. Indeed, ectopic hepatic HO-1 overexpression activates intracellular SIRT1/autophagy axis to serve as a key cellular self-defense mechanism in both mouse and human OLT recipients. Recent translational studies in rodents and human liver transplant patients provide novel insights into HO-1 mediated cytoprotection against sterile hepatic inflammation. In this review, we summarize the current bench-to-bedside knowledge on HO-1 molecular signaling and discuss their future therapeutic potential to mitigate IRI in OLT.
Collapse
Affiliation(s)
- Hirofumi Hirao
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Kenneth J Dery
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Shoichi Kageyama
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Kojiro Nakamura
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA; Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Surgery, Nishi-Kobe Medical Center, 5-7-1 Koji-dai, Nishi-ku, Kobe, Hyogo, 651-2273, Japan
| | - Jerzy W Kupiec-Weglinski
- Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
93
|
Kwong KK, Chan ST. The role of carbon monoxide and heme oxygenase-1 in COVID-19. Toxicol Rep 2020; 7:1170-1171. [PMID: 32904637 PMCID: PMC7457907 DOI: 10.1016/j.toxrep.2020.08.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Indexed: 11/25/2022] Open
Abstract
•Low level carbon monoxide (CO) and heme oxygenase-1 (HO-1) may play a role on the effect of smoking on COVID-19. •CO and HO-1 are anti-inflammatory and cytoprotective and HO-1 is most protective if it is induced before the occurrence of an oxidative insult. •COVID-19 may by itself induce HO-1 and CO which may be potential markers of inflammation or cytokine storm. •Some asymptomatic patients of COVID-19 may have a high baseline level of HO-1 or CO before their COVID-19 infection. •Non-invasive measurement of carboxyhemoglobin using a pulse CO-oximeter may be used to index CO and HO-1 level.
Collapse
Affiliation(s)
- Kenneth K. Kwong
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| | - Suk-tak Chan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
94
|
Han HL, Zhang JF, Yan EF, Shen MM, Wu JM, Gan ZD, Wei CH, Zhang LL, Wang T. Effects of taurine on growth performance, antioxidant capacity, and lipid metabolism in broiler chickens. Poult Sci 2020; 99:5707-5717. [PMID: 33142488 PMCID: PMC7647726 DOI: 10.1016/j.psj.2020.07.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/07/2020] [Accepted: 07/23/2020] [Indexed: 10/31/2022] Open
Abstract
To investigate the effects of dietary taurine supplementation on growth performance, antioxidant status, and lipid metabolism in broilers, 384 male broilers (Arbor Acres, 1 D of age) were randomly allocated into 4 groups with 8 replicates of 8 birds. Dietary treatments were supplemented with taurine at the level of 0.00, 2.50, 5.00, and 7.50 g/kg of the diet (denoted as CON, TAU1, TAU2, TAU3, respectively). The BW gain from 1 to 21 D and from 22 to 42 D were all increased linearly (linear, P < 0.001) by taurine supplementation. Throughout the trial period, the highest BW gain and favorable gain-to-feed ratio were observed in the TAU2 group. Taurine supplementation increased the antioxidant enzyme activities and decreased (linear, P < 0.001) the content of malondialdehyde in both serum and the liver of broilers and alleviated oxidative damage through enhancing (P < 0.05) the hepatic genes expression of nuclear factor erythroid-2-related factor 2 (NRF2), glutathione peroxidase (GPX), and heme oxygenase-1 (HO-1). Correspondingly, in serum, the activities of hepatic lipase and total lipase were decreased linearly and quadratically (linear and quadratic, P < 0.001) with the increasing inclusion of taurine in the diet. Meanwhile, in serum, the content of triglycerides was significantly decreased (P < 0.05), and except for TAU3, the total cholesterol content was also significantly decreased (P < 0.05) by taurine supplementation. In addition, the hepatic content of triglycerides was significantly decreased (P < 0.05) in the TAU1 and TAU2 groups. Compared with the CON group, the hepatic genes expression of adenosine monophosphate-activated protein kinase alpha (AMPKα), silent 1, (SIRT1) and carnitine palmitoyltransferase 1 (CPT-1) were all increased (P < 0.05), and sterol regulatory element-binding protein-1 (SREBP-1) expression was decreased (P < 0.05) in the TAU2 group. These results indicated that taurine supplementation improved the growth performance, antioxidant capacity, and lipid metabolism of broilers.
Collapse
Affiliation(s)
- H L Han
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - J F Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - E F Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - M M Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - J M Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - Z D Gan
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - C H Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - L L Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China
| | - T Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu 210095, Nanjing, People's Republic of China.
| |
Collapse
|
95
|
Zhou J, Terluk MR, Basso L, Mishra UR, Orchard PJ, Cloyd JC, Schröder H, Kartha RV. N-acetylcysteine Provides Cytoprotection in Murine Oligodendrocytes through Heme Oxygenase-1 Activity. Biomedicines 2020; 8:biomedicines8080240. [PMID: 32717964 PMCID: PMC7460204 DOI: 10.3390/biomedicines8080240] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Oligodendrocytic injury by oxidative stress can lead to demyelination, contributing to neurodegeneration. We investigated the mechanisms by which an antioxidant, N-acetylcysteine (NAC), reduces oxidative stress in murine oligodendrocytes. We used normal 158N and mutant 158JP cells with endogenously high reactive oxygen species (ROS) levels. Oxidative stress was induced in 158N cells using hydrogen peroxide (H2O2, 500 μM), and both cells were treated with NAC (50 µM to 500 µM). ROS production, total glutathione (GSH) and cell survival were measured 24 h after treatment. In normal cells, H2O2 treatment resulted in a ~5.5-fold increase in ROS and ~50% cell death. These deleterious effects of oxidative stress were attenuated by NAC, resulting in improved cell survival. Similarly, NAC treatment resulted in decreased ROS levels in 158JP cells. Characterization of mechanisms underlying cytoprotection in both cell lines revealed an increase in GSH levels by NAC, which was partially blocked by an inhibitor of GSH synthesis. Interestingly, we observed heme oxygenase-1 (HO-1), a cytoprotective enzyme, play a critical role in cytoprotection. Inhibition of HO-1 activity abolished the cytoprotective effect of NAC with a corresponding decrease in total antioxidant capacity. Our results indicate that NAC promotes oligodendrocyte survival in oxidative stress-related conditions through multiple pathways.
Collapse
Affiliation(s)
- Jie Zhou
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Marcia R. Terluk
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Lisa Basso
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Usha R. Mishra
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Paul J. Orchard
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, Medical School, University of Minnesota, 425 East River Parkway, Minneapolis, MN 55455, USA;
| | - James C. Cloyd
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
| | - Henning Schröder
- Department of Pharmaceutics, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA;
| | - Reena V. Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (L.B.); (U.R.M.); (J.C.C.)
- Correspondence: ; Tel.: +1-612-626-2436
| |
Collapse
|
96
|
Figliuzzi M, Tironi M, Longaretti L, Mancini A, Teoldi F, Sangalli F, Remuzzi A. Copper-dependent biological effects of particulate matter produced by brake systems on lung alveolar cells. Arch Toxicol 2020; 94:2965-2979. [PMID: 32577786 DOI: 10.1007/s00204-020-02812-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022]
Abstract
Road traffic is one of the main sources of particulate emissions into the environment and has an increasing, negative impact on the release of potentially dangerous materials. Vehicle brakes release a significant amount of wear particles, and knowledge regarding their possible adverse effects is limited. One of the most dangerous elements contained in brake pads is copper (Cu), known to be toxic for human health. Therefore, our aim was to study the cell toxicity of particulate matter (PM) produced by different combinations of braking discs and pads containing different amounts of Cu. We investigated whether brake-derived microparticles have toxic effects on lung cells proportionally to their Cu content. Analyte content was measured in friction materials by XRFS and in PM2.5 captured during braking tests using SEM/EDX. The biological impact of brake-derived PM2.5 was investigated on a human epithelial alveolar cell line (A549). Cell viability, oxidative stress, mitochondrial membrane potential, apoptosis, and the pro-inflammatory response of the cells, as well as gene expression, were assessed following exposure to increasing PM2.5 concentrations (1, 10, 100, 200, and 500 µg/ml). The brake debris with the lowest Cu content did not induce significant changes in biological effects on A549 cells compared to normal controls, except for ROS production and IL6 gene expression. PM2.5 containing higher Cu quantities induced cell toxicity that correlated with Cu concentration. Our data suggest that the toxicity of PM2.5 from the brake system is mainly related to Cu content, thus confirming that eliminating Cu from brake pads will be beneficial for human health in urbanized environments.
Collapse
Affiliation(s)
- Marina Figliuzzi
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126, Bergamo, Italy.
| | - Matteo Tironi
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126, Bergamo, Italy
| | - Lorena Longaretti
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, Bergamo, Italy
| | - Alessandro Mancini
- Laboratorio Materiali Advanced R&D Brembo S.P.A, Viale Europa, 2, Stezzano, BG, Italy
| | - Federico Teoldi
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Negri 2, Milan, Italy
| | - Fabio Sangalli
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126, Bergamo, Italy
| | - Andrea Remuzzi
- Department of Management, Information and Production Engineering, University of Bergamo, Viale Marconi 5, Dalmine, BG, Italy
| |
Collapse
|
97
|
Chillappagari S, Garapati V, Mahavadi P, Naehrlich L, Schmeck BT, Schmitz ML, Guenther A. Defective BACH1/HO-1 regulatory circuits in cystic fibrosis bronchial epithelial cells. J Cyst Fibros 2020; 20:140-148. [PMID: 32534959 DOI: 10.1016/j.jcf.2020.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/01/2020] [Accepted: 05/23/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The stress-regulated enzyme hemeoxygenase-1 (HO-1) contributes to the cell response towards inflammation and oxidative stress. We previously reported on curtailed HO-1 expression in cystic fibrosis (CF) bronchial epithelial (CFBE41o-) cells and CF-mice, but the molecular mechanisms for this are not known. Here, we compared healthy and CF bronchial epithelial cells for regulatory circuits controlling HO-1 protein levels. METHODS In this study, we employed immunohistochemistry on CF and healthy lung sections to examine the BACH1 protein expression. Alteration of BACH1 protein levels in 16HBE14o- and CFBE41o- cells was achieved by using either siRNA-mediated knockdown of BACH1 or by increasing miRNA-155 levels. HO-1 luciferase reporter assay was chosen to examine the downstream affects after BACH1 modulation. RESULTS Human CF lungs and cells showed increased levels of the HO-1 transcriptional repressor, BACH1, and increased miR-155 expression. Knockdown studies using BACH1 siRNA and overexpression of miR-155 did not significantly rescue HO-1 expression in CFBE41o- cells. Elevated BACH1 expression detected in CF cells was refractory to the inhibitory function of miR-155 and was instead due to increased protein stability. CONCLUSION We observed defects in the inhibitory activities of miR-155 and BACH1 on HO-1 expression in CF cells. Thus various defective regulatory loops account for dysregulated BACH1 expression in CF, which in turn may contribute to low HO-1 levels.
Collapse
Affiliation(s)
- Shashipavan Chillappagari
- Institute of Biochemistry, Justus-Liebig-University, D-35392, Giessen, Germany; Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany.
| | - Virajith Garapati
- Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Lutz Naehrlich
- University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany; Department of Pediatrics, Justus Liebig University, Giessen, Feulgenstrasse 12, 35392 Giessen, Germany
| | - Bernd T Schmeck
- University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany; Institute for Lung Research, Department of Respiratory and Critical Care Medicine, University Medical Center Marburg, Center for Synthetic Microbiology (SYNMIKRO), Philipps-University, Marburg, Germany, Member of the German Center for Lung Research (DZL), and the German Center of Infection Research (DZIF), Marburg, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus-Liebig-University, D-35392, Giessen, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig University, Giessen, Germany; University of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany; Lung Clinic Waldhof-Elgershausen, Greifenstein, Germany
| |
Collapse
|
98
|
Club Cell Heme Oxygenase-1 Deletion: Effects in Hyperoxia-Exposed Adult Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2908271. [PMID: 32587658 PMCID: PMC7303751 DOI: 10.1155/2020/2908271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/29/2020] [Accepted: 05/16/2020] [Indexed: 02/06/2023]
Abstract
Thioredoxin reductase-1 (TXNRD1) inhibition activates nuclear factor (erythroid-derived 2)-like 2 (Nrf2) responses and prevents acute lung injury (ALI). Heme oxygenase-1 (HO-1) induction following TXNRD1 inhibition is Nrf2-dependent in airway epithelial (club) cells in vitro. The influence of club cell HO-1 on lung development and lung injury responses is poorly understood. The present studies characterized the effects of hyperoxia on club cell-specific HO-1 knockout (KO) mice. These mice were generated by crossing Hmox1 flox mice with transgenic mice expressing cre recombinase under control of the club cell-specific Scgb1a1 promoter. Baseline analyses of lung architecture and function performed in age-matched adult wild-type and KO mice indicated an increased alveolar size and airway resistance in HO-1 KO mice. In subsequent experiments, adult wild-type and HO-1 KO mice were either continuously exposed to >95% hyperoxia or room air for 72 h or exposed to >95 hyperoxia for 48 h followed by recovery in room air for 48 h. Injury was quantitatively assessed by calculating right lung/body weight ratios (g/kg). Analyses indicated an independent effect of hyperoxia but not genotype on right lung/body weight ratios in both wild-type and HO-1 KO mice. The magnitude of increases in right lung/body weight ratios was similar in mice of both genotypes. In the recovery model, an independent effect of hyperoxia but not genotype was also detected. In contrast to the continuous exposure model, right lung/body weight ratio mice were significantly elevated in HO-1 KO but not wild-type mice. Though club cell HO-1 does not alter hyperoxic sensitivity in adult mice, it significantly influences lung development and resolution of lung injury following acute hyperoxic exposure.
Collapse
|
99
|
Abstract
This review is focusing on the understanding of various factors and components governing and controlling the occurrence of ventricular arrhythmias including (i) the role of various ion channel-related changes in the action potential (AP), (ii) electrocardiograms (ECGs), (iii) some important arrhythmogenic mediators of reperfusion, and pharmacological approaches to their attenuation. The transmembrane potential in myocardial cells is depending on the cellular concentrations of several ions including sodium, calcium, and potassium on both sides of the cell membrane and active or inactive stages of ion channels. The movements of Na+, K+, and Ca2+ via cell membranes produce various currents that provoke AP, determining the cardiac cycle and heart function. A specific channel has its own type of gate, and it is opening and closing under specific transmembrane voltage, ionic, or metabolic conditions. APs of sinoatrial (SA) node, atrioventricular (AV) node, and Purkinje cells determine the pacemaker activity (depolarization phase 4) of the heart, leading to the surface manifestation, registration, and evaluation of ECG waves in both animal models and humans. AP and ECG changes are key factors in arrhythmogenesis, and the analysis of these changes serve for the clarification of the mechanisms of antiarrhythmic drugs. The classification of antiarrhythmic drugs may be based on their electrophysiological properties emphasizing the connection between basic electrophysiological activities and antiarrhythmic properties. The review also summarizes some important mechanisms of ventricular arrhythmias in the ischemic/reperfused myocardium and permits an assessment of antiarrhythmic potential of drugs used for pharmacotherapy under experimental and clinical conditions.
Collapse
Affiliation(s)
- Arpad Tosaki
- Department of Pharmacology, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
100
|
Thongtip S, Siviroj P, Prapamontol T, Deesomchok A, Wisetborisut A, Nangola S, Khacha-ananda S. A suitable biomarker of effect, club cell protein 16, from crystalline silica exposure among Thai stone-carving workers. Toxicol Ind Health 2020; 36:287-296. [DOI: 10.1177/0748233720920137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Exposure to respirable crystalline silica (RCS) reportedly induces chronic lung injury. We investigated the association between RCS exposure and two biomarkers of the effect, plasma club cell protein 16 (CC16) and heme oxygenase-1 (HO-1) levels, in stone-carving workers. Fifty-seven exposed workers (EWs) and 20 unexposed workers (UWs) were enrolled onto the study. Cumulative exposure to RCS was individually estimated using a filter-based gravimetric method. The plasma CC16 and HO-1 levels were determined using commercial kits. The 8-h time-weighted average for RCS concentration in the EW was significantly greater than this concentration in the UW ( p < 0.001). The health risk characterization for RCS exposure expressed as a hazard quotient (HQ) indicated that crystalline silica might be a risk factor where there is chronic exposure (HQ = 4.48). The EW group presented a significant decrease in CC16 and an increase in HO-1 levels in comparison to the UW group ( p < 0.001). In addition, we found a significant association between RCS concentration and plasma CC16 only. Therefore, our findings representing a significant decrease in CC16 in the plasma of stone-carving workers and this biological marker were significantly associated with RCS concentration. Our data indicated that CC16 might be a suitable biomarker to use to predict the health risk to stone-carving workers of exposure to RCS.
Collapse
Affiliation(s)
- Sakesun Thongtip
- Department of Environmental Health, Faculty of Medicine, University of Phayao, Phayao, Thailand
| | - Penprapa Siviroj
- Department of Community Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Tippawan Prapamontol
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Athavudh Deesomchok
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anawat Wisetborisut
- Department of Family Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sawitree Nangola
- Department of Clinical Immunology and Transfusion Sciences, Faculty of Allied Health Sciences, University of Phayao, Phayao, Thailand
| | - Supakit Khacha-ananda
- Department of Forensic Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|