1
|
Baguma M, Kessels S, Bito V, Brône B, Triller A, Maynard S, Legendre P, Rigo JM, Le Corronc H, Chabwine JN. New insight into the molecular etiopathogenesis of konzo: Cyanate could be a plausible neurotoxin contributing to konzo, contrary to thiocyanate. Neurotoxicology 2024; 105:323-333. [PMID: 39608576 DOI: 10.1016/j.neuro.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
INTRODUCTION Chronic cassava-derived cyanide poisoning is associated with the appearance of konzo, a tropical spastic paraparesis due to selective upper motor neuron damage. Whether the disease is caused by a direct action of cyanide or its metabolites is still an open question. This preliminary study assessed the neurotoxic effects of thiocyanate (SCN) and cyanate (OCN), two cyanide metabolites hypothesized to be plausible toxic agents in konzo. METHODS Cultured mouse neuroblastoma (Neuro-2A) and human neuroblastoma (SH-SY5Y) cell lines were incubated (24, 48, and 72 hours) in sodium OCN or sodium SCN in a disease-relevant concentration range. Cell viability, caspase (3, 8, and 9) activities, and reactive oxygen species (ROS) generation were evaluated using appropriate assay kits. Additionally, electrophysiological responses induced by OCN and SCN in primary spinal cord neurons (from Sprague Dawley rats) were assessed by whole-cell patch-clamp techniques. RESULTS Both OCN and SCN were toxic in a dose-dependent way, even if SCN toxicity appeared at very high concentrations (30 mM, corresponding to more than 100-fold higher than normal plasmatic levels), contrary to OCN (0.3-3 mM). OCN was markedly more toxic in a poor culture medium (MEM; IC50 = 3.2 mM) compared to a glucose- and amino acid-rich medium (DMEM; IC50=7.6 mM). OCN treatment increased the ROS generation by 8.9 folds, as well as the Caspase-3, Caspase-8, and Caspase-9 activities by 3.2, 2.5, and 2.6 folds, respectively. Finally, OCN (and SCN to a lesser extent) induced depolarizing currents in primary spinal cord neurons, through an activation of ionotropic glutamate receptors. CONCLUSION Our results suggest OCN as the most plausible neurotoxic agent involved in konzo, while SCN toxicity could be questioned at such high concentrations. Also, they support apoptosis, oxidative stress, and excitotoxicity as probable mechanisms of OCN neurotoxicity.
Collapse
Affiliation(s)
- Marius Baguma
- UHasselt, Neuroscience Research Group (NEURO), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium; Université Catholique de Bukavu (UCB), Center for Tropical Diseases and Global Health (CTDGH), Bukavu, Democratic Republic Congo; Hôpital Provincial Général de Référence de Bukavu (HPGRB), Neurology Ward, Bukavu, Democratic Republic Congo.
| | - Sofie Kessels
- UHasselt, Neuroscience Research Group (NEURO), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium
| | - Virginie Bito
- UHasselt, Cardio & Organ Systems (COST), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium
| | - Bert Brône
- UHasselt, Neuroscience Research Group (NEURO), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium
| | - Antoine Triller
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, Inserm, Université PSL, Paris, France
| | - Stéphanie Maynard
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, Inserm, Université PSL, Paris, France
| | - Pascal Legendre
- Sorbonne Université, INSERM, CNRS, Neurosciences Paris Seine - Institut de Biologie, Paris Seine (NPS - IBPS), Paris 75005, France
| | - Jean-Michel Rigo
- UHasselt, Neuroscience Research Group (NEURO), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium
| | - Hervé Le Corronc
- Sorbonne Université, INSERM, CNRS, Neurosciences Paris Seine - Institut de Biologie, Paris Seine (NPS - IBPS), Paris 75005, France; Université d'Angers, Angers, France
| | - Joelle Nsimire Chabwine
- Université Catholique de Bukavu (UCB), Center for Tropical Diseases and Global Health (CTDGH), Bukavu, Democratic Republic Congo; University of Fribourg, Faculty of Science and Medicine, Department of Neuroscience and Movement Science, Fribourg, Switzerland
| |
Collapse
|
2
|
Nayak M, Sonowal L, Pradhan L, Upadhyay A, Kamath P, Mukherjee S. Multifunctional (4-in-1) Therapeutic Applications of Nickel Thiocyanate Nanoparticles Impregnated Cotton Gauze as Antibacterial, Antibiofilm, Antioxidant and Wound Healing Agent. Chem Asian J 2024; 19:e202400187. [PMID: 38665128 DOI: 10.1002/asia.202400187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/16/2024] [Indexed: 08/27/2024]
Abstract
The wounds, arises from accidents, burns, surgeries, diabetes, and trauma, can significantly impact well-being and present persistent clinical challenges. Ideal wound dressings should be flexible, stable, antibacterial, antioxidant and anti-inflammatory in nature, facilitating a scarless rapid wound healing. Initiatives were taken to create antibacterial cotton fabrics by incorporating agents like antibiotics and metallic nanoparticles. However, due to a lack of multifunctionality, these materials were not highly effective in causing scarless and rapid wound healing. In this article, nickel thiocyanate nanoparticle (NiSCN-NPs) impregnated cotton gauze wound dressing (NiSCN-CG) was developed. These nanoparticles were non-toxic to normal human cell lines till 1 mg/mL dose and did not cause skin irritation in the rat model. Further, NiSCN-NPs exhibited antimicrobial, antibiofilm and antioxidant activities confirmed using different in vitro experiments. In vivo wound healing studies in rat models using NiSCN-CG demonstrated rapid scarless wound healing. The nickel thiocyanate impregnated cotton gauze presents a novel approach in scarless wound healing, and as an antimicrobial agent, offering a promising solution for diverse wounds and infections in the future.
Collapse
Affiliation(s)
- Malay Nayak
- School of Biomedical Engineering, IIT (BHU), Varanasi, Varanasi, 221005, Uttar Pradesh, India
| | - Lidiya Sonowal
- School of Biomedical Engineering, IIT (BHU), Varanasi, Varanasi, 221005, Uttar Pradesh, India
| | - Lipi Pradhan
- School of Biomedical Engineering, IIT (BHU), Varanasi, Varanasi, 221005, Uttar Pradesh, India
| | - Anjali Upadhyay
- School of Biomedical Engineering, IIT (BHU), Varanasi, Varanasi, 221005, Uttar Pradesh, India
| | - Prajwal Kamath
- School of Biomedical Engineering, IIT (BHU), Varanasi, Varanasi, 221005, Uttar Pradesh, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, IIT (BHU), Varanasi, Varanasi, 221005, Uttar Pradesh, India
| |
Collapse
|
3
|
Vaillancourt M, Aguilar D, Fernandes SE, Jorth PA. A chronic Pseudomonas aeruginosa mouse lung infection modeling the pathophysiology and inflammation of human cystic fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617039. [PMID: 39416002 PMCID: PMC11482824 DOI: 10.1101/2024.10.07.617039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Investigation of chronic cystic fibrosis (CF) lung infections has been limited by a lack of murine models that reproduce obstructive lung pathology, chronicity of bacterial infections, and complex inflammation in human CF lung pathology. Three different approaches have been used separately to address these limitations, including using transgenic Scnn1b-Tg mice overexpressing a lung epithelial sodium channel to mimic the mucus-rich and hyperinflammatory CF lung environment, using synthetic CF sputum medium (SCFM) in an acute infection to induce bacterial phenotypes consistent with human CF, or using agar beads to promote chronic infections. Here, we combine these three models to establish a chronic Pseudomonas aeruginosa lung infection model using SCFM agar beads and Scnn1b-Tg mice (SCFM-Tg-mice) to recapitulate nutrients, mucus, and inflammation characteristic of the human CF lung environment. Like people with CF, SCFM-Tg-mice failed to clear bacterial infections. Lung function measurements showed that infected SCFM-Tg-mice had decreased inspiratory capacity and compliance, elevated airway resistance, and significantly reduced FVC and FEV0.1. Using spectral flow cytometry and multiplex cytokine arrays we show that, like people with CF, SCFM-Tg-mice developed inflammation characterized by eosinophil infiltration and Th2 lymphocytic cytokine responses. Chronically infected SCFM-Tg-mice developed an exacerbated mix of innate and Th1, Th2, and Th17-mediated inflammation, causing higher lung cellular damage, and elevated numbers of unusual Siglec F+ neutrophils. Thus, SCFM-Tg-mice represents a powerful tool to investigate bacterial pathogenesis and potential treatments for chronic CF lung infections and reveal a potential role for Siglec F+ neutrophils in CF inflammation.
Collapse
Affiliation(s)
- Mylene Vaillancourt
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Diane Aguilar
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sheryl E. Fernandes
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter A. Jorth
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
4
|
Wang D, Zhang Y, He Y, Song F, Tang Y, Chen L, Wang Y, Yang F, Yao X. Associations of Perchlorate, Nitrate, and Thiocyanate with Bone Mineral Density in the US General Population: A Multi-Cycle Study of NHANES 2011-2018. Nutrients 2024; 16:2658. [PMID: 39203795 PMCID: PMC11487404 DOI: 10.3390/nu16162658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Perchlorate, nitrate, and thiocyanate are widely recognized as endocrine disrupting chemicals, which are closely related to thyroid function. Animal and human studies show a correlation between thyroid hormone and bone mineral density (BMD). However, it remains unknown whether perchlorate, nitrate, and thiocyanate were associated with BMD. This study aimed to explore the association between perchlorate, nitrate, and thiocyanate exposure with BMD. METHOD A cross-sectional analysis among 5607 participants from the 2011-2018 National Health and Nutrition Examination Survey (NHANES) was conducted in the present study. Perchlorate, nitrate, and thiocyanate were detected in urine by ion chromatography. Survey-weighted generalized linear regression, restricted cubic splines, and qgcomp models were used to assess the association of BMDs with single and mixed perchlorate, nitrate, and thiocyanate exposures. In addition, age, gender, and BMI stratified these associations. RESULTS Negative associations were found between perchlorate and nitrate with BMDs. Furthermore, based on the qgcomp model results, the combined association of perchlorate, nitrate, and thiocyanate exposure was negatively associated with BMDs (β = -0.017, 95% CI: -0.041, -0.024 for total BMD; β = -0.017, 95% CI: -0.029, -0.005 for lumbar BMD). Additionally, there was a significant effect after gender, age, and BMI stratification between perchlorate, nitrate, and thiocyanate with BMDs in the normal weight group (β = -0.015, 95% CI: -0.020, -0.011 for total BMD; β = -0.022, 95% CI: -0.028, -0.016 for lumbar BMD) and children and adolescents group (β = -0.025, 95% CI: -0.031, -0.019 for total BMD; β -0.017, 95% CI: -0.029, -0.005 for lumbar BMD). CONCLUSIONS The present study indicated a negative correlation between BMDs and urinary perchlorate, nitrate, and thiocyanate levels, with nitrate being the main contributor to the mixture effect. People with normal weight and children and adolescents were more likely to be affected.
Collapse
Affiliation(s)
- Donglan Wang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
| | - Ying Zhang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
| | - Yayu He
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
| | - Fengmei Song
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
| | - Yan Tang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
| | - Limou Chen
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
| | - Yangcan Wang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
| | - Fei Yang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha 410000, China
| | - Xueqiong Yao
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421009, China (Y.H.)
- Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421009, China
| |
Collapse
|
5
|
Ashtiwi NM, Kim SO, Chandler JD, Rada B. The therapeutic potential of thiocyanate and hypothiocyanous acid against pulmonary infections. Free Radic Biol Med 2024; 219:104-111. [PMID: 38608822 PMCID: PMC11088529 DOI: 10.1016/j.freeradbiomed.2024.04.217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Hypothiocyanous acid (HOSCN) is an endogenous oxidant produced by peroxidase oxidation of thiocyanate (SCN-), an ubiquitous sulfur-containing pseudohalide synthesized from cyanide. HOSCN serves as a potent microbicidal agent against pathogenic bacteria, viruses, and fungi, functioning through thiol-targeting mechanisms, independent of currently approved antimicrobials. Additionally, SCN- reacts with hypochlorous acid (HOCl), a highly reactive oxidant produced by myeloperoxidase (MPO) at sites of inflammation, also producing HOSCN. This imparts both antioxidant and antimicrobial potential to SCN-. In this review, we discuss roles of HOSCN/SCN- in immunity and potential therapeutic implications for combating infections.
Collapse
Affiliation(s)
- Nuha Milad Ashtiwi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Susan O Kim
- Pediatrics, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Joshua D Chandler
- Pediatrics, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep Medicine, Emory University, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| |
Collapse
|
6
|
Kim SO, Shapiro JP, Cottrill KA, Collins GL, Shanthikumar S, Rao P, Ranganathan S, Stick SM, Orr ML, Fitzpatrick AM, Go YM, Jones DP, Tirouvanziam RM, Chandler JD. Substrate-dependent metabolomic signatures of myeloperoxidase activity in airway epithelial cells: Implications for early cystic fibrosis lung disease. Free Radic Biol Med 2023; 206:180-190. [PMID: 37356776 PMCID: PMC10513041 DOI: 10.1016/j.freeradbiomed.2023.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/14/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023]
Abstract
Myeloperoxidase (MPO) is released by neutrophils in inflamed tissues. MPO oxidizes chloride, bromide, and thiocyanate to produce hypochlorous acid (HOCl), hypobromous acid (HOBr), and hypothiocyanous acid (HOSCN), respectively. These oxidants are toxic to pathogens, but may also react with host cells to elicit biological activity and potential toxicity. In cystic fibrosis (CF) and related diseases, increased neutrophil inflammation leads to increased airway MPO and airway epithelial cell (AEC) exposure to its oxidants. In this study, we investigated how equal dose-rate exposures of MPO-derived oxidants differentially impact the metabolome of human AECs (BEAS-2B cells). We utilized enzymatic oxidant production with rate-limiting glucose oxidase (GOX) coupled to MPO, and chloride, bromide (Br-), or thiocyanate (SCN-) as substrates. AECs exposed to GOX/MPO/SCN- (favoring HOSCN) were viable after 24 h, while exposure to GOX/MPO (favoring HOCl) or GOX/MPO/Br- (favoring HOBr) developed cytotoxicity after 6 h. Cell glutathione and peroxiredoxin-3 oxidation were insufficient to explain these differences. However, untargeted metabolomics revealed GOX/MPO and GOX/MPO/Br- diverged significantly from GOX/MPO/SCN- for dozens of metabolites. We noted methionine sulfoxide and dehydromethionine were significantly increased in GOX/MPO- or GOX/MPO/Br--treated cells, and analyzed them as potential biomarkers of lung damage in bronchoalveolar lavage fluid from 5-year-olds with CF (n = 27). Both metabolites were associated with increasing bronchiectasis, neutrophils, and MPO activity. This suggests MPO production of HOCl and/or HOBr may contribute to inflammatory lung damage in early CF. In summary, our in vitro model enabled unbiased identification of exposure-specific metabolite products which may serve as biomarkers of lung damage in vivo. Continued research with this exposure model may yield additional oxidant-specific biomarkers and reveal explicit mechanisms of oxidant byproduct formation and cellular redox signaling.
Collapse
Affiliation(s)
- Susan O Kim
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, USA
| | - Joseph P Shapiro
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, USA
| | - Kirsten A Cottrill
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, USA
| | - Genoah L Collins
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, USA
| | - Shivanthan Shanthikumar
- Respiratory and Sleep Medicine, Royal Children's Hospital, Parkville, VIC, Australia; Respiratory Diseases, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Padma Rao
- Medical Imaging, Royal Children's Hospital, Parkville, VIC, Australia
| | - Sarath Ranganathan
- Respiratory and Sleep Medicine, Royal Children's Hospital, Parkville, VIC, Australia; Respiratory Diseases, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Perth, Western Australia, Australia
| | - Michael L Orr
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Anne M Fitzpatrick
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Young-Mi Go
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Rabindra M Tirouvanziam
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Joshua D Chandler
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, USA; Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA.
| |
Collapse
|
7
|
Li W, Wu H, Xu X, Zhang Y. Environmental exposure to perchlorate, nitrate, and thiocyanate in relation to chronic kidney disease in the general US population, NHANES 2005-2016. Chin Med J (Engl) 2023:00029330-990000000-00571. [PMID: 37154820 DOI: 10.1097/cm9.0000000000002586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Few studies have explored the impact of perchlorate, nitrate, and thiocyanate (PNT) on kidney function. This study aimed to evaluate the association of urinary levels of PNT with renal function as well as the prevalence of chronic kidney disease (CKD) among the general population in the United States. METHODS This analysis included data from 13,373 adults (≥20 years) from the National Health and Nutrition Examination Survey 2005 to 2016. We used multivariable linear and logistic regression, to explore the associations of urinary PNT with kidney function. Restricted cubic splines were used to assess the potentially non-linear relationships between PNT exposure and outcomes. RESULTS After traditional creatinine adjustment, perchlorate (P-traditional) was positively associated with estimated glomerular filtration rate (eGFR) (adjusted β: 2.75; 95% confidence interval [CI]: 2.25 to 3.26; P < 0.001), and negatively associated with urinary albumin-to-creatinine ratio (ACR) (adjusted β: -0.05; 95% CI: -0.07 to -0.02; P = 0.001) in adjusted models. After both traditional and covariate-adjusted creatinine adjustment, urinary nitrate and thiocyanate were positively associated with eGFR (all P values <0.05), and negatively associated with ACR (all P values <0.05); higher nitrate or thiocyanate was associated with a lower risk of CKD (all P values <0.001). Moreover, there were L-shaped non-linear associations between nitrate, thiocyanate, and outcomes. In the adjusted models, for quartiles of PNT, statistically significant dose-response associations were observed in most relationships. Most results were consistent in the stratified and sensitivity analyses. CONCLUSIONS Exposures to PNT might be associated with kidney function, indicating a potential beneficial effect of environmental PNT exposure (especially nitrate and thiocyanate) on the human kidney.
Collapse
Affiliation(s)
- Wei Li
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hong Wu
- Department of Liver Surgery and Liver Transplantation, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Xuewen Xu
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yange Zhang
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
8
|
Reynolds WF, Malle E, Maki RA. Thiocyanate Reduces Motor Impairment in the hMPO-A53T PD Mouse Model While Reducing MPO-Oxidation of Alpha Synuclein in Enlarged LYVE1/AQP4 Positive Periventricular Glymphatic Vessels. Antioxidants (Basel) 2022; 11:antiox11122342. [PMID: 36552550 PMCID: PMC9774557 DOI: 10.3390/antiox11122342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) is due to the oxidation of alpha synuclein (αSyn) contributing to motor impairment. We developed a transgenic mouse model of PD that overexpresses the mutated human αSyn gene (A53T) crossed to a mouse expressing the human MPO gene. This model exhibits increased oxidation and chlorination of αSyn leading to greater motor impairment. In the current study, the hMPO-A53T mice were treated with thiocyanate (SCN-) which is a favored substrate of MPO as compared to chlorine. We show that hMPO-A53T mice treated with SCN- have less chlorination in the brain and show an improvement in motor skills compared to the nontreated hMPO-A53T mice. Interestingly, in the hMPO-A53T mice we found a possible link between MPO-related disease and the glymphatic system which clears waste including αSyn from the brain. The untreated hMPO-A53T mice exhibited an increase in the size of periventricular glymphatic vessels expressing the glymphatic marker LYVE1 and aquaporin 4 (AQP4). These vessels also exhibited an increase in MPO and HOCl-modified epitopes in the glymphatic vessels correlating with loss of ependymal cells lining the ventricles. These findings suggest that MPO may significantly promote the impairment of the glymphatic waste removal system thus contributing to neurodegeneration in PD. Moreover, the inhibition of MPO chlorination/oxidation by SCN- may provide a potential therapeutic approach to this disease.
Collapse
Affiliation(s)
- Wanda F. Reynolds
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- Correspondence:
| | - Ernst Malle
- Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | - Richard A. Maki
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
9
|
The Use of Thiocyanate Formulations to Create Manganese Porphyrin Antioxidants That Supplement Innate Immunity. Antioxidants (Basel) 2022; 11:antiox11071252. [PMID: 35883743 PMCID: PMC9311894 DOI: 10.3390/antiox11071252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/02/2022] [Accepted: 06/09/2022] [Indexed: 01/25/2023] Open
Abstract
The innate immune response to infection results in inflammation and oxidative damage, creating a paradox where most anti-inflammatory and antioxidant therapies can further suppress an already inadequate immune response. We have previously reported the beneficial effects of the exogenous supplementation of innate immunity with small pseudohalide thiocyanate (−SCN) in a mouse model of a cystic fibrosis (CF) lung infection and inflammation. The object of this study was to evaluate the use of −SCN as a counter anion for cationic manganese porphyrin (MnP) catalytic antioxidants, which could increase the parent compound’s antioxidant spectrum against hypohalous acids while supplementing innate immunity. The antioxidant activities of the parent compound were examined, as its chloride salt was compared with the −SCN-anion exchanged compound, (MnP(SCN) versus MnP(Cl)). We measured the superoxide dismutase activity spectrophotometrically and performed hydrogen peroxide scavenging using oxygen and hydrogen peroxide electrodes. Peroxidase activity was measured using an amplex red assay. The inhibition of lipid peroxidation was assessed using a thiobarbituric acid reactive species (TBARS) assay. The effects of the MnP compounds on macrophage phagocytosis were assessed by flow cytometry. The abilities of the MnP(Cl) formulations to protect human bronchiolar epithelial cells against hypochlorite (HOCl) and glycine chloramine versus their MnP(SCN) formulations were assessed using a cell viability assay. We found that anions exchanging out the chloride for −SCN improved the cellular bioavailability but did not adversely affect the cell viability or phagocytosis and that they switched hydrogen-peroxide scavenging from a dismutation reaction to a peroxidase reaction. In addition, the −SCN formulations improved the ability of MnPs to protect human bronchiolar epithelial cells against hypochlorous acid (HOCl) and glycine chloramine toxicity. These novel types of antioxidants may be more beneficial in treating lung disease that is associated with chronic infections or acute infectious exacerbations.
Collapse
|
10
|
Abstract
Heme-containing peroxidases are widely distributed in the animal and plant kingdoms and play an important role in host defense by generating potent oxidants. Myeloperoxidase (MPO), the prototype of heme-containing peroxidases, exists in neutrophils and monocytes. MPO has a broad spectrum of microbial killing. The difficulty of producing MPO at a large scale hinders its study and utilization. This study aimed to overexpress recombinant human MPO and characterize its microbicidal activities in vitro and in vivo. A human HEK293 cell line stably expressing recombinant MPO (rMPO) was established as a component of this study. rMPO was overexpressed and purified for studies on its biochemical and enzymatic properties, as well as its microbicidal activities. In this study, rMPO was secreted into culture medium as a monomer. rMPO revealed enzymatic activity similar to that of native MPO. rMPO, like native MPO, was capable of killing a broad spectrum of microorganisms, including Gram-negative and -positive bacteria and fungi, at low nM levels. Interestingly, rMPO could kill antibiotic-resistant bacteria, making it very useful for treatment of nosocomial infections and mixed infections. The administration of rMPO significantly reduced the morbidity and mortality of murine lung infections induced by Pseudomonas aeruginosa or methicillin-resistant Staphylococcus aureus. In animal safety tests, the administration of 100 nM rMPO via tail vein did not result in any sign of toxic effects. Taken together, the data suggest that rMPO purified from a stably expressing human cell line is a new class of antimicrobial agents with the ability to kill a broad spectrum of pathogens, including bacteria and fungi with or without drug resistance. IMPORTANCE Over the past 2 decades, more than 20 new infectious diseases have emerged. Unfortunately, novel antimicrobial therapeutics are discovered at much lower rates. Infections caused by resistant microorganisms often fail to respond to conventional treatment, resulting in prolonged illness, greater risk of death, and high health care costs. Currently, this is best seen with the lack of a cure for coronavirus disease 2019 (COVID-19). To combat such untreatable microorganisms, there is an urgent need to discover new classes of antimicrobial agents. Myeloperoxidase (MPO) plays an important role in host defense. The difficulty of producing MPO on a large scale hinders its study and utilization. We have produced recombinant MPO at a large scale and have characterized its antimicrobial activities. Most importantly, recombinant MPO significantly reduced the morbidity and mortality of murine pneumonia induced by Pseudomonas aeruginosa or methicillin-resistant Staphylococcus aureus. Our data suggest that recombinant MPO from human cells is a new class of antimicrobials with a broad spectrum of activity.
Collapse
|
11
|
Sarr D, Gingerich AD, Asthiwi NM, Almutairi F, Sautto GA, Ecker J, Nagy T, Kilgore MB, Chandler JD, Ross TM, Tripp RA, Rada B. Dual oxidase 1 promotes antiviral innate immunity. Proc Natl Acad Sci U S A 2021; 118:e2017130118. [PMID: 34168077 PMCID: PMC8256044 DOI: 10.1073/pnas.2017130118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Indexed: 12/30/2022] Open
Abstract
Dual oxidase 1 (DUOX1) is an NADPH oxidase that is highly expre-ssed in respiratory epithelial cells and produces H2O2 in the airway lumen. While a line of prior in vitro observations suggested that DUOX1 works in partnership with an airway peroxidase, lactoperoxidase (LPO), to produce antimicrobial hypothiocyanite (OSCN-) in the airways, the in vivo role of DUOX1 in mammalian organisms has remained unproven to date. Here, we show that Duox1 promotes antiviral innate immunity in vivo. Upon influenza airway challenge, Duox1-/- mice have enhanced mortality, morbidity, and impaired lung viral clearance. Duox1 increases the airway levels of several cytokines (IL-1β, IL-2, CCL1, CCL3, CCL11, CCL19, CCL20, CCL27, CXCL5, and CXCL11), contributes to innate immune cell recruitment, and affects epithelial apoptosis in the airways. In primary human tracheobronchial epithelial cells, OSCN- is generated by LPO using DUOX1-derived H2O2 and inactivates several influenza strains in vitro. We also show that OSCN- diminishes influenza replication and viral RNA synthesis in infected host cells that is inhibited by the H2O2 scavenger catalase. Binding of the influenza virus to host cells and viral entry are both reduced by OSCN- in an H2O2-dependent manner in vitro. OSCN- does not affect the neuraminidase activity or morphology of the influenza virus. Overall, this antiviral function of Duox1 identifies an in vivo role of this gene, defines the steps in the infection cycle targeted by OSCN-, and proposes that boosting this mechanism in vivo can have therapeutic potential in treating viral infections.
Collapse
Affiliation(s)
- Demba Sarr
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Aaron D Gingerich
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Nuha Milad Asthiwi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Faris Almutairi
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602
| | - Giuseppe A Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Jeffrey Ecker
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Tamás Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Matthew B Kilgore
- Department of Pediatrics, Division of Pulmonary, Allergy and Immunology, Cystic Fibrosis, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322
- Center for Cystic Fibrosis and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA 30322
| | - Joshua D Chandler
- Department of Pediatrics, Division of Pulmonary, Allergy and Immunology, Cystic Fibrosis, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA 30322
- Center for Cystic Fibrosis and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA 30322
| | - Ted M Ross
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602;
| |
Collapse
|
12
|
Brao KJ, Wille BP, Lieberman J, Ernst RK, Shirtliff ME, Harro JM. Scnn1b-Transgenic BALB/c Mice as a Model of Pseudomonas aeruginosa Infections of the Cystic Fibrosis Lung. Infect Immun 2020; 88:e00237-20. [PMID: 32631918 PMCID: PMC7440770 DOI: 10.1128/iai.00237-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa is responsible for much of the morbidity and mortality associated with cystic fibrosis (CF), a condition that predisposes patients to chronic lung infections. P. aeruginosa lung infections are difficult to treat because P. aeruginosa adapts to the CF lung, can develop multidrug resistance, and can form biofilms. Despite the clinical significance of P. aeruginosa, modeling P. aeruginosa infections in CF has been challenging. Here, we characterize Scnn1b-transgenic (Tg) BALB/c mice as P. aeruginosa lung infection models. Scnn1b-Tg mice overexpress the epithelial Na+ channel (ENaC) in their lungs, driving increased sodium absorption that causes lung pathology similar to CF. We intranasally infected Scnn1b-Tg mice and wild-type littermates with the laboratory P. aeruginosa strain PAO1 and CF clinical isolates and then assessed differences in bacterial clearance, cytokine responses, and histological features up to 12 days postinfection. Scnn1b-Tg mice carried higher bacterial burdens when infected with biofilm-grown rather than planktonic PAO1; Scnn1b-Tg mice also cleared infections more slowly than their wild-type littermates. Infection with PAO1 elicited significant increases in proinflammatory and Th17-linked cytokines on day 3. Scnn1b-Tg mice infected with nonmucoid early CF isolates maintained bacterial burdens and mounted immune responses similar to those of PAO1-infected Scnn1b-Tg mice. In contrast, Scnn1b-Tg mice infected with a mucoid CF isolate carried high bacterial burdens, produced significantly more interleukin 1β (IL-1β), IL-13, IL-17, IL-22, and KC, and showed severe immune cell infiltration into the bronchioles. Taken together, these results show the promise of Scnn1b-Tg mice as models of early P. aeruginosa colonization in the CF lung.
Collapse
Affiliation(s)
- Kristen J Brao
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Brendan P Wille
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland, USA
| | - Joshua Lieberman
- Division of Microbiology, Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mark E Shirtliff
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Janette M Harro
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Lee EH, Shin MH, Gi M, Park J, Song D, Hyun YM, Ryu JH, Seong JK, Jeon Y, Han G, Namkung W, Park MS, Choi JY. Inhibition of Pendrin by a small molecule reduces Lipopolysaccharide-induced acute Lung Injury. Theranostics 2020; 10:9913-9922. [PMID: 32929324 PMCID: PMC7481407 DOI: 10.7150/thno.46417] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Pendrin is encoded by SLC26A4 and its mutation leads to congenital hearing loss. Additionally, pendrin is up-regulated in inflammatory airway diseases such as chronic obstructive pulmonary disease, allergic rhinitis, and asthma. In this study, the effects of a novel pendrin inhibitor, YS-01, were investigated in an LPS-induced acute lung injury (ALI) mice model, and the mechanism underlying the effect of YS-01 was examined. Methods: Lipopolysaccharide (LPS, 10 mg/kg) was intranasally instilled in wild type (WT) and pendrin-null mice. YS-01 (10 mg/kg) was administered intra-peritoneally before or after LPS inhalation. Lung injury parameters were assessed in the lung tissue and bronchoalveolar lavage fluid (BALF). Pendrin levels in the BALF of 41 patients with acute respiratory distress syndrome (ARDS) due to pneumonia and 25 control (solitary pulmonary nodule) patients were also measured. Results: LPS instillation induced lung injury in WT mice but not in pendrin-null mice. Pendrin expression was increased by LPS stimulation both in vitro and in vivo. YS-01 treatment dramatically attenuated lung injury and reduced BALF cell counts and protein concentration after LPS instillation in WT mice. Proinflammatory cytokines and NF-κB activation were suppressed by YS-01 treatment in LPS-induced ALI mice. In BALF of patients whose ARDS was caused by pneumonia, pendrin expression was up-regulated compared to that in controls (mean, 24.86 vs. 6.83 ng/mL, P < 0.001). Conclusions: A novel pendrin inhibitor, YS-01, suppressed lung injury in LPS-induced ALI mice and our data provide a new strategy for the treatment of inflammatory airway diseases including sepsis-induced ALI.
Collapse
|
14
|
Liu Y, Burton T, Rayner BS, San Gabriel PT, Shi H, El Kazzi M, Wang X, Dennis JM, Ahmad G, Schroder AL, Gao A, Witting PK, Chami B. The role of sodium thiocyanate supplementation during dextran sodium sulphate-stimulated experimental colitis. Arch Biochem Biophys 2020; 692:108490. [PMID: 32721434 DOI: 10.1016/j.abb.2020.108490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 12/15/2022]
Abstract
Ulcerative colitis is a condition characterised by the infiltration of leukocytes into the gastrointestinal wall. Leukocyte-MPO catalyses hypochlorous acid (HOCl) and hypothiocyanous acid (HOSCN) formation from chloride (Cl-) and thiocyanous (SCN-) anions, respectively. While HOCl indiscriminately oxidises biomolecules, HOSCN primarily targets low-molecular weight protein thiols. Oxidative damage mediated by HOSCN may be reversible, potentially decreasing MPO-associated host tissue destruction. This study investigated the effect of SCN- supplementation in a model of acute colitis. Female mice were supplemented dextran sodium sulphate (DSS, 3% w/v) in the presence of 10 mM Cl- or SCN- in drinking water ad libitum, or with salts (NaCl and NaSCN only) or water only (controls). Behavioural studies showed mice tolerated NaSCN and NaCl-treated water with water-seeking frequency. Ion-exchange chromatography showed increased fecal and plasma SCN- levels in thiocyanate supplemented mice; plasma SCN- reached similar fold-increase for smokers. Overall there was no difference in weight loss and clinical score, mucin levels, crypt integrity and extent of cellular infiltration between DSS/SCN- and DSS/Cl- groups. Neutrophil recruitment remained unchanged in DSS-treated mice, as assessed by fecal calprotectin levels. Total thiol and tyrosine phosphatase activity remained unchanged between DSS/Cl- and DSS/SCN- groups, however, colonic tissue showed a trend in decreased 3-chlorotyrosine (1.5-fold reduction, p < 0.051) and marked increase in colonic GCLC, the rate-limiting enzyme in glutathione synthesis. These data suggest that SCN- administration can modulate MPO activity towards a HOSCN-specific pathway, however, this does not alter the development of colitis within a DSS murine model.
Collapse
Affiliation(s)
- Yuyang Liu
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Thomas Burton
- Animal Behavioural Facility, Charles Perkins Centre, School of Medical Sciences and the Bosch Institute, The University of Sydney, NSW, 2006, Australia.
| | - Benjamin Saul Rayner
- Heart Research Institute, Sydney Medical School, The University of Sydney, NSW, 2006, Australia.
| | - Patrick T San Gabriel
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Han Shi
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Mary El Kazzi
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - XiaoSuo Wang
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Joanne M Dennis
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Gulfam Ahmad
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Angie L Schroder
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Antony Gao
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Paul Kenneth Witting
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| | - Belal Chami
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| |
Collapse
|
15
|
Dickerhof N, Huang J, Min E, Michaëlsson E, Lindstedt EL, Pearson JF, Kettle AJ, Day BJ. Myeloperoxidase inhibition decreases morbidity and oxidative stress in mice with cystic fibrosis-like lung inflammation. Free Radic Biol Med 2020; 152:91-99. [PMID: 32142878 DOI: 10.1016/j.freeradbiomed.2020.03.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/23/2020] [Accepted: 03/02/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cystic fibrosis (CF) lung disease is characterized by severe bacterial infections, excessive neutrophilic inflammation and oxidative stress. The neutrophil enzyme myeloperoxidase (MPO), which produces hypochlorous acid, is associated with worse disease outcomes. Therefore, pharmacological inhibition of MPO in the airways has therapeutic potential. We investigated whether treating mice with an MPO inhibitor during pulmonary infection decreases oxidative stress and improves infection outcomes in mice with CF-like lung inflammation without impacting on bacterial clearance. METHODS Transgenic β-epithelial sodium channel (βENaC)-overexpressing mice (n = 10) were infected with Burkholderia multivorans and treated twice daily with the MPO inhibitor AZM198 (125 μmol/kg) or vehicle administered by oral gavage for two days. Bodyweight was recorded daily. MPO activity, markers of oxidative stress, inflammatory cytokines and leukocytes numbers were measured in bronchoalveolar lavage fluid (BALF). Bacterial burden was determined in lung tissue homogenates. RESULTS During the course of infection, mice treated with AZM198 lost less weight than vehicle-treated mice (p < 0.01). MPO activity and glutathione sulfonamide, a hypochlorous acid-specific glutathione oxidation product, were significantly lower in BALF from AZM198-treated mice (p < 0.05). The inflammatory cytokines CXCL1 and TNF-α in BALF and bacterial burden in the lung were not significantly different between treated and control mice. CONCLUSIONS Orally administered AZM198 inhibits MPO activity in epithelial lining fluid. Blocking hypochlorous acid production in epithelial lining fluid during pulmonary infections through inhibition of MPO improves morbidity in mice with CF-like lung inflammation without interfering with clearance of bacteria. Pharmacological inhibition of MPO is an approach to limit destructive oxidative stress in cystic fibrosis lung disease in humans.
Collapse
Affiliation(s)
- Nina Dickerhof
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand.
| | - Jie Huang
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Elysia Min
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Erik Michaëlsson
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eva-Lotte Lindstedt
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - John F Pearson
- Biostatistics and Computational Biology Unit, University of Otago Christchurch, Christchurch, New Zealand
| | - Anthony J Kettle
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Brian J Day
- Department of Medicine, National Jewish Health, Denver, CO, USA
| |
Collapse
|
16
|
Maki RA, Holzer M, Motamedchaboki K, Malle E, Masliah E, Marsche G, Reynolds WF. Human myeloperoxidase (hMPO) is expressed in neurons in the substantia nigra in Parkinson's disease and in the hMPO-α-synuclein-A53T mouse model, correlating with increased nitration and aggregation of α-synuclein and exacerbation of motor impairment. Free Radic Biol Med 2019; 141:115-140. [PMID: 31175983 PMCID: PMC6774439 DOI: 10.1016/j.freeradbiomed.2019.05.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/15/2019] [Accepted: 05/29/2019] [Indexed: 10/26/2022]
Abstract
α-Synuclein (αSyn) is central to the neuropathology of Parkinson's disease (PD) due to its propensity for misfolding and aggregation into neurotoxic oligomers. Nitration/oxidation of αSyn leads to dityrosine crosslinking and aggregation. Myeloperoxidase (MPO) is an oxidant-generating enzyme implicated in neurodegenerative diseases. In the present work we have examined the impact of MPO in PD through analysis of postmortem PD brain and in a novel animal model in which we crossed a transgenic mouse expressing the human MPO (hMPO) gene to a mouse expressing human αSyn-A53T mutant (A53T) (hMPO-A53T). Surprisingly, our results show that in PD substantia nigra, the hMPO gene is expressed in neurons containing aggregates of nitrated αSyn as well as MPO-generated HOCl-modified epitopes. In our hMPO-A53T mouse model, we also saw hMPO expression in neurons but not mouse MPO. In the mouse model, hMPO was expressed in neurons colocalizing with nitrated αSyn, carbamylated lysine, nitrotyrosine, as well as HOCl-modified epitopes/proteins. RNAscope in situ hybridization confirmed hMPO mRNA expression in neurons. Interestingly, the hMPO protein expressed in hMPO-A53T brain is primarily the precursor proMPO, which enters the secretory pathway potentially resulting in interneuronal transmission of MPO and oxidative species. Importantly, the hMPO-A53T mouse model, when compared to the A53T model, exhibited significant exacerbation of motor impairment on rotating rods, balance beams, and wire hang tests. Further, hMPO expression in the A53T model resulted in earlier onset of end stage paralysis. Interestingly, there was a high concentration of αSyn aggregates in the stratum lacunosum moleculare of hippocampal CA2 region, which has been associated in humans with accumulation of αSyn pathology and neural atrophy in dementia with Lewy bodies. This accumulation of αSyn aggregates in CA2 was associated with markers of endoplasmic reticulum (ER) stress and the unfolded protein response with expression of activating transcription factor 4 (ATF4), C/EBP homologous protein (CHOP), MPO, and cleaved caspase-3. Together these findings suggest that MPO plays an important role in nitrative and oxidative damage that contributes to αSyn pathology in synucleinopathies.
Collapse
Affiliation(s)
- Richard A Maki
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Michael Holzer
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - Khatereh Motamedchaboki
- Tumor Initiation & Maintenance Program and NCI Cancer Centre Proteomics Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ernst Malle
- Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Eliezer Masliah
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA; Department Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA; Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - Wanda F Reynolds
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
17
|
The science of licking your wounds: Function of oxidants in the innate immune system. Biochem Pharmacol 2019; 163:451-457. [DOI: 10.1016/j.bcp.2019.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/08/2019] [Indexed: 02/07/2023]
|
18
|
Huang L, Xuan W, Sarna T, Hamblin MR. Comparison of thiocyanate and selenocyanate for potentiation of antimicrobial photodynamic therapy. JOURNAL OF BIOPHOTONICS 2019; 12:e201800092. [PMID: 29885019 PMCID: PMC6286685 DOI: 10.1002/jbio.201800092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/07/2018] [Indexed: 06/08/2023]
Abstract
We have previously shown that antimicrobial photodynamic therapy (aPDT) mediated by different photosensitizers (PS) can be potentiated by a variety of inorganic salts. Potassium thiocyanate (KSCN) potentiated aPDT mediated by methylene blue (MB), while potassium selenocyanate (KSeCN) potentiated aPDT mediated by MB, Rose Bengal and the anionic porphyrin 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrin dihydrochloride. However, the mechanisms of action that were proposed were fundamentally different. In the present study, we compare these two salts (KSCN and KSeCN) with different light-activated PS and different oxidative reactions for killing gram-positive and gram-negative bacteria. Overall KSeCN was more powerful than KSCN, and worked with a wider range of PS, while KSCN only worked with phenothiazinium salts. KSeCN produced killing when cells were added after light suggesting production of a semistable species called selenocyanogen (SeCN)2 . We tested three different oxidative reactions that can all potentially kill bacteria: lead tetraacetate (Pb[OAc]4 ); Fenton reagent (hydrogen peroxide [H2 O2 ] and ferrous sulfate) H2 O2 and horseradish peroxidase (HRP). In every case, KSeCN was substantially more effective (several logs) than KSCN in potentiating the bacterial killing. We conclude that (SeCN)2 is the mediator for aPDT using KSeCN, while sulfur trioxide radical anion is the mediator for KSCN using phenothiaziums. For H2 O2 /HRP with KSCN, hypothiocyanite is proposed to be the antibacterial agent in the literature, while hyposelenocyanite is said not to exist. Pb[OAc]4 is known to produce (SeCN)2 from KSeCN as well as the analogous (SCN)2 from KSCN. The mediators from Fenton reaction are unclear (pseudohalogen radical ions?) Both KSCN (which occurs naturally in the human body) and KSeCN may be clinically applicable.
Collapse
Affiliation(s)
- Liyi Huang
- Department of Infectious Diseases, First Affiliated
Hospital, Guangxi Medical University, Nanning, China
- Wellman Center for Photomedicine, Massachusetts General
Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston,
MA, USA
| | - Weijun Xuan
- Wellman Center for Photomedicine, Massachusetts General
Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston,
MA, USA
- Department of Otorhinolaryngology, Head and Neck Surgery,
First Clinical Medical College and Hospital, Guangxi University of Chinese Medicine,
Nanning, China
| | - Tadeusz Sarna
- Department of Biophysics, Faculty of Biochemistry,
Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General
Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston,
MA, USA
- Harvard-MIT Division of Health Sciences and Technology,
Cambridge, MA, USA
| |
Collapse
|
19
|
Maroney MJ, Hondal RJ. Selenium versus sulfur: Reversibility of chemical reactions and resistance to permanent oxidation in proteins and nucleic acids. Free Radic Biol Med 2018; 127:228-237. [PMID: 29588180 PMCID: PMC6158117 DOI: 10.1016/j.freeradbiomed.2018.03.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/14/2018] [Accepted: 03/18/2018] [Indexed: 12/16/2022]
Abstract
This review highlights the contributions of Jean Chaudière to the field of selenium biochemistry. Chaudière was the first to recognize that one of the main reasons that selenium in the form of selenocysteine is used in proteins is due to the fact that it strongly resists permanent oxidation. The foundations for this important concept was laid down by Al Tappel in the 1960's and even before by others. The concept of oxygen tolerance first recognized in the study of glutathione peroxidase was further advanced and refined by those studying [NiFeSe]-hydrogenases, selenosubtilisin, and thioredoxin reductase. After 200 years of selenium research, work by Marcus Conrad and coworkers studying glutathione peroxidase-4 has provided definitive evidence for Chaudière's original hypothesis (Ingold et al., 2018) [36]. While the reaction of selenium with oxygen is readily reversible, there are many other examples of this phenomenon of reversibility. Many reactions of selenium can be described as "easy in - easy out". This is due to the strong nucleophilic character of selenium to attack electrophiles, but then this reaction can be reversed due to the strong electrophilic character of selenium and the weakness of the selenium-carbon bond. Several examples of this are described.
Collapse
Affiliation(s)
- Michael J Maroney
- Department of Chemistry and Program in Molecular and Cellular Biology, University of Massachusetts, Life Sciences Laboratories, 240 Thatcher Road, Room N373, Amherst, MA 01003-9364, United States
| | - Robert J Hondal
- Department of Biochemistry, 89 Beaumont Ave, Given Building Room B413, Burlington, VT 05405, United States.
| |
Collapse
|
20
|
Chandler JD, Horati H, Walker DI, Pagliano E, Tirouvanziam R, Veltman M, Scholte BJ, Janssens HM, Go YM, Jones DP. Determination of thiocyanate in exhaled breath condensate. Free Radic Biol Med 2018; 126:334-340. [PMID: 30144632 PMCID: PMC6166650 DOI: 10.1016/j.freeradbiomed.2018.08.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/09/2018] [Accepted: 08/11/2018] [Indexed: 01/09/2023]
Abstract
Thiocyanate is a heme peroxidase substrate that scavenges oxidants produced during inflammation and regulates host defense. In cystic fibrosis (CF) patients, increased airway thiocyanate levels are associated with improved lung function. Research on airway thiocyanate is limited, however, because convenient non-invasive airway sampling methods, such as exhaled breath condensate (EBC), yield low concentrations that are difficult to detect with available assays. In the present study, we developed a method for the determination of thiocyanate in dilute samples using isotope dilution headspace gas chromatography-coupled high-resolution, accurate-mass mass spectrometry (GC-HRMS). The method reliably quantified as little as 4 pmol thiocyanate in EBC and could detect even lower amounts. We successfully measured thiocyanate in EBC from seven healthy donors, with a mean ± SD of 27 ± 16 nM and a median inter-assay coefficient of variation of 10.4% over six months. The method was applied to other biological fluids (plasma from the same visit as EBC donation; bronchoalveolar lavage fluid [BALF] from infants with CF; and healthy adult mouse BALF), giving reliable quantification of samples ranging from 10 nM to 100 µM. Thiocyanate concentrations in fluids besides EBC were (from lowest to highest): 0.73 ± 0.39 µM in BALF of healthy adult mice (n = 6); 1.4 ± 1.4 µM in BALF from infants with CF (n = 24); 46 ± 22 µM in the plasma of adult volunteers (n = 7). These results demonstrate the utility of this new method for clinical determination of thiocyanate in EBC and other biological fluids.
Collapse
Affiliation(s)
- Joshua D Chandler
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Hamed Horati
- Department of Pediatric Pulmonology, Erasmus MC, Rotterdam, the Netherlands; Department of Cell Biology, Erasmus MC, Rotterdam, the Netherlands
| | - Douglas I Walker
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Enea Pagliano
- Measurement Science and Standards, National Research Council of Canada, Ottawa, Canada
| | - Rabindra Tirouvanziam
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Mieke Veltman
- Department of Pediatric Pulmonology, Erasmus MC, Rotterdam, the Netherlands; Department of Cell Biology, Erasmus MC, Rotterdam, the Netherlands
| | - Bob J Scholte
- Department of Pediatric Pulmonology, Erasmus MC, Rotterdam, the Netherlands; Department of Cell Biology, Erasmus MC, Rotterdam, the Netherlands
| | - Hettie M Janssens
- Department of Pediatric Pulmonology, Erasmus MC, Rotterdam, the Netherlands
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
21
|
Leiva-Juárez MM, Kolls JK, Evans SE. Lung epithelial cells: therapeutically inducible effectors of antimicrobial defense. Mucosal Immunol 2018; 11:21-34. [PMID: 28812547 PMCID: PMC5738267 DOI: 10.1038/mi.2017.71] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 07/14/2017] [Indexed: 02/06/2023]
Abstract
Lung epithelial cells are increasingly recognized to be active effectors of microbial defense, contributing to both innate and adaptive immune function in the lower respiratory tract. As immune sentinels, lung epithelial cells detect diverse pathogens through an ample repertoire of membrane-bound, endosomal, and cytosolic pattern-recognition receptors (PRRs). The highly plastic epithelial barrier responds to detected threats via modulation of paracellular flux, intercellular communications, mucin production, and periciliary fluid composition. Epithelial PRR stimulation also induces production of cytokines that recruit and sculpt leukocyte-mediated responses, and promotes epithelial generation of antimicrobial effector molecules that are directly microbicidal. The epithelium can alternately enhance tolerance to pathogens, preventing tissue damage through PRR-induced inhibitory signals, opsonization of pathogen-associated molecular patterns, and attenuation of injurious leukocyte responses. The inducibility of these protective responses has prompted attempts to therapeutically harness epithelial defense mechanisms to protect against pneumonias. Recent reports describe successful strategies for manipulation of epithelial defenses to protect against a wide range of respiratory pathogens. The lung epithelium is capable of both significant antimicrobial responses that reduce pathogen burdens and tolerance mechanisms that attenuate immunopathology. This manuscript reviews inducible lung epithelial defense mechanisms that offer opportunities for therapeutic manipulation to protect vulnerable populations against pneumonia.
Collapse
Affiliation(s)
- Miguel M. Leiva-Juárez
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jay K. Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Scott E. Evans
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA,The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
22
|
Willemin ME, Lumen A. Thiocyanate: a review and evaluation of the kinetics and the modes of action for thyroid hormone perturbations. Crit Rev Toxicol 2017. [DOI: 10.1080/10408444.2017.1281590] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Marie-Emilie Willemin
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Annie Lumen
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
23
|
Moualek I, Iratni Aiche G, Mestar Guechaoui N, Lahcene S, Houali K. Antioxidant and anti-inflammatory activities of Arbutus unedo aqueous extract. Asian Pac J Trop Biomed 2016. [DOI: 10.1016/j.apjtb.2016.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
24
|
McElroy CS, Min E, Huang J, Loader JE, Hendry-Hofer TB, Garlick RB, Rioux JS, Veress LA, Smith R, Osborne C, Anderson DR, Holmes WW, Paradiso DC, White CW, Day BJ. From the Cover: Catalytic Antioxidant Rescue of Inhaled Sulfur Mustard Toxicity. Toxicol Sci 2016; 154:341-353. [PMID: 27605419 DOI: 10.1093/toxsci/kfw170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sulfur mustard (bis 2-chloroethyl ethyl sulfide, SM) is a powerful bi-functional vesicating chemical warfare agent. SM tissue injury is partially mediated by the overproduction of reactive oxygen species resulting in oxidative stress. We hypothesized that using a catalytic antioxidant (AEOL 10150) to alleviate oxidative stress and secondary inflammation following exposure to SM would attenuate the toxic effects of SM inhalation. Adult male rats were intubated and exposed to SM (1.4 mg/kg), a dose that produces an LD50 at approximately 24 h. Rats were randomized and treated via subcutaneous injection with either sterile PBS or AEOL 10150 (5 mg/kg, sc, every 4 h) beginning 1 h post-SM exposure. Rats were euthanized between 6 and 48 h after exposure to SM and survival and markers of injury were determined. Catalytic antioxidant treatment improved survival after SM inhalation in a dose-dependent manner, up to 52% over SM PBS at 48 h post-exposure. This improvement was sustained for at least 72 h after SM exposure when treatments were stopped after 48 h. Non-invasive monitoring throughout the duration of the studies also revealed blood oxygen saturations were improved by 10% and clinical scores were reduced by 57% after SM exposure in the catalytic antioxidant treatment group. Tissue analysis showed catalytic antioxidant therapy was able to decrease airway cast formation by 69% at 48 h post-exposure. To investigate antioxidant induced changes at the peak of injury, several biomarkers of oxidative stress and inflammation were evaluated at 24 h post-exposure. AEOL 10150 attenuated SM-mediated lung lipid oxidation, nitrosative stress and many proinflammatory cytokines. The findings indicate that catalytic antioxidants may be useful medical countermeasure against inhaled SM exposure.
Collapse
Affiliation(s)
- Cameron S McElroy
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045.,Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Elysia Min
- Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Jie Huang
- Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Joan E Loader
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | | | - Rhonda B Garlick
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Jackie S Rioux
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Livia A Veress
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Russell Smith
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Chris Osborne
- Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Dana R Anderson
- Analytical Toxicology Division, Proving Grounds United States Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen, Maryland 21010
| | - Wesley W Holmes
- Analytical Toxicology Division, Proving Grounds United States Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen, Maryland 21010
| | - Danielle C Paradiso
- Analytical Toxicology Division, Proving Grounds United States Army Medical Research Institute of Chemical Defense (USAMRICD), Aberdeen, Maryland 21010
| | - Carl W White
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045.,Department of Pediatrics, University of Colorado, Aurora, Colorado 80045
| | - Brian J Day
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado 80045 .,Department of Medicine, National Jewish Health, Denver, Colorado 80206
| |
Collapse
|
25
|
Wang Z, DiDonato JA, Buffa J, Comhair SA, Aronica MA, Dweik RA, Lee NA, Lee JJ, Thomassen MJ, Kavuru M, Erzurum SC, Hazen SL. Eosinophil Peroxidase Catalyzed Protein Carbamylation Participates in Asthma. J Biol Chem 2016; 291:22118-22135. [PMID: 27587397 DOI: 10.1074/jbc.m116.750034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Indexed: 12/21/2022] Open
Abstract
The biochemical mechanisms through which eosinophils contribute to asthma pathogenesis are unclear. Here we show eosinophil peroxidase (EPO), an abundant granule protein released by activated eosinophils, contributes to characteristic asthma-related phenotypes through oxidative posttranslational modification (PTM) of proteins in asthmatic airways through a process called carbamylation. Using a combination of studies we now show EPO uses plasma levels of the pseudohalide thiocyanate (SCN-) as substrate to catalyze protein carbamylation, as monitored by PTM of protein lysine residues into Nϵ-carbamyllysine (homocitrulline), and contributes to the pathophysiological sequelae of eosinophil activation. Studies using EPO-deficient mice confirm EPO serves as a major enzymatic source for protein carbamylation during eosinophilic inflammatory models, including aeroallergen challenge. Clinical studies similarly revealed significant enrichment in carbamylation of airway proteins recovered from atopic asthmatics versus healthy controls in response to segmental allergen challenge. Protein-bound homocitrulline is shown to be co-localized with EPO within human asthmatic airways. Moreover, pathophysiologically relevant levels of carbamylated protein either incubated with cultured human airway epithelial cells in vitro, or provided as an aerosolized exposure in non-sensitized mice, induced multiple asthma-associated phenotypes including induction of mucin, Th2 cytokines, IFNγ, TGFβ, and epithelial cell apoptosis. Studies with scavenger receptor-A1 null mice reveal reduced IL-13 generation following exposure to aerosolized carbamylated protein, but no changes in other asthma-related phenotypes. In summary, EPO-mediated protein carbamylation is promoted during allergen-induced asthma exacerbation, and can both modulate immune responses and trigger a cascade of many of the inflammatory signals present in asthma.
Collapse
Affiliation(s)
- Zeneng Wang
- From the Departments of Cellular and Molecular Medicine
| | | | | | | | | | | | - Nancy A Lee
- the Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona 85259
| | - James J Lee
- the Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, Arizona 85259
| | - Mary Jane Thomassen
- the Division of Pulmonary, Critical Care & Sleep Medicine, East Carolina University, Greenville, North Carolina 27834, and
| | - Mani Kavuru
- the Division of Pulmonary and Critical Care Medicine, Thomas Jefferson University and Hospital, Philadelphia, Pennsylvania 19107
| | | | - Stanley L Hazen
- From the Departments of Cellular and Molecular Medicine, Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio 44195,
| |
Collapse
|
26
|
New insights into thiocyanate oxidation by human myeloperoxidase. J Inorg Biochem 2016; 162:117-126. [PMID: 27343172 DOI: 10.1016/j.jinorgbio.2016.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/26/2016] [Accepted: 06/14/2016] [Indexed: 11/20/2022]
Abstract
Human myeloperoxidase (MPO) uses chloride and thiocyanate as physiological substrates at neutral pH. Oxidation of thiocyanate to hypothiocyanite mediated by the redox intermediate Compound I rapidly restores the ferric state of MPO. At low thiocyanate concentration and in the presence of hydrogen peroxide the observed reaction sequence is Compound I→ferric MPO→Compound II→MPO-cyanide complex, whereas at high thiocyanate concentrations and in the absence of H2O2 the only observed transition is Compound I→ferric MPO. The reaction of ferric MPO with hypothiocyanite directly forms the MPO-cyanide complex, whereas a transient product derived from the reaction between hypothiocyanite and hydrogen peroxide is demonstrated to mediate the conversion of ferric MPO to Compound II. Mechanisms for those reactions are discussed and proposed.
Collapse
|
27
|
Therapeutic Effects of α1-Antitrypsin on Psedumonas aeruginosa Infection in ENaC Transgenic Mice. PLoS One 2015; 10:e0141232. [PMID: 26509529 PMCID: PMC4624966 DOI: 10.1371/journal.pone.0141232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease with many airway pathological features, including aberrant epithelial sodium channel (ENaC) function, persistent Pseudomonas aeruginosa (PA) infection and neutrophil-dominant inflammation. PA infection in CF airways is difficult to treat due to antibiotic resistance and other factors. Recently, α1-antitrypsin (A1AT) have been shown to be effective to reduce CF airway PA infection. However, there is a dearth of studies about the mechanisms underlying A1AT's therapeutic effects. The goal of our study is to provide an animal model of A1AT therapy in CF lungs. ENaC transgenic mice with PA infection were used as a CF-like model. Mice were intratracheally treated with PA or saline (control) in a fibrin plug. Two hours after PA infection, aerosolized A1AT were delivered to mouse lungs once daily. At day 1 and day 3 post PA infection, lung inflammation, PA load as well as host defence protein short palate, lung, and nasal epithelium clone 1 (SPLUNC1) were measured. At day 1 post PA infection when A1AT was delivered once to ENaC transgenic mouse lungs, A1AT did not reduce lung inflammation (e.g., neutrophils) and PA load. However, at day 3 post PA infection when ENaC transgenic mice received three repeated A1AT treatments, a significant decrease in airspace inflammation and PA load was observed. Although A1AT prevented the loss of SPLUNC1 in bronchoalveolar lavage fluid of PA-infected wild-type mice, it did not restore SPLUNC1 levels in ENaC transgenic mice. Our current study has provided a valid and quick A1AT therapeutic model in CF-like lungs that may serve as a platform for future mechanistic studies about how A1AT exerts beneficial effects in human CF patients.
Collapse
|
28
|
Chandler JD, Min E, Huang J, McElroy CS, Dickerhof N, Mocatta T, Fletcher AA, Evans CM, Liang L, Patel M, Kettle AJ, Nichols DP, Day BJ. Antiinflammatory and Antimicrobial Effects of Thiocyanate in a Cystic Fibrosis Mouse Model. Am J Respir Cell Mol Biol 2015; 53:193-205. [PMID: 25490247 DOI: 10.1165/rcmb.2014-0208oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Thiocyanate (SCN) is used by the innate immune system, but less is known about its impact on inflammation and oxidative stress. Granulocytes oxidize SCN to evolve the bactericidal hypothiocyanous acid, which we previously demonstrated is metabolized by mammalian, but not bacterial, thioredoxin reductase (TrxR). There is also evidence that SCN is dysregulated in cystic fibrosis (CF), a disease marked by chronic infection and airway inflammation. To investigate antiinflammatory effects of SCN, we administered nebulized SCN or saline to β epithelial sodium channel (βENaC) mice, a phenotypic CF model. SCN significantly decreased airway neutrophil infiltrate and restored the redox ratio of glutathione in lung tissue and airway epithelial lining fluid to levels comparable to wild type. Furthermore, in Pseudomonas aeruginosa-infected βENaC and wild-type mice, SCN decreased inflammation, proinflammatory cytokines, and bacterial load. SCN also decreased airway neutrophil chemokine keratinocyte chemoattractant (also known as C-X-C motif chemokine ligand 1) and glutathione sulfonamide, a biomarker of granulocyte oxidative activity, in uninfected βENaC mice. Lung tissue TrxR activity and expression increased in inflamed lung tissue, providing in vivo evidence for the link between hypothiocyanous acid metabolism by TrxR and the promotion of selective biocide of pathogens. SCN treatment both suppressed inflammation and improved host defense, suggesting that nebulized SCN may have important therapeutic utility in diseases of both chronic airway inflammation and persistent bacterial infection, such as CF.
Collapse
Affiliation(s)
- Joshua D Chandler
- 1 Department of Pharmaceutical Sciences and.,Departments of 2 Medicine and
| | | | | | - Cameron S McElroy
- 1 Department of Pharmaceutical Sciences and.,Departments of 2 Medicine and
| | - Nina Dickerhof
- 3 Centre for Free Radical Research, Department of Pathology, University of Otago Christchurch, Christchurch, New Zealand
| | - Tessa Mocatta
- 3 Centre for Free Radical Research, Department of Pathology, University of Otago Christchurch, Christchurch, New Zealand
| | - Ashley A Fletcher
- 4 Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado
| | - Christopher M Evans
- 4 Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, Colorado
| | | | | | - Anthony J Kettle
- 3 Centre for Free Radical Research, Department of Pathology, University of Otago Christchurch, Christchurch, New Zealand
| | - David P Nichols
- Departments of 2 Medicine and.,5 Pediatrics, National Jewish Health, Denver, Colorado; and
| | - Brian J Day
- 1 Department of Pharmaceutical Sciences and.,Departments of 2 Medicine and
| |
Collapse
|
29
|
Nichols DP, Chmiel JF. Inflammation and its genesis in cystic fibrosis. Pediatr Pulmonol 2015; 50 Suppl 40:S39-56. [PMID: 26335954 DOI: 10.1002/ppul.23242] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/07/2015] [Accepted: 06/16/2015] [Indexed: 12/17/2022]
Abstract
The host inflammatory response in cystic fibrosis (CF) lung disease has long been recognized as a central pathological feature and an important therapeutic target. Indeed, many believe that bronchiectasis results largely from the oxidative and proteolytic damage comprised within an exuberant airway inflammatory response that is dominated by neutrophils. In this review, we address the longstanding argument of whether or not the inflammatory response is directly attributable to impairment of the cystic fibrosis transmembrane conductance regulator or only secondary to airway obstruction and chronic bacterial infection and challenge the importance of this distinction in the context of therapy. We also review the centrality of neutrophils in CF lung pathophysiology and highlight more recent data that suggest the importance of other cell types and signaling beyond NF-κB activation. We discuss how protease and redox imbalance are critical factors in CF airway inflammation and end by reviewing some of the more promising therapeutic approaches now under development.
Collapse
Affiliation(s)
- David P Nichols
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.,National Jewish Health, Denver, Colorado
| | - James F Chmiel
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| |
Collapse
|
30
|
Abstract
Cystic fibrosis (CF) is a heterogeneous multiorgan disease caused by mutations in the CFTR gene leading to misfolding (and other defects) and consequent dysfunction of CFTR protein. The majority of mutations cause a severe CF phenotype, and people with this condition will require a wide variety of medical interventions and therapies throughout their lives to address the symptoms of their condition. CF affects many different organ systems, but the most serious consequence of the disease is degeneration of lung function due to chronic respiratory infection and colonization of the airways with opportunistic microbial pathogens. Improvements in therapeutics, particularly the effective use of antibiotics, have led to significant gradual increases in life expectancy. There remains, however, a continuing need for newer, safer and more effective antimicrobials and mucolytic agents to maintain and improve our ability to combat CF lung infections before other curative approaches which target the root cause of the disease become available.
Collapse
|
31
|
Morgan PE, Laura RP, Maki RA, Reynolds WF, Davies MJ. Thiocyanate supplementation decreases atherosclerotic plaque in mice expressing human myeloperoxidase. Free Radic Res 2015; 49:743-9. [PMID: 25812586 DOI: 10.3109/10715762.2015.1019347] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Elevated levels of the heme enzyme myeloperoxidase (MPO) are associated with adverse cardiovascular outcomes. MPO predominantly catalyzes formation of the oxidants hypochlorous acid (HOCl) from Cl(-), and hypothiocyanous acid (HOSCN) from SCN(-), with these anions acting as competitive substrates. HOSCN is a less powerful and more specific oxidant than HOCl, and selectively targets thiols; such damage is largely reversible, unlike much HOCl-induced damage. We hypothesized that increased plasma SCN(-), and hence HOSCN formation instead of HOCl, may decrease artery wall damage. This was examined using high-fat fed atherosclerosis-prone LDLR(-/-) mice transgenic for human MPO, with and without SCN(-) (10 mM) added to drinking water. Serum samples, collected fortnightly, were analyzed for cholesterol, triglycerides, thiols, MPO, and SCN(-); study-long exposure was calculated by area under the curve (AUC). Mean serum SCN(-) concentrations were elevated in the supplemented mice (200-320 μM) relative to controls (< 120 μM). Normalized aortic root plaque areas at sacrifice were 26% lower in the SCN(-)-supplemented mice compared with controls (P = 0.0417), but plaque morphology was not appreciably altered. Serum MPO levels steadily increased in mice on the high-fat diet, however, comparison of SCN(-)-supplemented versus control mice showed no significant changes in MPO protein, cholesterol, or triglyceride levels; thiol levels were decreased in supplemented mice at one time-point. Plaque areas increased with higher cholesterol AUC (r = 0.4742; P = 0.0468), and decreased with increasing SCN(-) AUC (r = - 0.5693; P = 0.0134). These data suggest that increased serum SCN(-) levels, which can be achieved in humans by dietary manipulation, may decrease atherosclerosis burden.
Collapse
Affiliation(s)
- P E Morgan
- The Heart Research Institute , Newtown, NSW , Australia
| | | | | | | | | |
Collapse
|
32
|
Caverly LJ, Caceres SM, Fratelli C, Happoldt C, Kidwell KM, Malcolm KC, Nick JA, Nichols DP. Mycobacterium abscessus morphotype comparison in a murine model. PLoS One 2015; 10:e0117657. [PMID: 25675351 PMCID: PMC4326282 DOI: 10.1371/journal.pone.0117657] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 12/30/2014] [Indexed: 11/18/2022] Open
Abstract
Pulmonary infections with Mycobacterium abscessus (M. abscessus) are increasingly prevalent in patients with lung diseases such as cystic fibrosis. M. abscessus exists in two morphotypes, smooth and rough, but the impact of morphotype on virulence is unclear. We developed an immune competent mouse model of pulmonary M. abscessus infection and tested the differences in host inflammatory response between the morphotypes of M. abscessus. Smooth and rough morphotypes of M. abscessus were isolated from the same American Type Culture Collection strain. Wild type and cystic fibrosis mice were intratracheally inoculated with known quantities of M. abscessus suspended in fibrin plugs. At the time of sacrifice lung and splenic tissues and bronchoalveolar lavage fluid were collected and cultured. Bronchoalveolar lavage fluid was analyzed for leukocyte count, differential and cytokine expression. Pulmonary infection with M. abscessus was present at both 3 days and 14 days post-inoculation in all groups at greater levels than systemic infection. Inoculation with M. abscessus rough morphotype resulted in more bronchoalveolar lavage fluid neutrophils compared to smooth morphotype at 14 days post-inoculation in both wild type (p = 0.01) and cystic fibrosis (p<0.01) mice. Spontaneous in vivo conversion from smooth to rough morphotype occurred in 12/57 (21%) of mice. These mice trended towards greater weight loss than mice in which morphotype conversion did not occur. In the described fibrin plug model of M. abscessus infection, pulmonary infection with minimal systemic dissemination is achieved with both smooth and rough morphotypes. In this model M. abscessus rough morphotype causes a greater host inflammatory response than the smooth based on bronchoalveolar lavage fluid neutrophil levels.
Collapse
Affiliation(s)
- Lindsay J. Caverly
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Silvia M. Caceres
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Cori Fratelli
- Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
| | - Carrie Happoldt
- Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
| | - Kelley M. Kidwell
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kenneth C. Malcolm
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Jerry A. Nick
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, United States of America
| | - David P. Nichols
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
- Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, United States of America
| |
Collapse
|
33
|
Chandler JD, Day BJ. Biochemical mechanisms and therapeutic potential of pseudohalide thiocyanate in human health. Free Radic Res 2015; 49:695-710. [PMID: 25564094 DOI: 10.3109/10715762.2014.1003372] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Thiocyanate (SCN(-)) is a ubiquitous molecule in mammalian biology, reaching up to mM concentrations in extracellular fluids. Two- electron oxidation of SCN(-) by H2O2 produces hypothiocyanous acid (HOSCN), a potent anti-microbial species. This reaction is catalyzed by chordate peroxidases (e.g., myeloperoxidase and lactoperoxidase), occurring in human secretory mucosa, including the oral cavity, airway, and alimentary tract, and regulates resident and transient flora as part of innate immunity. Increasing SCN(-) levels limits the concentrations of a family of 2-electron oxidants (H2O2, hypohalous acids, and haloamines) in favor of HOSCN formation, altering the oxidative impact on host tissue by substitution of repairable thiol and selenol oxidations instead of biomolecule degradation. This fine-tuning of inflammatory oxidation paradoxically associates with maintained host defense and decreased host injury during infections, due in part to phylogenetic differences in the thioredoxin reductase system between mammals and their pathogens. These differences could be exploited by pharmacologic use of SCN(-). Recent preclinical studies have identified anti-microbial and anti-inflammatory effects of SCN(-) in pulmonary and cardiovascular animal models, with implications for treatment of infectious lung disease and atherogenesis. Further research is merited to expand on these findings and identify other diseases where SCN(-) may be of use. High oral bioavailability and an increased knowledge of the biochemical effects of SCN(-) on a subset of pro-inflammatory reactions suggest clinical utility.
Collapse
|
34
|
Wong H, Zou S, Li J, Ma C, Chen J, Leong P, Leung H, Chan W, Ko K. <i>Lily bulb</i> Nectar Produces Expectorant and Anti-Tussive Activities, and Suppresses Cigarette Smoke-Induced Inflammatory Response in the Respiratory Tract in Mice. Chin Med 2015. [DOI: 10.4236/cm.2015.62015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
35
|
Rees MD, Maiocchi SL, Kettle AJ, Thomas SR. Mechanism and regulation of peroxidase-catalyzed nitric oxide consumption in physiological fluids: critical protective actions of ascorbate and thiocyanate. Free Radic Biol Med 2014; 72:91-103. [PMID: 24704973 DOI: 10.1016/j.freeradbiomed.2014.03.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 03/26/2014] [Accepted: 03/27/2014] [Indexed: 01/01/2023]
Abstract
Catalytic consumption of nitric oxide (NO) by myeloperoxidase and related peroxidases is implicated as playing a key role in impairing NO bioavailability during inflammatory conditions. However, there are major gaps in our understanding of how peroxidases consume NO in physiological fluids, in which multiple reactive enzyme substrates and antioxidants are present. Notably, ascorbate has been proposed to enhance myeloperoxidase-catalyzed NO consumption by forming NO-consuming substrate radicals. However, we show that in complex biological fluids ascorbate instead plays a critical role in inhibiting NO consumption by myeloperoxidase and related peroxidases (lactoperoxidase, horseradish peroxidase) by acting as a competitive substrate for protein-bound redox intermediates and by efficiently scavenging peroxidase-derived radicals (e.g., urate radicals), yielding ascorbyl radicals that fail to consume NO. These data identify a novel mechanistic basis for how ascorbate preserves NO bioavailability during inflammation. We show that NO consumption by myeloperoxidase Compound I is significant in substrate-rich fluids and is resistant to competitive inhibition by ascorbate. However, thiocyanate effectively inhibits this process and yields hypothiocyanite at the expense of NO consumption. Hypothiocyanite can in turn form NO-consuming radicals, but thiols (albumin, glutathione) readily prevent this. Conversely, where ascorbate is absent, glutathione enhances NO consumption by urate radicals via pathways that yield S-nitrosoglutathione. Theoretical kinetic analyses provide detailed insights into the mechanisms by which ascorbate and thiocyanate exert their protective actions. We conclude that the local depletion of ascorbate and thiocyanate in inflammatory microenvironments (e.g., due to increased metabolism or dysregulated transport) will impair NO bioavailability by exacerbating peroxidase-catalyzed NO consumption.
Collapse
Affiliation(s)
- Martin D Rees
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; Rural Clinical School, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Sophie L Maiocchi
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Anthony J Kettle
- Centre for Free Radical Research, Department of Pathology, University of Otago, 8140 Christchurch, New Zealand
| | - Shane R Thomas
- Centre for Vascular Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
36
|
Abstract
Cystic fibrosis (CF) is the most common inherited genetic condition amongst Caucasian ethnicities, affecting 1 in 2500 live births. There remains a significant unmet medical need for more and better therapies for this chronic, degenerative condition, in particular those that address the respiratory dysfunction and respiratory infections that characterise CF. CF is caused by mutations in the cystic transmembrane conductance regulator gene (CFTR). The key pathology driver of CF is dysregulated ion transport across the epithelial cell barriers that line the respiratory tract, gastrointestinal tract and other organ systems. This review focuses on the state-of-the-art advances and future directions in therapeutic strategies to combat and manage the symptoms of CF and/or restore functionality of the defective CFTR.
Collapse
|
37
|
Ziady AG, Hansen J. Redox balance in cystic fibrosis. Int J Biochem Cell Biol 2014; 52:113-23. [PMID: 24657650 DOI: 10.1016/j.biocel.2014.03.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/28/2014] [Accepted: 03/11/2014] [Indexed: 11/30/2022]
Abstract
The homeostatic balance between oxidants and antioxidants in biological systems is known as redox balance, and is regulated by complex processes. Redox balance regulates many of the known cellular pathways and disease processes. The dysregulation of redox balance can lead to acute or long-term oxidative or reductive stresses that are associated with many of the abnormalities observed in cystic fibrosis (CF). Over the past 5 decades researchers have examined contributors to redox dysregulation, their molecular products, and their impact on ion transport, cell proliferation, inflammation, bacterial killing, and the metabolism of nucleic acids, proteins, and lipids in CF. CF patients exhibit elevated markers of oxidative stress when compared to non-CF healthy controls; however, whether the reported redox imbalance is sufficient to produce pathology has been controversial. In addition, comparisons between CF and non-CF disease controls have been lacking. To better understand the mechanisms which mediate the generation of oxidants and antioxidants in CF and the importance of their balance in effecting oxidative or reductive stress, we will review the determinants of redox balance in the blood, lumen, and cellular compartments. From the perspective of methodological application, we will focus on the approaches most often used to study oxidant and antioxidants in CF, including biochemical, proteomic, metabolomic, and lipidomic studies, with a discussion of the few transcriptomic analyses that predict changes in the expression of regulators of redox. Finally, we will discuss the utility of oxidants and antioxidants as biomarkers of disease and the use of antioxidant therapy in CF.
Collapse
Affiliation(s)
- Assem G Ziady
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - Jason Hansen
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|