1
|
Fotouh A, Abdel-Maguid DS, Abdelhaseib M, Zaki RS, Darweish M. Pathological and pharmacovigilance monitoring as toxicological imputations of azithromycin and its residues in broilers. Vet World 2024; 17:1271-1280. [PMID: 39077436 PMCID: PMC11283599 DOI: 10.14202/vetworld.2024.1271-1280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/13/2024] [Indexed: 07/31/2024] Open
Abstract
Background and Aim The importance of monitoring antimicrobial residues in food is underlined by increasing worries about food safety and public health. The potential toxicity of azithromycin (Az) on broilers and its impact on chicken meat residues require further investigation. This study assesses Az's toxicity effects and associated risks in broiler chickens through evaluation. Materials and Methods One hundred and twenty chicks were distributed into four equal groups randomly. Each group received different daily oral doses of Az: 200 mg/kg for Az1, 100 mg/kg for Az2, and 50 mg/kg for Az3. The FAz group was given plain water. High-performance liquid chromatography was used to measure Az residue levels in muscle and liver. Oxidative markers (malondialdehyde [MDA], superoxide dismutase [SOD], catalase [CAT]), liver and kidney function tests, and histopathological examination were conducted. Results The levels of alanine aminotransferase and aspartate aminotransferase increased in Az1 and Az2 groups from 8 h to 3 days and decreased slightly in Az2 by 7 days, while they remained normal in Az3. The levels of uric acid and creatine in the Az1 and Az2 groups increased from 8 h to 3 days and subsequently decreased in Az2 by the 7th day. Az1 group showed the highest increase in MDA levels within 7 days. With higher Az doses, SOD and CAT levels showed a more significant decrease post-treatment. 9.1 μg/kg Az1 liver had the highest residues, whereas none were detected in muscle. Conclusion At higher doses, Az caused significant liver and kidney damage, whereas lower doses had negligible effects. Muscle tissue contains fewer Az residues than liver. Assessing risks and ensuring compliance with regulations necessitate constant surveillance of Az residues in food. The health implications and risk management insights necessitate further investigation into the long-term effects of Az residues.
Collapse
Affiliation(s)
- Ahmed Fotouh
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, New Valley University, El Kharga, Egypt
- MBA, Marywood University, Pennsylvania, USA
| | - Doaa Safwat Abdel-Maguid
- Department of Forensic and Toxicology, Faculty of Veterinary Medicine, New Valley University, El Kharga, Egypt
| | - Maha Abdelhaseib
- Department of Food Hygiene, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Rania Samir Zaki
- Department of Food Hygiene, Safety and Technology, Faculty of Veterinary Medicine, New Valley University, El Kharga, Egypt
| | - Marwa Darweish
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, 13736, Moshtohor, Toukh, Qaluiobia, Egypt
| |
Collapse
|
2
|
Lauriola M, Pani A, Ippoliti G, Mortara A, Milighetti S, Mazen M, Perseghin G, Pastori D, Grosso P, Scaglione F. Effect of Combination Therapy of Hydroxychloroquine and Azithromycin on Mortality in Patients With COVID-19. Clin Transl Sci 2020; 13:1071-1076. [PMID: 32926573 PMCID: PMC7719367 DOI: 10.1111/cts.12860] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/13/2020] [Indexed: 01/10/2023] Open
Abstract
Conflicting evidence regarding the use of hydroxychloroquine (HCQ) and azithromycin for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection do exist. We performed a retrospective single-center cohort study including 377 consecutive patients admitted for pneumonia related to coronavirus disease 2019 (COVID-19). Of these, 297 were in combination treatment, 17 were on HCQ alone, and 63 did not receive either of these 2 drugs because of contraindications. The primary end point was in-hospital death. Mean age was 71.8 ± 13.4 years and 34.2% were women. We recorded 146 deaths: 35 in no treatment, 7 in HCQ treatment group, and 102 in HCQ + azithromycin treatment group (log rank test for Kaplan-Meier curve P < 0.001). At multivariable Cox proportional hazard regression analysis, age (hazard ratio (HR) 1.057, 95% confidence interval (CI) 1.035-1.079, P < 0.001), mechanical ventilation/continuous positive airway pressure (HR 2.726, 95% CI 1.823-4.074, P < 0.001), and C reactive protein above the median (HR 2.191, 95% CI 1.479-3.246, P < 0.001) were directly associated with death, whereas use of HCQ + azithromycin (vs. no treatment; HR 0.265, 95% CI 0.171-0.412, P < 0.001) was inversely associated. In this study, we found a reduced in-hospital mortality in patients treated with a combination of HCQ and azithromycin after adjustment for comorbidities. A large randomized trial is necessary to confirm these findings.
Collapse
Affiliation(s)
| | - Arianna Pani
- Department of Oncology and Hemato‐OncologyUniversità degli Studi di MilanoMilanItaly
| | | | | | | | - Marjieh Mazen
- Internal Medicine DepartmentPoliclinico di MonzaMonzaItaly
| | - Gianluca Perseghin
- Internal Medicine DepartmentPoliclinico di MonzaMonzaItaly
- Department of Medicine and SurgeryUniversità degli Studi di Milano BicoccaMilanItaly
| | - Daniele Pastori
- Department of Clinical, Internal, Anesthesiologic, and Cardiovascular SciencesSapienza University of RomeRomeItaly
| | - Paolo Grosso
- Intensive Care UnitPoliclinico di MonzaMonzaItaly
| | - Francesco Scaglione
- Department of Oncology and Hemato‐OncologyUniversità degli Studi di MilanoMilanItaly
| |
Collapse
|
3
|
Sadeghi S, Tapak M, Ghazanfari T, Mosaffa N. A review of Sulfur Mustard-induced pulmonary immunopathology: An Alveolar Macrophage Approach. Toxicol Lett 2020; 333:115-129. [PMID: 32758513 DOI: 10.1016/j.toxlet.2020.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/27/2022]
Abstract
Despite many studies investigating the mechanism of Sulfur Mustard (SM) induced lung injury, the underlying mechanism is still unclear. Inflammatory and subsequent fibroproliferative stages of SM-toxicity are based upon several highly-related series of events controlled by the immune system. The inhalation of SM gas variably affects different cell populations within the lungs. Various studies have shown the critical role of macrophages in triggering a pulmonary inflammatory response as well as its maintenance, resolution, and repair. Importantly, macrophages can serve as either pro-inflammatory or anti-inflammatory populations depending on the present conditions at any pathological stage. Different characteristics of macrophages, including their differentiation, phenotypic, and functional properties, as well as interactions with other cell populations determine the outcomes of lung diseases and the extent of long- or short-term pulmonary damage induced by SM. In this paper, we summarize the current state of knowledge regarding the role of alveolar macrophages and their mediators in the pathogenesis of SM in pulmonary injury. Investigating the specific cells and mechanisms involved in SM-lung injury may be useful in finding new target opportunities for treatment of this injury.
Collapse
Affiliation(s)
- Somaye Sadeghi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahtab Tapak
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of Immunology, Shahed University, Tehran, Iran.
| | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Etemad L, Moshiri M, Balali-Mood M. Delayed Complications and Long-Term Management of Sulfur Mustard Poisoning: A Narrative Review of Recent Advances by Iranian Researchers Part ІІ: Clinical Management and Therapy. IRANIAN JOURNAL OF MEDICAL SCIENCES 2018; 43:235-247. [PMID: 29892141 PMCID: PMC5993908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The present study aimed to review and discuss the recommended and recently suggested protocols by Iranian researchers for a long-term treatment of delayed complications of sulfur mustard (DCSM) in veterans. As indicated clinically, patients who suffer from delayed ocular complications of sulfur mustard (DOCS) benefit from treatments for dry eyes, therapeutic contact lenses, amniotic membrane transplantation; blepharorrhaphy, tarsorrhaphy, limbal stem cell transplantation; corneal transplantation, topical steroids, and immunosuppressive. In spite of penetrating keratoplasty, lamellar keratoplasty and keratolimbal allograft had a good long-term survival. Delayed respiratory complications (DRCS) are the most common effects and life-threatening in Iranian veterans. The recommended treatment protocols include regular clinical evaluations, respiratory physiotherapy and rehabilitation, N-acetyl cysteine; warm humidified air, long-acting b2-agonists, and inhaled corticosteroids. Azithromycin has also been effective in improving clinical conditions, pulmonary function tests, inflammatory indexes, and life quality of the veterans. Interferon gamma (IFN-γ) and helium: oxygen combination were also used in severe DRCS with good results. Some of the delayed cutaneous complications (DCCS) such as itching affects the quality of life of victims. Regular but not frequent showering and bathing, applying sunscreen compounds, topical corticosteroids, and systemic antihistamines reduce the problems of DCCS patients. Several compounds such as capsaicin cream, pimecrolimus, IFN-γ, phenol-menthol; Aloe vera/olive oil cream, cetirizine, doxepine, and hydroxyzine were evaluated in DCCS patients with some benefits. The physicians in charge of veterans emphasize the importance of a healthy lifestyle, appropriate financial/social/cultural supports, and a degree of reassurance and supportive care on the clinical improvement of patients.
Collapse
Affiliation(s)
- Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Moshiri
- Medical Toxicology Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdi Balali-Mood
- Medical Toxicology Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Pérez-Del Palacio J, Díaz C, Vergara N, Algieri F, Rodríguez-Nogales A, de Pedro N, Rodríguez-Cabezas ME, Genilloud O, Gálvez J, Vicente F. Exploring the Role of CYP3A4 Mediated Drug Metabolism in the Pharmacological Modulation of Nitric Oxide Production. Front Pharmacol 2017; 8:202. [PMID: 28446877 PMCID: PMC5388737 DOI: 10.3389/fphar.2017.00202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/28/2017] [Indexed: 11/15/2022] Open
Abstract
Nitric-oxide synthase, the enzyme responsible for mammalian nitric oxide generation, and cytochrome P450, the major enzymes involved in drug metabolism, share striking similarities. Therefore, it makes sense that cytochrome P450 drug mediated biotransformations might play an important role in the pharmacological modulation of nitric oxide synthase. In this work, we have undertaken an integrated in vitro assessment of the hepatic metabolism and nitric oxide modulation of previously described dual inhibitors (imidazoles and macrolides) of these enzymes in order assess the implication of CYP450 activities over production of nitric oxide. In vitro systems based in human liver microsomes and activated mouse macrophages were developed for these purposes. Additionally in vitro production the hepatic metabolites of dual inhibitor, roxithromycin, was investigated achieving the identification and isolation of main hepatic biotransformation products. Our results suggested that for some macrolide compounds, the cytochrome P450 3A4 derived drug metabolites have an important effect on nitric oxide production and might critically contribute to the pharmacological immunomodulatory activity observed.
Collapse
Affiliation(s)
- José Pérez-Del Palacio
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores de AndalucíaGranada, Spain
| | - Caridad Díaz
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores de AndalucíaGranada, Spain
| | - Noemí Vergara
- Calcium Metabolism and Vascular Calcification Unit, Maimonides Institute for Biomedical Research, University Hospital Reina Sofia, Nephrology Service, University of CórdobaCordoba, Spain
| | - Francesca Algieri
- Department of Pharmacology, ibs, CIBER-EHD, Center for Biomedical Research, University of GranadaGranada, Spain
| | - Alba Rodríguez-Nogales
- Department of Pharmacology, ibs, CIBER-EHD, Center for Biomedical Research, University of GranadaGranada, Spain
| | - Nuria de Pedro
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores de AndalucíaGranada, Spain
| | - M Elena Rodríguez-Cabezas
- Department of Pharmacology, ibs, CIBER-EHD, Center for Biomedical Research, University of GranadaGranada, Spain
| | - Olga Genilloud
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores de AndalucíaGranada, Spain
| | - Julio Gálvez
- Department of Pharmacology, ibs, CIBER-EHD, Center for Biomedical Research, University of GranadaGranada, Spain
| | - Francisca Vicente
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores de AndalucíaGranada, Spain
| |
Collapse
|
6
|
Weinberger B, Malaviya R, Sunil VR, Venosa A, Heck DE, Laskin JD, Laskin DL. Mustard vesicant-induced lung injury: Advances in therapy. Toxicol Appl Pharmacol 2016; 305:1-11. [PMID: 27212445 PMCID: PMC5119915 DOI: 10.1016/j.taap.2016.05.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/18/2016] [Indexed: 01/17/2023]
Abstract
Most mortality and morbidity following exposure to vesicants such as sulfur mustard is due to pulmonary toxicity. Acute injury is characterized by epithelial detachment and necrosis in the pharynx, trachea and bronchioles, while long-term consequences include fibrosis and, in some instances, cancer. Current therapies to treat mustard poisoning are primarily palliative and do not target underlying pathophysiologic mechanisms. New knowledge about vesicant-induced pulmonary disease pathogenesis has led to the identification of potentially efficacious strategies to reduce injury by targeting inflammatory cells and mediators including reactive oxygen and nitrogen species, proteases and proinflammatory/cytotoxic cytokines. Therapeutics under investigation include corticosteroids, N-acetyl cysteine, which has both mucolytic and antioxidant properties, inducible nitric oxide synthase inhibitors, liposomes containing superoxide dismutase, catalase, and/or tocopherols, protease inhibitors, and cytokine antagonists such as anti-tumor necrosis factor (TNF)-α antibody and pentoxifylline. Antifibrotic and fibrinolytic treatments may also prove beneficial in ameliorating airway obstruction and lung remodeling. More speculative approaches include inhibitors of transient receptor potential channels, which regulate pulmonary epithelial cell membrane permeability, non-coding RNAs and mesenchymal stem cells. As mustards represent high priority chemical threat agents, identification of effective therapeutics for mitigating toxicity is highly significant.
Collapse
Affiliation(s)
- Barry Weinberger
- Division of Neonatal and Perinatal Medicine, Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, NY 11040, USA.
| | - Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Vasanthi R Sunil
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Diane E Heck
- Department of Environmental Health Science, New York Medical College, School of Public Health, Valhalla, NY 10595, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
7
|
Nourani MR, Mahmoodzadeh Hosseini H, Azimzadeh Jamalkandi S, Imani Fooladi AA. Cellular and molecular mechanisms of acute exposure to sulfur mustard: a systematic review. J Recept Signal Transduct Res 2016; 37:200-216. [DOI: 10.1080/10799893.2016.1212374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Mohammad Reza Nourani
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Khazdair MR, Boskabady MH, Ghorani V. Respiratory effects of sulfur mustard exposure, similarities and differences with asthma and COPD. Inhal Toxicol 2015; 27:731-44. [PMID: 26635274 DOI: 10.3109/08958378.2015.1114056] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Previous research has found relationships between sulfur mustard (SM) toxicity and its adverse effects. OBJECTIVE SM is highly toxic to the respiratory system, leading to hacking cough, rhinorrheachest tightness, acute pharyngitis and laryngitis, chronic bronchitis and lung fibrosis. In this review, based on the scientific literature, we provide an updated summary of information on SM exposures and their differences with asthma and COPD. METHOD Information of this review was obtained by searching Medline/PubMed, ScienceDirect, Scopus, Google Scholar, ISI Web of Knowledge and Chemical Abstracts. RESULTS SM exposure can decrease pulmonary function tests (PFTs) values. In addition, inflammatory cell accumulation in the respiratory tract and increased expression of some pro-inflammatory cytokines including tumor necrosis factor-α (TNFα), IL-1a, IL-1β, and reactive oxygen radicals due to SM exposure have been shown. Matrix metalloproteinase (MMP) which degrade extracellular matrix proteins, contributing to inflammatory cell recruitment, tissue injury and fibrosis are also up-regulated in the lung after SM exposure. In the lung, SM exposure also can cause serious pathological changes including airway inflammation, parenchymal tissue destruction and airway obstruction which can lead to asthma or chronic obstructive pulmonary disease (COPD). Following SM poisoning, DNA damage, apoptosis and autophagy are observed in the lung along with the increased expression of activated caspases and DNA repair enzymes. CONCLUSION In the present article, respiratory symptoms, changes in PFTs, lung pathology and lung inflammation due to SM exposure and the similarities and differences between them and those observed in asthma and COPD were reviewed.
Collapse
Affiliation(s)
- Mohammad Reza Khazdair
- a Pharmaceutical Research Center and Department of Physiology, School of Medicine .,b Student Research Committee , and
| | - Mohammad Hossein Boskabady
- c Neurogenic Inflammation Research Centre and Department of Physiology, School of Medicine, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Vahideh Ghorani
- a Pharmaceutical Research Center and Department of Physiology, School of Medicine
| |
Collapse
|
9
|
Park WH, Kang S, Piao Y, Pak CJ, Oh MS, Kim J, Kang MS, Pak YK. Ethanol extract of Bupleurum falcatum and saikosaponins inhibit neuroinflammation via inhibition of NF-κB. JOURNAL OF ETHNOPHARMACOLOGY 2015; 174:37-44. [PMID: 26231448 DOI: 10.1016/j.jep.2015.07.039] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/16/2015] [Accepted: 07/27/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of Bupleurum falcatum L. (BF) has been used in traditional Korean and Chinese medicines for over 2000 years to treat infections, fever, and chronic liver diseases. Among the many active compounds in BF ethanol extract (BFE), saikosaponins exert pharmacological activities including anti-inflammatory effects. Activated microglial cells release a variety of pro-inflammatory substances, leading to neuronal cell death and neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. The aim of the present study was to investigate the mechanism of the anti-neuroinflammatory effects of BFE using lipopolysaccharide (LPS)-stimulated microglial cells and LPS-intraperitoneal injected C57BL/6 mice. MATERIALS AND METHODS Dried roots of BF were extracted with 70% ethanol (tenfold volume) on a stirring plate for 24h at room temperature to prepare BFE. Pure saikosaponins (SB3, SB4, and SD) were prepared by solvent extraction and column chromatography fractionation. BV2 murine microglial cells were treated with BFE or saikosaponins for 4h and stimulated with LPS. Generation of nitric oxide (NO), inflammatory cytokines, and reactive oxygen species (ROS) from activated microglial cells were monitored. The effects of BFE on NF-κB activation were determined using RT-PCR, reporter assay, and immunostaining. The in vivo effects of BFE were also assessed by immunohistochemical staining of tissue sections from LPS-injected mouse brains. RESULTS Treatment with BFE or saikosaponins dose-dependently attenuated LPS-induced production of NO, iNOS mRNA, and ROS by 30-50%. They reduced LPS-mediated increases in the mRNA levels of IL-6, IL-1β, and TNF-α by approximately 30-70% without affecting cell viability, and decreased LPS-mediated NF-κB activity via reducing p65/RELA mRNA, transcriptional activity, and nuclear localization of NF-κB. BFE also reduced LPS-induced activation of microglia and astrocytes in the hippocampus and substantia nigra of LPS-injected mice. CONCLUSION Our data suggest that BFE may be effective for reducing neuroinflammation-mediated neurodegeneration through suppressing NF-κB-mediated inflammatory pathways.
Collapse
Affiliation(s)
- Wook Ha Park
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Sora Kang
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Ying Piao
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Christine Jeehye Pak
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Myung Sook Oh
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Jinwoong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Min Seo Kang
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea
| | - Youngmi Kim Pak
- Neurodegeneration Control Research Center, Department of Physiology, College of Medicine, Kyung Hee University, Seoul 130-731, Republic of Korea.
| |
Collapse
|
10
|
Al-Ali A, Singh N, Manshian B, Wilkinson T, Wills J, Jenkins GJS, Doak SH. Quantum dot induced cellular perturbations involving varying toxicity pathways. Toxicol Res (Camb) 2015. [DOI: 10.1039/c4tx00175c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Quantum dots (QD) with varying surface chemistry can have an impact on cellular uptake and a range of indicators for cell perturbation.
Collapse
Affiliation(s)
- Abdullah Al-Ali
- Institute of Life Science
- College of Medicine
- Swansea University
- Swansea
- UK
| | - Neenu Singh
- Institute of Life Science
- College of Medicine
- Swansea University
- Swansea
- UK
| | - Bella Manshian
- Biomedical NMR unit-MoSAIC
- Department of Medicine
- KU Leuven
- B-3000 Leuven
- Belgium
| | - Tom Wilkinson
- Institute of Life Science
- College of Medicine
- Swansea University
- Swansea
- UK
| | - John Wills
- Institute of Life Science
- College of Medicine
- Swansea University
- Swansea
- UK
| | | | - Shareen H. Doak
- Institute of Life Science
- College of Medicine
- Swansea University
- Swansea
- UK
| |
Collapse
|
11
|
Shohrati M, Karimzadeh I, Saburi A, Khalili H, Ghanei M. The role ofN-acetylcysteine in the management of acute and chronic pulmonary complications of sulfur mustard: a literature review. Inhal Toxicol 2014; 26:507-23. [DOI: 10.3109/08958378.2014.920439] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
12
|
Keyser BM, Andres DK, Holmes WW, Paradiso D, Appell A, Letukas VA, Benton B, Clark OE, Gao X, Ray P, Anderson DR, Ray R. Mustard Gas Inhalation Injury. Int J Toxicol 2014; 33:271-281. [DOI: 10.1177/1091581814532959] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mustard gas (sulfur mustard [SM], bis-[2-chloroethyl] sulfide) is a vesicating chemical warfare agent and a potential chemical terrorism agent. Exposure of SM causes debilitating skin blisters (vesication) and injury to the eyes and the respiratory tract; of these, the respiratory injury, if severe, may even be fatal. Therefore, developing an effective therapeutic strategy to protect against SM-induced respiratory injury is an urgent priority of not only the US military but also the civilian antiterrorism agencies, for example, the Homeland Security. Toward developing a respiratory medical countermeasure for SM, four different classes of therapeutic compounds have been evaluated in the past: anti-inflammatory compounds, antioxidants, protease inhibitors and antiapoptotic compounds. This review examines all of these different options; however, it suggests that preventing cell death by inhibiting apoptosis seems to be a compelling strategy but possibly dependent on adjunct therapies using the other drugs, that is, anti-inflammatory, antioxidant, and protease inhibitor compounds.
Collapse
Affiliation(s)
- Brian M. Keyser
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Devon K. Andres
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Wesley W. Holmes
- Analytical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Danielle Paradiso
- Analytical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Ashley Appell
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Valerie A. Letukas
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Betty Benton
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Offie E. Clark
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Xiugong Gao
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Prabhati Ray
- Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Dana R. Anderson
- Analytical Toxicology Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Radharaman Ray
- Research Division, US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| |
Collapse
|
13
|
Malaviya R, Venosa A, Hall L, Gow AJ, Sinko PJ, Laskin JD, Laskin DL. Attenuation of acute nitrogen mustard-induced lung injury, inflammation and fibrogenesis by a nitric oxide synthase inhibitor. Toxicol Appl Pharmacol 2012; 265:279-91. [PMID: 22981630 DOI: 10.1016/j.taap.2012.08.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 08/25/2012] [Accepted: 08/27/2012] [Indexed: 11/19/2022]
Abstract
Nitrogen mustard (NM) is a toxic vesicant known to cause damage to the respiratory tract. Injury is associated with increased expression of inducible nitric oxide synthase (iNOS). In these studies we analyzed the effects of transient inhibition of iNOS using aminoguanidine (AG) on NM-induced pulmonary toxicity. Rats were treated intratracheally with 0.125 mg/kg NM or control. Bronchoalveolar lavage fluid (BAL) and lung tissue were collected 1 d-28 d later and lung injury, oxidative stress and fibrosis assessed. NM exposure resulted in progressive histopathological changes in the lung including multifocal lesions, perivascular and peribronchial edema, inflammatory cell accumulation, alveolar fibrin deposition, bronchiolization of alveolar septal walls, and fibrosis. This was correlated with trichrome staining and expression of proliferating cell nuclear antigen (PCNA). Expression of heme oxygenase (HO)-1 and manganese superoxide dismutase (Mn-SOD) was also increased in the lung following NM exposure, along with levels of protein and inflammatory cells in BAL, consistent with oxidative stress and alveolar-epithelial injury. Both classically activated proinflammatory (iNOS⁺ and cyclooxygenase-2⁺) and alternatively activated profibrotic (YM-1⁺ and galectin-3⁺) macrophages appeared in the lung following NM administration; this was evident within 1d, and persisted for 28 d. AG administration (50 mg/kg, 2×/day, 1d-3 d) abrogated NM-induced injury, oxidative stress and inflammation at 1d and 3d post exposure, with no effects at 7 d or 28 d. These findings indicate that nitric oxide generated via iNOS contributes to acute NM-induced lung toxicity, however, transient inhibition of iNOS is not sufficient to protect against pulmonary fibrosis.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Sahebkar A. Baicalin as a potentially promising drug for the management of sulfur mustard induced cutaneous complications: a review of molecular mechanisms. Cutan Ocul Toxicol 2012; 31:226-234. [PMID: 22107027 DOI: 10.3109/15569527.2011.633950] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Sulfur mustard (SM) is a bifunctional alkylating agent with strong blistering, irritant, mutagenic and cytotoxic properties. SM has been widely deployed as a chemical warfare agent for over a century, leading to extensive casualties. Skin is among the first and most heavily damaged organs upon SM exposure. Unfortunately, a considerable fraction of SM-intoxicated patients are still suffering from chronic cutaneous complications. While these complications adversely affect patients' quality of life, there is as yet no ideal treatment for them and therapeutic options are limited and mainly symptomatic. During recent decades, remarkable progress has been made in understanding molecular mechanisms underlying SM-induced dermatotoxicity and several intra- and extracellular targets have been identified. This review argues that baicalin, a bioactive flavonoid from the roots of Scutellaria spp., could counteract different molecular and biochemical abnormalities that mediate SM dermatotoxicity and could therefore be regarded as a promising therapeutic option for the management of SM-induced cutaneous lesions.
Collapse
Affiliation(s)
- Amirhossein Sahebkar
- Biotechnology Research Center and School of Pharmacy, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran.
| |
Collapse
|
15
|
Gao X, Anderson DR, Brown AW, Lin H, Amnuaysirikul J, Chua AL, Holmes WW, Ray P. Pathological studies on the protective effect of a macrolide antibiotic, roxithromycin, against sulfur mustard inhalation toxicity in a rat model. Toxicol Pathol 2011; 39:1056-64. [PMID: 21934141 DOI: 10.1177/0192623311422079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Macrolide antibiotics have been shown to protect airway epithelial cells and macrophages from sulfur mustard (SM)-induced cytotoxicity. In the current study, the efficacy of roxithromycin in ameliorating SM-induced respiratory injury was further evaluated in a rat model. Anesthetized rats (N = 8/group) were intratracheally exposed to SM by vapor inhalation. For the drug treatment groups, rats were orally given 10, 20, or 40 mg/kg roxithromycin one hr prior to exposure and every twenty-four hr thereafter. After one, three, or seven days of treatment, sections of the lung were examined and scored for histopathological parameters. Treatment with roxithromycin ameliorated many of the symptoms caused by SM in some animals. In particular, treatment at 40 mg/kg for three days showed significant improvements (p < .05) over the untreated group. When the evaluation was focused on trachea, treatment with roxithromycin for three days showed a trend of dose-dependent protection; moreover, the groups treated with 20 or 40 mg/kg of roxithromycin were statistically different (p < .001 and p < .05, respectively) from the untreated group. These results suggest that roxithromycin protects against some damages associated with SM injury in the lung, particularly in the upper respiratory tract.
Collapse
Affiliation(s)
- Xiugong Gao
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Seagrave J, Weber WM, Grotendorst GR. Sulfur mustard vapor effects on differentiated human lung cells. Inhal Toxicol 2011; 22:896-902. [PMID: 20569120 DOI: 10.3109/08958378.2010.493901] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
CONTEXT Sulfur mustard (SM) causes skin blistering and long-term pulmonary dysfunction. Its adverse effects have been studied in battlefield-exposed humans, but lack of knowledge regarding confounding factors makes interpretation challenging. Animal studies are critical to understanding mechanisms, but differences between animals and humans must be addressed. Studies of cultured human cells can bridge animal studies and humans. OBJECTIVE Evaluate effects of SM vapor on airway cells. MATERIALS AND METHODS We examined responses of differentiated human tracheal/bronchial epithelial cells, cultured at an air-liquid interface, to SM vapors. SM effects on metabolic activity (Water Soluble Tetrazolium (WST) assay), cytokine and metalloproteinase secretion, and cellular heme oxygenase 1 (HO-1), an oxidative stress indicator, were measured after 24 h. RESULTS At noncytotoxic levels of exposure, interleukin 8 and matrix metalloproteinase-13 were significantly increased in these cultures, but HO-1 was not significantly affected. DISCUSSION AND CONCLUSION Exposure of differentiated airway epithelial cells to sub-cytotoxic levels of SM vapor induced inflammatory and degradative responses that could contribute to the adverse health effects of inhaled SM.
Collapse
|
17
|
Matsumura Y, Mitani A, Suga T, Kamiya Y, Kikuchi T, Tanaka S, Aino M, Noguchi T. Azithromycin may inhibit interleukin-8 through suppression of Rac1 and a nuclear factor-kappa B pathway in KB cells stimulated with lipopolysaccharide. J Periodontol 2011; 82:1623-31. [PMID: 21417583 DOI: 10.1902/jop.2011.100721] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Recent studies have shown that the 15-member macrolide antibiotic azithromycin (AZM) not only has antibacterial activity, but also results in the role of immunomodulator. Interleukin (IL)-8 is an important inflammatory mediator in periodontal disease. However, there have been no reports on the effects of AZM on IL-8 production from human oral epithelium. Therefore, we investigated the effects of AZM on IL-8 production in an oral epithelial cell line. METHODS KB cells were stimulated by Escherichia coli or Aggregatibacter actinomycetemcomitans (previously Actinobacillus actinomycetemcomitans) lipopolysaccharide (LPS) with or without AZM. IL-8 mRNA and protein expression and production in response to LPS were analyzed by quantitative polymerase chain reaction, flow cytometry, and enzyme-linked immunosorbent assay. The activation of nuclear factor-kappa B (NF-κB) and Rac1, which is important for IL-8 expression, was analyzed by enzyme-linked immunosorbent assay and Western blotting, respectively. RESULTS IL-8 mRNA expression, IL-8 production, and NF-κB activation in LPS-stimulated KB cells were inhibited by the addition of AZM. LPS-induced Rac1 activation was also suppressed by AZM. CONCLUSIONS This study suggests that AZM inhibits LPS-induced IL-8 production in an oral epithelial cell line, in part caused by the suppression of Rac1 and NF-κB activation. The use of AZM might provide possible benefits in periodontal therapy, with respect to both its antibacterial action and apparent anti-inflammatory effect.
Collapse
Affiliation(s)
- Yusuke Matsumura
- Department of Periodontology, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ghabili K, Agutter PS, Ghanei M, Ansarin K, Panahi Y, Shoja MM. Sulfur mustard toxicity: history, chemistry, pharmacokinetics, and pharmacodynamics. Crit Rev Toxicol 2011; 41:384-403. [PMID: 21329486 DOI: 10.3109/10408444.2010.541224] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sulfur mustard (SM) and similar bifunctional agents have been used as chemical weapons for almost 100 years. Victims of high-dose exposure, both combatants and civilians, may die within hours or weeks, but low-dose exposure causes both acute injury to the eyes, skin, respiratory tract and other parts of the body, and chronic sequelae in these organs are often debilitating and have a serious impact on quality of life. Ever since they were first used in warfare in 1917, SM and other mustard agents have been the subjects of intensive research, and their chemistry, pharmacokinetics and mechanisms of toxic action are now fairly well understood. In the present article we review this knowledge and relate the molecular-biological basis of SM toxicity, as far as it has been elucidated, to the pathological effects on exposure victims.
Collapse
Affiliation(s)
- Kamyar Ghabili
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | | | | | |
Collapse
|
19
|
Sulfur mustard-induced pulmonary injury: therapeutic approaches to mitigating toxicity. Pulm Pharmacol Ther 2010; 24:92-9. [PMID: 20851203 DOI: 10.1016/j.pupt.2010.09.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 09/03/2010] [Accepted: 09/09/2010] [Indexed: 11/23/2022]
Abstract
Sulfur mustard (SM) is highly toxic to the lung inducing both acute and chronic effects including upper and lower obstructive disease, airway inflammation, and acute respiratory distress syndrome, and with time, tracheobronchial stenosis, bronchitis, and bronchiolitis obliterans. Thus it is essential to identify effective strategies to mitigate the toxicity of SM and related vesicants. Studies in animals and in cell culture models have identified key mechanistic pathways mediating their toxicity, which may be relevant targets for the development of countermeasures. For example, following SM poisoning, DNA damage, apoptosis, and autophagy are observed in the lung, along with increased expression of activated caspases and DNA repair enzymes, biochemical markers of these activities. This is associated with inflammatory cell accumulation in the respiratory tract and increased expression of tumor necrosis factor-α and other proinflammatory cytokines, as well as reactive oxygen and nitrogen species. Matrix metalloproteinases are also upregulated in the lung after SM exposure, which are thought to contribute to the detachment of epithelial cells from basement membranes and disruption of the pulmonary epithelial barrier. Findings that production of inflammatory mediators correlates directly with altered lung function suggests that they play a key role in toxicity. In this regard, specific therapeutic interventions currently under investigation include anti-inflammatory agents (e.g., steroids), antioxidants (e.g., tocopherols, melatonin, N-acetylcysteine, nitric oxide synthase inhibitors), protease inhibitors (e.g., doxycycline, aprotinin, ilomastat), surfactant replacement, and bronchodilators. Effective treatments may depend on the extent of lung injury and require a multi-faceted pharmacological approach.
Collapse
|
20
|
Malaviya R, Sunil VR, Cervelli J, Anderson DR, Holmes WW, Conti ML, Gordon RE, Laskin JD, Laskin DL. Inflammatory effects of inhaled sulfur mustard in rat lung. Toxicol Appl Pharmacol 2010; 248:89-99. [PMID: 20659490 DOI: 10.1016/j.taap.2010.07.018] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 07/16/2010] [Accepted: 07/19/2010] [Indexed: 12/19/2022]
Abstract
Inhalation of sulfur mustard (SM), a bifunctional alkylating agent that causes severe lung damage, is a significant threat to both military and civilian populations. The mechanisms mediating its cytotoxic effects are unknown and were investigated in the present studies. Male rats Crl:CD(SD) were anesthetized, and then intratracheally intubated and exposed to 0.7-1.4mg/kg SM by vapor inhalation. Animals were euthanized 6, 24, 48h or 7days post-exposure and bronchoalveolar lavage fluid (BAL) and lung tissue collected. Exposure of rats to SM resulted in rapid pulmonary toxicity, including focal ulceration and detachment of the trachea and bronchial epithelia from underlying mucosa, thickening of alveolar septal walls and increased numbers of inflammatory cells in the tissue. There was also evidence of autophagy and apoptosis in the tissue. This was correlated with increased BAL protein content, a marker of injury to the alveolar epithelial lining. SM exposure also resulted in increased expression of markers of inflammation including cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNFα), inducible nitric oxide synthase (iNOS), and matrix metalloproteinase-9 (MMP-9), each of which has been implicated in pulmonary toxicity. Whereas COX-2, TNFα and iNOS were mainly localized in alveolar regions, MMP-9 was prominent in bronchial epithelium. In contrast, expression of the anti-oxidant hemeoxygenase, and the anti-inflammatory collectin, surfactant protein-D, decreased in the lung after SM exposure. These data demonstrate that SM-induced oxidative stress and injury are associated with the generation of cytotoxic inflammatory proteins which may contribute to the pathogenic response to this vesicant.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Gao X, Ray R, Xiao Y, Ishida K, Ray P. Macrolide antibiotics improve chemotactic and phagocytic capacity as well as reduce inflammation in sulfur mustard-exposed monocytes. Pulm Pharmacol Ther 2010; 23:97-106. [DOI: 10.1016/j.pupt.2009.10.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 10/06/2009] [Accepted: 10/21/2009] [Indexed: 10/20/2022]
|
22
|
Wang D, Lu Z, Hu L, Zhang Y, Hu X. Macrolide antibiotics aggravate experimental autoimmune encephalomyelitis and inhibit inducible nitric oxide synthase. Immunol Invest 2010; 38:602-12. [PMID: 19811424 DOI: 10.1080/08820130903062194] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Recent studies have implicated Chlamydia pneumoniae (C. pneumoniae) is present in a subset of patients with multiple sclerosis (MS) in which C. pneumoniae could act as a cofactor in the development of the disease. Macrolide antibiotics are most widely used anti-chlamydial agents and have immunomodulatory effect independently of their anti-bacterial activity. To investigate their effects on experimental autoimmune encephalomyelitis (EAE), EAE was induced by immunization with MBP68-86 peptide emulsified in complete Freund's adjuvant (CFA). Clarithromycin (CM) or azithromycin (AM, 50 mg/100 g body weight) was administrated daily from day 2 before immunization. All rats developed and survived EAE, but the groups administrated CM or AM had more severe symptoms. On day 11 post-immunization, mononuclear cells (MNCs) were prepared from the spleen of control group and cultured with or without macrolide antibiotics (10mug/ml). We evaluated nitric oxide (NO) production in the serum and culture supernatant. Inducible nitric oxide synthase (iNOS) mRNA and protein expression in the spinal cords and cultured MNCs were measured. The results showed that CM and AM similarly inhibited NO production and iNOS mRNA and protein expression in vivo and in vitro. Macrolide antibiotics may aggravate EAE by inhibiting iNOS mRNA and protein expression. Further studies are needed to investigate the effect of macrolide antibiotics on MS and to compare the effect of different anti-chlamydial antibiotics on MS.
Collapse
Affiliation(s)
- Dunjing Wang
- Department of Neurology, The Third Affiliated hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | |
Collapse
|