1
|
Sana Vilela V, Andrighetti de Oliveira Braga K, Moreira Ruiz L, Nepomuceno NA, Oliveira Melo P, Manzuti GM, Alcantara de Oliveira Costa V, de Campos Ramos J, Tadeu Correia A, Pêgo-Fernandes PM. Anti-inflammatory effect of thalidomide in an experimental lung donor model of brain death. Sci Rep 2024; 14:8796. [PMID: 38627574 PMCID: PMC11021429 DOI: 10.1038/s41598-024-59267-1] [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: 01/05/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
Lung transplantation stands as a vital treatment for severe lung diseases, primarily sourcing organs from donors with brain death (BD). This research delved into the potential anti-inflammatory effects of thalidomide in rats with BD-induced lung complications. In this study twenty-four Wistar rats were divided into three groups: the control (CTR), brain death (BD) and brain death + thalidomide (TLD) groups. Post specific procedures, a 360 min monitoring period ensued. Comprehensive analyses of blood and heart-lung samples were conducted. Elevated IL-6 levels characterized both BD and TLD groups relative to the CTR (p = 0.0067 and p = 0.0137). Furthermore, TNF-α levels were notably higher in the BD group than both CTR and TLD (p = 0.0152 and p = 0.0495). Additionally, IL-1β concentrations were significantly pronounced in both BD and TLD compared to CTR, with the BD group surpassing TLD (p = 0.0256). Immunohistochemical assessments revealed augmented NF-ĸB expression in the BD group in comparison to both CTR and TLD (p = 0.0006 and p = 0.0005). With this study we can conclude that BD induced acute pulmonary inflammation, whereas thalidomide manifested a notable capability in diminishing key inflammatory markers, indicating its prospective therapeutic significance in lung transplantation scenarios.
Collapse
Affiliation(s)
- Vanessa Sana Vilela
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Rua Dr. Eneas de Carvalho Aguiar 44, bloco 1, SS, sala 25, Cerqueira Cezar, Sao Paulo, SP, 05403-000, Brazil.
| | - Karina Andrighetti de Oliveira Braga
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Liliane Moreira Ruiz
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Natalia Aparecida Nepomuceno
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Paolo Oliveira Melo
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Giovana Maria Manzuti
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Vinícius Alcantara de Oliveira Costa
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Jhonatan de Campos Ramos
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Aristides Tadeu Correia
- Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Paulo Manuel Pêgo-Fernandes
- Departamento de Cardiopneumologia, Laboratorio de Pesquisa em Cirurgia Toracica, Instituto do Coração, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
2
|
Barbarossa A, Ceramella J, Carocci A, Iacopetta D, Rosato A, Limongelli F, Carrieri A, Bonofiglio D, Sinicropi MS. Benzothiazole-Phthalimide Hybrids as Anti-Breast Cancer and Antimicrobial Agents. Antibiotics (Basel) 2023; 12:1651. [PMID: 38136685 PMCID: PMC10740580 DOI: 10.3390/antibiotics12121651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
The benzothiazole nucleus is a major heterocyclic scaffold whose therapeutic potential has been thoroughly explored due to its structural simplicity and ease of synthesis. In fact, several benzothiazole derivatives have been synthesized over time, demonstrating numerous pharmacological properties such as anticancer, antimicrobial, anti-inflammatory, and antioxidant activities. Herein, we propose a new series of benzothiazole-phthalimide hybrids obtained by linking the phthalimide moiety to differently substituted benzothiazole nuclei through the N atom. These compounds have been screened for their anticancer properties against two human breast cancer cell lines. Furthermore, we delved into the mechanism of action of the most active hybrid, compound 3h, by assessing its capability to damage the nuclear DNA, trigger the apoptotic process in the high metastatic MDA-MB-231 cells, and prevent cellular migration. Moreover, in view of the documented antimicrobial activities of the two scaffolds involved, we explored the antibacterial and antifungal effects of the studied compounds by means of the broth microdilution method. Among the studied compounds, 3h showed the highest antimicrobial activity, both against gram-positive and gram-negative bacterial strains belonging to the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) and against fungal strains of the Candida species with MICs values ranging from 16 to 32 µg/mL.
Collapse
Affiliation(s)
- Alexia Barbarossa
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.B.); (A.R.); (F.L.); (A.C.)
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (J.C.); (D.B.); (M.S.S.)
| | - Alessia Carocci
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.B.); (A.R.); (F.L.); (A.C.)
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (J.C.); (D.B.); (M.S.S.)
| | - Antonio Rosato
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.B.); (A.R.); (F.L.); (A.C.)
| | - Francesco Limongelli
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.B.); (A.R.); (F.L.); (A.C.)
| | - Antonio Carrieri
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.B.); (A.R.); (F.L.); (A.C.)
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (J.C.); (D.B.); (M.S.S.)
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (J.C.); (D.B.); (M.S.S.)
| |
Collapse
|
3
|
Dsouza NN, Alampady V, Baby K, Maity S, Byregowda BH, Nayak Y. Thalidomide interaction with inflammation in idiopathic pulmonary fibrosis. Inflammopharmacology 2023; 31:1167-1182. [PMID: 36966238 PMCID: PMC10039777 DOI: 10.1007/s10787-023-01193-1] [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: 11/10/2022] [Accepted: 03/04/2023] [Indexed: 03/27/2023]
Abstract
The "Thalidomide tragedy" is a landmark in the history of the pharmaceutical industry. Despite limited clinical trials, there is a continuous effort to investigate thalidomide as a drug for cancer and inflammatory diseases such as rheumatoid arthritis, lepromatous leprosy, and COVID-19. This review focuses on the possibilities of targeting inflammation by repurposing thalidomide for the treatment of idiopathic pulmonary fibrosis (IPF). Articles were searched from the Scopus database, sorted, and selected articles were reviewed. The content includes the proven mechanisms of action of thalidomide relevant to IPF. Inflammation, oxidative stress, and epigenetic mechanisms are major pathogenic factors in IPF. Transforming growth factor-β (TGF-β) is the major biomarker of IPF. Thalidomide is an effective anti-inflammatory drug in inhibiting TGF-β, interleukins (IL-6 and IL-1β), and tumour necrosis factor-α (TNF-α). Thalidomide binds cereblon, a process that is involved in the proposed mechanism in specific cancers such as breast cancer, colon cancer, multiple myeloma, and lung cancer. Cereblon is involved in activating AMP-activated protein kinase (AMPK)-TGF-β/Smad signalling, thereby attenuating fibrosis. The past few years have witnessed an improvement in the identification of biomarkers and diagnostic technologies in respiratory diseases, partly because of the COVID-19 pandemic. Hence, investment in clinical trials with a systematic plan can help repurpose thalidomide for pulmonary fibrosis.
Collapse
Affiliation(s)
- Nikitha Naomi Dsouza
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Varun Alampady
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krishnaprasad Baby
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Swastika Maity
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bharath Harohalli Byregowda
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
4
|
Gheshlaghi F, Haghirzavareh J, Wong A, Golshiri P, Gheshlaghi S, Eizadi-Mood N. Prediction of mortality and morbidity following paraquat poisoning based on trend of liver and kidney injury. BMC Pharmacol Toxicol 2022; 23:67. [PMID: 36068596 PMCID: PMC9450277 DOI: 10.1186/s40360-022-00609-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 08/31/2022] [Indexed: 11/10/2022] Open
Abstract
Background Paraquat is a non-selective herbicide that causes severe tissue damage in various organs including the liver and kidney. The aim of this study was to determine the trend of the liver and kidney injury in patients with paraquat poisoning. Methods This retrospective cross-sectional study was performed at the Khorshid Hospital referral poisoning emergency center. The medical records of all patients with acute paraquat poisoning admitted from March 2017 to October 2020 were reviewed. Demographic factors, liver and kidney function tests and outcomes were recorded. Patients were divided into two groups based on the outcome of mortality (death or survived). The two groups were compared in terms of changes in creatinine and liver enzymes during hospitalization. Results A significant difference in mean creatinine levels between the two groups was observed from the third day after admission. The peak median Cr was 3.5 mg/dl for deceased patients in day 6 and 1.47 mg/dl for survived patients on 4th day. Minor elevations of ALT and AST were present in those who died. Logistic regression analysis shows patients who had level of creatinine higher than normal from the 2nd to 6th day post overdose, the risk of mortality was 4.83 to 7.44 times more than patients with normal creatinine level. The mean (SD) area under the curve for outcome prediction was reported to be excellent for creatinine on the 8th day post overdose (85.7 ± 13.2). Creatinine was higher than 2 on the 8th day post ingestion and had a sensitivity 100% and specificity 85.7% for mortality prediction (P value, 0.05). Conclusions The risk of mortality secondary to paraquat ingestion was highly associated with a rise in creatinine. Minor elevations of ALT and AST were also present in those who died. The creatinine concentration on different days post overdose can be helpful in predicting the severity of poisoning especially when the serum paraquat levels are not available.
Collapse
Affiliation(s)
- Farzad Gheshlaghi
- Department of Clinical Toxicology, School of Medicine; Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jamileh Haghirzavareh
- Medical Practitioner, Department of Clinical Toxicology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anselm Wong
- Victorian Poisons Information Centre, Austin Toxicology and Emergency Department Austin Health, Heidelberg, and Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Parastoo Golshiri
- Department of Community Medicine and Family Physician, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shayan Gheshlaghi
- Medical Practitioner, School of Medicine, Islamic Azad University Najafabad Branch, Isfahan, Iran.
| | - Nastaran Eizadi-Mood
- Department of Clinical Toxicology, School of Medicine; Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
5
|
Estornut C, Milara J, Bayarri MA, Belhadj N, Cortijo J. Targeting Oxidative Stress as a Therapeutic Approach for Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 12:794997. [PMID: 35126133 PMCID: PMC8815729 DOI: 10.3389/fphar.2021.794997] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 01/19/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease characterized by an abnormal reepithelialisation, an excessive tissue remodelling and a progressive fibrosis within the alveolar wall that are not due to infection or cancer. Oxidative stress has been proposed as a key molecular process in pulmonary fibrosis development and different components of the redox system are altered in the cellular actors participating in lung fibrosis. To this respect, several activators of the antioxidant machinery and inhibitors of the oxidant species and pathways have been assayed in preclinical in vitro and in vivo models and in different clinical trials. This review discusses the role of oxidative stress in the development and progression of IPF and its underlying mechanisms as well as the evidence of oxidative stress in human IPF. Finally, we analyze the mechanism of action, the efficacy and the current status of different drugs developed to inhibit the oxidative stress as anti-fibrotic therapy in IPF.
Collapse
Affiliation(s)
- Cristina Estornut
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - Javier Milara
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- *Correspondence: Cristina Estornut, ; Javier Milara,
| | - María Amparo Bayarri
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Nada Belhadj
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain
- Pharmacy Unit, University General Hospital Consortium, Valencia, Spain
- CIBERES, Health Institute Carlos III, Valencia, Spain
- Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
6
|
Zarkesh K, Entezar-Almahdi E, Ghasemiyeh P, Akbarian M, Bahmani M, Roudaki S, Fazlinejad R, Mohammadi-Samani S, Firouzabadi N, Hosseini M, Farjadian F. Drug-based therapeutic strategies for COVID-19-infected patients and their challenges. Future Microbiol 2021; 16:1415-1451. [PMID: 34812049 PMCID: PMC8610072 DOI: 10.2217/fmb-2021-0116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging epidemic-prone diseases have introduced numerous health and economic challenges in recent years. Given current knowledge of COVID-19, herd immunity through vaccines alone is unlikely. In addition, vaccination of the global population is an ongoing challenge. Besides, the questions regarding the prevalence and the timing of immunization are still under investigation. Therefore, medical treatment remains essential in the management of COVID-19. Herein, recent advances from beginning observations of COVID-19 outbreak to an understanding of the essential factors contributing to the spread and transmission of COVID-19 and its treatment are reviewed. Furthermore, an in-depth discussion on the epidemiological aspects, clinical symptoms and most efficient medical treatment strategies to mitigate the mortality and spread rates of COVID-19 is presented.
Collapse
Affiliation(s)
- Khatereh Zarkesh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elaheh Entezar-Almahdi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Ghasemiyeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Akbarian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Bahmani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrzad Roudaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rahil Fazlinejad
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Hosseini
- Department of Manufacturing & Industrial Engineering, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
7
|
Amirshahrokhi K. Thalidomide reduces glycerol-induced acute kidney injury by inhibition of NF-κB, NLRP3 inflammasome, COX-2 and inflammatory cytokines. Cytokine 2021; 144:155574. [PMID: 33975771 DOI: 10.1016/j.cyto.2021.155574] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is an important clinical complication of rhabdomyolysis. The inflammatory processes are involved in the pathogenesis of AKI induced by rhabdomyolysis. Thalidomide is an anti-inflammatory agent that has been used in the treatment of inflammatory disorders. The aim of this study was to investigate the therapeutic effect of thalidomide and its underlying mechanisms on a mouse model of rhabdomyolysis-induced AKI. Mice were injected with a single dose of glycerol (50%, 10 ml/kg, im) to induce AKI, and treated with thalidomide (40 and 80 mg/kg/day, orally) for 2 days. Renal tissue and blood samples were collected for histological and biochemical analysis. In thalidomide treated mice, blood urea nitrogen (BUN) (59.3 ± 19.6 vs. 223 ± 33 mg/dl), plasma creatinine (0.58 ± 0.3 vs. 1.28 ± 0.3 mg/dl), relative kidney weight (0.93 ± 0.13% vs. 1.22 ± 0.1%) and histopathological damage (1.5 ± 0.8 vs. 3.3 ± 1.1 score) were significantly lower as compared to the glycerol group. The results also showed that the levels of malondialdehyde (MDA) (0.13 ± 0.02 vs. 0.2 ± 0.01 µM/mg), myeloperoxidase (MPO) (0.1 ± 0.05 vs. 0.25 ± 0.02 U/mg) and the expression of nuclear factor kappa B (NF-κB) (1.7-fold), NLRP3 inflammasome (1.4-fold) and cyclooxygenase (COX)-2 (3-fold) in renal tissue were significantly lower in thalidomide treated group than those in the glycerol group. Thalidomide treatment resulted in lower renal pro-inflammatory cytokines tumor necrosis factor (TNF)-α (6.7 ± 0.8 vs. 12.3 ± 1.2 ng/ml), interleukin (IL)-1β (3.2 ± 0.5 vs. 5.1 ± 0.3 pg/mg), IL-6 (24.7 ± 2.4 vs. 33 ± 3 pg/mg) and transforming growth factor (TGF)-β1 (0.6 ± 0.17 vs. 1.56 ± 0.24 ng/ml) than those in the glycerol treated mice. In addition the levels of monocyte chemoattractant protein (MCP)-1 (9.5 ± 1 vs. 12.8 ± 1.1 pg/mg) and intercellular adhesion molecule (ICAM)-1 (22.8 ± 7.8 vs. 53.3 ± 5.5 pg/mg) were significantly lower in renal tissue of mice treated with thalidomide as compared to the glycerol treated mice. In conclusion these data revealed that thalidomide may be a potential therapeutic approach against rhabdomyolysis-induced AKI through inhibition of inflammatory responses.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
8
|
Vassileva S, Mateeva V, Drenovska K. Drug repurposing of dermatologic medications to treat coronavirus disease 2019: Science or fiction? Clin Dermatol 2021; 39:430-445. [PMID: 34518001 PMCID: PMC7959882 DOI: 10.1016/j.clindermatol.2021.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
No pharmaceutical products have been demonstrated to be safe and effective to specifically treat coronavirus disease 2019 (COVID-19); therefore, the therapy administered to infected patients remains symptomatic and empiric. Alongside the development of new, often high-cost drugs, a different tactic is being applied in parallel, investigating long-established, inexpensive medications originally designed for a variety of diseases to study their potential in treating COVID-19. The skin is the largest organ of the human body. With more than 3,000 skin conditions identified, the specialty of dermatology offers a rich armamentarium of systemic therapeutic agents aimed to treat the various chronic immunologically mediated, metabolic, infectious, occupational, inherited, or paraneoplastic dermatoses. Dermatologists have extensive experience with many drugs that have demonstrated promising in vitro antiviral action (directly targeting the viral replication). Many of these drugs have been used as nonspecific immunosuppressive strategies, such as glucocorticoids, synthetic antimalarials, colchicine, or other immunomodulators, and a number of targeted therapeutics have been directed at controlling hyperinflammatory processes similar to the "cytokine storm" associated with COVID-19 infection. We discuss several dermatologic drugs that have already been used or may have a promising role in the treatment of COVID-19.
Collapse
Affiliation(s)
- Snejina Vassileva
- Department of Dermatology and Venereology, Medical University-Sofia, Sofia, Bulgaria.
| | - Valeria Mateeva
- Department of Dermatology and Venereology, Medical University-Sofia, Sofia, Bulgaria
| | - Kossara Drenovska
- Department of Dermatology and Venereology, Medical University-Sofia, Sofia, Bulgaria
| |
Collapse
|
9
|
Huang KC, Li JC, Wang SM, Cheng CH, Yeh CH, Lin LS, Chiu HY, Chang CY, Chuu JJ. The effects of carbon monoxide releasing molecules on paraquat-induced pulmonary interstitial inflammation and fibrosis. Toxicology 2021; 456:152750. [PMID: 33737140 DOI: 10.1016/j.tox.2021.152750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/10/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023]
Abstract
Paraquat, an herbicide used extensively worldwide, can cause severe toxicity in humans and animals, leading to irreversible, lethal lung fibrosis. The potential of CO-releasing molecules (CORMs), substances that release CO (Carbon monoxide) within animal tissues, for treating paraquat-induced ROS generation and inflammation is investigated here. Our results show that the fast CO releaser CORM-3 (4-20 μM) acts as a potential scavenger of free radicals and decreases fibrosis progression by inhibiting paraquat-induced overexpression of connective tissue growth factor and angiotensin II in MRC-5 cells. The slow CO releaser CORM-A1 (5 mg/kg) clearly decreased expression of the lung profibrogenic cytokines COX-2, TNF-α, and α-SMA and serum hydroxyproline, resulting in a lower mortality rate in paraquat-treated mice. Mice treated with higher-dose CORM-A1 (10 mg/kg) had relatively intact lung lobes and fewer fibrotic patches by gross observation, with less collagen deposition, mesangial matrix accumulation, and pulmonary fibrosis resulting from the mitigation of TGF-β overexpression. In conclusion, our data demonstrate for the first time that CORM-A1 alleviated the development of the fibrotic process and improved survival rate in mice exposed to PQ, would be an attractive therapeutic approach to attenuate the progression of pulmonary fibrosis following PQ exposure.
Collapse
Affiliation(s)
- Kuo-Ching Huang
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Hospital, Liouying, Tainan, Taiwan; Department of Environmental and Occupational Health, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Jui-Chen Li
- Pharmacy Department, Wei-Gong Memorial Hospital, Miaoli, Taiwan
| | - Shu-Mei Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei, Taiwan
| | - Chia-Hui Cheng
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chun-Hsiang Yeh
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Li-Syun Lin
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hsin-Yi Chiu
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chia-Yu Chang
- Department of Neurology, Chi-Mei Medical Center, Tainan, Taiwan; Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| | - Jiunn-Jye Chuu
- Pharmacy Department, Wei-Gong Memorial Hospital, Miaoli, Taiwan; Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| |
Collapse
|
10
|
Zheng F, Zhu J, Zhang W, Fu Y, Lin Z. Thal protects against paraquat-induced lung injury through a microRNA-141/HDAC6/IκBα-NF-κB axis in rat and cell models. Basic Clin Pharmacol Toxicol 2021; 128:334-347. [PMID: 33015978 PMCID: PMC7894280 DOI: 10.1111/bcpt.13505] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 12/16/2022]
Abstract
The protective functions of thalidomide in paraquat (PQ)-induced injury have been reported. But the mechanisms remain largely unknown. In this research, a PQ-treated rat model was established and further treated with thalidomide. Oedema and pathological changes, oxidative stress, inflammation, fibrosis and cell apoptosis in rat lungs were detected. A PQ-treated RLE-6TN cell model was constructed, and the viability and apoptosis rate of cells were measured. Differentially expressed microRNAs (miRNAs) after thalidomide administration were screened out. Binding relationship between miR-141 and histone deacetylase 6 (HDAC6) was validated. Altered expression of miR-141 and HDAC6 was introduced to identify their involvements in thalidomide-mediated events. Consequently, thalidomide administration alone exerted no damage to rat lungs; in addition it reduced PQ-induced oedema. The oxidative stress, inflammation and cell apoptosis in rat lungs were reduced by thalidomide. In RLE-6TN cells, thalidomide increased cell viability and decreased apoptosis. miR-141 was responsible for thalidomide-mediated protective events by targeting HDAC6. Overexpression of HDAC6 blocked the protection of thalidomide against PQ-induced injury via activating the IkBα-NF-κB signalling pathway. Collectively, this study evidenced that thalidomide protects lung tissues from PQ-induced injury through a miR-141/HDAC6/IkBα-NF-κB axis.
Collapse
Affiliation(s)
- Fenshuang Zheng
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Junbo Zhu
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Wei Zhang
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Yangshan Fu
- Department of Emergency MedicineSecond People's Hospital of Yunnan ProvinceKunmingChina
| | - Zhaoheng Lin
- Department of Critical Care MedicinePeople's Hospital of Xishuangbanna Dai Nationality Autonomous PrefecturePingpongChina
| |
Collapse
|
11
|
Heimfarth L, Serafini MR, Martins-Filho PR, Quintans JDSS, Quintans-Júnior LJ. Drug repurposing and cytokine management in response to COVID-19: A review. Int Immunopharmacol 2020; 88:106947. [PMID: 32919216 PMCID: PMC7457938 DOI: 10.1016/j.intimp.2020.106947] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19), the infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is an aggressive disease that attacks the respiratory tract and has a higher fatality rate than seasonal influenza. The COVID-19 pandemic is a global health crisis, and no specific therapy or drug has been formally recommended for use against SARS-CoV-2 infection. In this context, it is a rational strategy to investigate the repurposing of existing drugs to use in the treatment of COVID-19 patients. In the meantime, the medical community is trialing several therapies that target various antiviral and immunomodulating mechanisms to use against the infection. There is no doubt that antiviral and supportive treatments are important in the treatment of COVID-19 patients, but anti-inflammatory therapy also plays a pivotal role in the management COVID-19 patients due to its ability to prevent further injury and organ damage or failure. In this review, we identified drugs that could modulate cytokines levels and play a part in the management of COVID-19. Several drugs that possess an anti-inflammatory profile in others illnesses have been studied in respect of their potential utility in the treatment of the hyperinflammation induced by SAR-COV-2 infection. We highlight a number of antivirals, anti-rheumatic, anti-inflammatory, antineoplastic and antiparasitic drugs that have been found to mitigate cytokine production and consequently attenuate the "cytokine storm" induced by SARS-CoV-2. Reduced hyperinflammation can attenuate multiple organ failure, and even reduce the mortality associated with severe COVID-19. In this context, despite their current unproven clinical efficacy in relation to the current pandemic, the repurposing of drugs with anti-inflammatory activity to use in the treatment of COVID-19 has become a topic of great interest.
Collapse
Affiliation(s)
- Luana Heimfarth
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), São Cristóvão, SE 49100-000 Brazil; Graduate Program of Health Sciences (PPGCS), São Cristóvão, SE 49100-000 Brazil.
| | - Mairim Russo Serafini
- Graduate Program of Pharmaceutical Sciences (PPGCF). Federal University of Sergipe (UFS), São Cristóvão, SE 49100-000 Brazil
| | | | - Jullyana de Souza Siqueira Quintans
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), São Cristóvão, SE 49100-000 Brazil; Graduate Program of Health Sciences (PPGCS), São Cristóvão, SE 49100-000 Brazil
| | - Lucindo José Quintans-Júnior
- Laboratory of Neuroscience and Pharmacological Assays (LANEF), São Cristóvão, SE 49100-000 Brazil; Graduate Program of Health Sciences (PPGCS), São Cristóvão, SE 49100-000 Brazil; Graduate Program of Pharmaceutical Sciences (PPGCF). Federal University of Sergipe (UFS), São Cristóvão, SE 49100-000 Brazil
| |
Collapse
|
12
|
Wang N, Li Y, Wang X, Ma Z, Wang Y, Zhang C, Yuan Y, Zhao M. Inhibition of TBK1 by amlexanox attenuates paraquat-induced acute lung injury. Toxicology 2020; 443:152555. [DOI: 10.1016/j.tox.2020.152555] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/24/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
|
13
|
Khalil A, Kamar A, Nemer G. Thalidomide-Revisited: Are COVID-19 Patients Going to Be the Latest Victims of Yet Another Theoretical Drug-Repurposing? Front Immunol 2020; 11:1248. [PMID: 32574274 PMCID: PMC7270289 DOI: 10.3389/fimmu.2020.01248] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/18/2020] [Indexed: 12/22/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a worldwide threatening health issue. The progression of this viral infection occurs in the airways of the lungs with an exaggerated inflammatory response referred to as the "cytokine storm" that can lead to lethal lung injuries. In the absence of an effective anti-viral molecule and until the formulation of a successful vaccine, anti-inflammatory drugs might offer a complementary tool for controlling the associated complications of COVID-19 and thus decreasing the subsequent fatalities. Drug repurposing for several molecules has emerged as a rapid temporary solution for COVID-19. Among these drugs is Thalidomide; a historically emblematic controversial molecule that harbors an FDA approval for treating erythema nodosum leprosum (ENL) and multiple myeloma (MM). Based on just one-case report that presented positive outcomes in a patient treated amongst others with Thalidomide, two clinical trials on the efficacy and safety of Thalidomide in treating severe respiratory complications in COVID-19 patients were registered. Yet, the absence of substantial evidence on Thalidomide usage in that context along with the discontinued studies on the efficiency of this drug in similar pulmonary diseases, might cause a significant obstacle for carrying out further clinical evaluations. Herein, we will discuss the theoretical effectiveness of Thalidomide in attenuating inflammatory complications that are encountered in COVID-19 patients while pinpointing the lack of the needed evidences to move forward with this drug.
Collapse
Affiliation(s)
- Athar Khalil
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Amina Kamar
- Vascular Medicine Program, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
- Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
14
|
Klotho Alleviates Lung Injury Caused by Paraquat via Suppressing ROS/P38 MAPK-Regulated Inflammatory Responses and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1854206. [PMID: 32509139 PMCID: PMC7244968 DOI: 10.1155/2020/1854206] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 01/09/2023]
Abstract
Acute lung injury (ALI) induced by paraquat (PQ) progresses rapidly with high mortality; however, there is no effective treatment, and the specific mechanism is not well understood. The antiaging protein klotho (KL) has multiple functions and exerts significant influences on various pathophysiological processes. This work evaluated the impact of KL on PQ-induced ALI and investigated its underlying mechanisms. As for in vivo research, C57BL/6 mice were treated with PQ (30 mg/kg) intraperitoneal (IP) injection to create a toxicity model of ALI (PQ group). The mice were divided into control group, KL group, PQ group, and PQ+KL group. For in vitro experiment, A549 cells were incubated with or without KL and then treated in the presence or absence of PQ for 24 h. In vivo result indicated that KL reduced the mortality, reduced IL-1β and IL-6 in the bronchoalveolar lavage fluid (BALF), attenuated ALI, and decreased apoptosis in situ. In vitro result revealed that KL significantly improved cell viability, reduced the levels of IL-1β and IL-6 in culture supernatants, suppressed cell apoptosis, inhibited caspase-3 activation, and enhanced mitochondrial membrane potential (ΔΨm) after PQ treatment. Besides, KL effectively abated reactive oxygen species (ROS) production, improved GSH content, and lowered lipid peroxidation in PQ-exposed A549 cells. Further experiments indicated that phosphorylated JNK and P38 MAPK was increased after PQ treatment; however, KL pretreatment could significantly lower the phosphorylation of P38 MAPK. Suppression of P38 MAPK improved cell viability, alleviated inflammatory response, and reduced apoptosis-related signals; however, it had no obvious effect on the production of ROS. Treatment with N-acetylcysteine (NAC), a classic ROS scavenger, could suppress ROS production and P38 MAPK activation. These findings suggested that KL could alleviate PQ-caused ALI via inhibiting ROS/P38 MAPK signaling-regulated inflammatory responses and mitochondria-dependent apoptosis.
Collapse
|
15
|
Zhao Y, Chen B, He Y, Zhang S, Qiu Y, Feng R, Yang H, Zeng Z, Ben-Horin S, Chen M, Mao R. Risk Factors Associated with Impaired Ovarian Reserve in Young Women of Reproductive Age with Crohn's Disease. Intest Res 2020; 18:200-209. [PMID: 32224833 PMCID: PMC7206342 DOI: 10.5217/ir.2019.00103] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND/AIMS Crohn's disease (CD) primarily affects young female adults of reproductive age. Few studies have been conducted on this population's ovarian reserve status. The aim of study was to investigate potential risk factors associated with low ovarian reserve, as reflected by serum anti-Müllerian hormone (AMH) in women of reproductive age with CD. METHODS This was a case-control study. Cases included 87 patients with established CD, and healthy controls were matched by age, height and weight in a 1:1 ratio. Serum AMH levels were measured by enzyme-linked immunosorbent assay. RESULTS The average serum AMH level was significantly lower in CD patients than in control group (2.47±2.08 ng/mL vs. 3.87±1.96 ng/mL, respectively, P<0.001). Serum AMH levels were comparable between CD patients and control group under 25 years of age (4.41±1.52 ng/mL vs. 3.49±2.10 ng/mL, P=0.06), however, serum AMH levels were significantly lower in CD patients over 25 years of age compared to control group (P<0.05). Multivariable analysis showed that an age greater than 25 (odds ratio [OR], 10.03; 95% confidence interval [CI], 1.90-52.93, P=0.007), active disease state (OR, 27.99; 95% CI, 6.13-127.95, P<0.001) and thalidomide use (OR, 15.66; 95% CI, 2.22-110.65, P=0.006) were independent risk factors associated with low ovarian reserve (serum AMH levels <2 ng/mL) in CD patients. CONCLUSIONS Ovarian reserve is impaired in young women of reproductive age with CD. Age over 25 and an active disease state were both independently associated with low ovarian reserve. Thalidomide use could result in impaired ovarian reserve.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Baili Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yao He
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shenghong Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yun Qiu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Rui Feng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hongsheng Yang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhirong Zeng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shomron Ben-Horin
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Gastroenterology, Sheba Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
16
|
Khalil A, Kamar A, Nemer G. Thalidomide-Revisited: Are COVID-19 Patients Going to Be the Latest Victims of Yet Another Theoretical Drug-Repurposing? Front Immunol 2020. [PMID: 32574274 DOI: 10.3389/fimmu.2020.01248/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a worldwide threatening health issue. The progression of this viral infection occurs in the airways of the lungs with an exaggerated inflammatory response referred to as the "cytokine storm" that can lead to lethal lung injuries. In the absence of an effective anti-viral molecule and until the formulation of a successful vaccine, anti-inflammatory drugs might offer a complementary tool for controlling the associated complications of COVID-19 and thus decreasing the subsequent fatalities. Drug repurposing for several molecules has emerged as a rapid temporary solution for COVID-19. Among these drugs is Thalidomide; a historically emblematic controversial molecule that harbors an FDA approval for treating erythema nodosum leprosum (ENL) and multiple myeloma (MM). Based on just one-case report that presented positive outcomes in a patient treated amongst others with Thalidomide, two clinical trials on the efficacy and safety of Thalidomide in treating severe respiratory complications in COVID-19 patients were registered. Yet, the absence of substantial evidence on Thalidomide usage in that context along with the discontinued studies on the efficiency of this drug in similar pulmonary diseases, might cause a significant obstacle for carrying out further clinical evaluations. Herein, we will discuss the theoretical effectiveness of Thalidomide in attenuating inflammatory complications that are encountered in COVID-19 patients while pinpointing the lack of the needed evidences to move forward with this drug.
Collapse
Affiliation(s)
- Athar Khalil
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Amina Kamar
- Vascular Medicine Program, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
- Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
17
|
Dastan F, Tabarsi P, Marjani M, Moniri A, Hashemian SMR, Tavakoli-Ardakani M, Saffaei A. Thalidomide against Coronavirus Disease 2019 (COVID-19): A Medicine with a Thousand Faces. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:1-2. [PMID: 32922463 PMCID: PMC7462477 DOI: 10.22037/ijpr.2020.113369.14259] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Farzaneh Dastan
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Majid Marjani
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Moniri
- Virology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Mohammad Reza Hashemian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maria Tavakoli-Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ali Saffaei
- Student Research Committee, Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Yao J, Zhang J, Tai W, Deng S, Li T, Wu W, Pu L, Fan D, Lei W, Zhang T, Dong Z. High-Dose Paraquat Induces Human Bronchial 16HBE Cell Death and Aggravates Acute Lung Intoxication in Mice by Regulating Keap1/p65/Nrf2 Signal Pathway. Inflammation 2019; 42:471-484. [PMID: 30734183 PMCID: PMC6449493 DOI: 10.1007/s10753-018-00956-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Paraquat (PQ) intoxication seriously endangers human beings’ health, however, the underlying mechanisms are still unclear. Here we found that PQ inhibits human bronchial 16HBE cell proliferation and promotes cell apoptosis, necrosis as well as ROS generation in a dose dependent manner. Of note, low-dose PQ (50 μM) induces cell autophagy, increases Nrf2 as well as p65 levels and has little impacts on Keap1, while high-dose PQ (500 μM) inhibits autophagy, upregulates Keap1 as well as downregulates p65 and Nrf2. In addition, we verified that p65 overexpression increases Nrf2 and its downstream targets in 16HBE cells, which are reversed by synergistically knocking down Nrf2. Our further results showed that high-dose PQ’s effects on cell proliferation, apoptosis, ROS levels and autophagy are reversed by p65 overexpression. Besides, the protective effects of overexpressed p65 on high-dose PQ (500 μM) treated 16HBE cells are abrogated by synergistically knocking down Nrf2. In vivo experiments also showed that high-dose PQ promotes inflammatory cytokines secretion, lung fibrosis and cell apoptosis, inhibits cell proliferation in mice models by regulating Keap1/p65/Nrf2 signal pathway. Therefore, we concluded that high-dose PQ (500 μM) inhibits 16HBE cell proliferation and autophagy, promotes cell death and mice lung fibrosis by regulating Keap1/p65/Nrf2 signal pathway.
Collapse
Affiliation(s)
- Jiexiong Yao
- Department of Internal Medicine Ward 5, Guangdong Provincial Corps Hospital of Chinese People's Armed Police Forces, Guangzhou Medical University, Guangzhou, Guangdong, 510507, People's Republic of China
| | - Jihua Zhang
- Department of Pulmonary and Critical Care Medicine, The People Hospital of Yuxi City, Yuxi, China
| | - Wenlin Tai
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road, Kunming, Yunnan, China
| | - Shuhao Deng
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Ting Li
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Wenjuan Wu
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Lin Pu
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Du Fan
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Wen Lei
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Tao Zhang
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Zhaoxing Dong
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China.
| |
Collapse
|
19
|
Tsai YR, Tweedie D, Navas-Enamorado I, Scerba MT, Chang CF, Lai JH, Wu JCC, Chen YH, Kang SJ, Hoffer BJ, de Cabo R, Greig NH, Chiang YH, Chen KY. Pomalidomide Reduces Ischemic Brain Injury in Rodents. Cell Transplant 2019; 28:439-450. [PMID: 31094216 PMCID: PMC6628558 DOI: 10.1177/0963689719850078] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Stroke is a leading cause of death and severe disability worldwide. After cerebral
ischemia, inflammation plays a central role in the development of permanent neurological
damage. Reactive oxygen species (ROS) are involved in the mechanism of post-ischemic
inflammation. The activation of several inflammatory enzymes produces ROS, which
subsequently suppress mitochondrial activity, leading to further tissue damage.
Pomalidomide (POM) is a clinically available immunomodulatory and anti-inflammatory agent.
Prior cellular studies demonstrate that POM can mitigate oxidative stress and lower levels
of pro-inflammatory cytokines, particularly TNF-α, which plays a prominent role in
ischemic stroke-induced brain damage and functional deficits. To evaluate the potential
value of POM in cerebral ischemia, POM was initially administered to transgenic mice
chronically over-expressing TNF-α surfactant protein (SP)-C promoter (SP-C/TNF-α mice) to
assess whether systemically administered drug could lower systemic TNF-α level. POM
significantly lowered serum levels of TNF-α and IL-5. Pharmacokinetic studies were then
undertaken in mice to evaluate brain POM levels following systemic drug administration.
POM possessed a brain/plasma concentration ratio of 0.71. Finally, rats were subjected to
transient middle cerebral artery occlusion (MCAo) for 60 min, and subsequently treated
with POM 30 min thereafter to evaluate action on cerebral ischemia. POM reduced the
cerebral infarct volume in MCAo-challenged rats and improved motor activity, as evaluated
by the elevated body swing test. POM’s neuroprotective actions on ischemic injury
represent a potential therapeutic approach for ischemic brain damage and related
disorders, and warrant further evaluation.
Collapse
Affiliation(s)
- Yan-Rou Tsai
- 1 The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei.,2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei
| | - David Tweedie
- 3 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ignacio Navas-Enamorado
- 3 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michael T Scerba
- 3 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Cheng-Fu Chang
- 2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,4 Department of Neurosurgery, Taipei City Hospital, Zhongxiao Branch, Taipei.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei
| | - Jing-Huei Lai
- 2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei
| | - John Chung-Che Wu
- 2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei.,6 Department of Neurosurgery, Taipei Medical University Hospital, Taipei
| | - Yen-Hua Chen
- 2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei
| | - Shuo-Jhen Kang
- 2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei
| | - Barry J Hoffer
- 2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,7 Department of Neurosurgery, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Rafael de Cabo
- 3 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nigel H Greig
- 3 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yung-Hsiao Chiang
- 1 The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei.,2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei.,6 Department of Neurosurgery, Taipei Medical University Hospital, Taipei
| | - Kai-Yun Chen
- 1 The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei.,2 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei
| |
Collapse
|
20
|
Liu B, Bing Q, Li S, Han B, Lu J, Baiyun R, Zhang X, Lv Y, Wu H, Zhang Z. Role of A 2B adenosine receptor-dependent adenosine signaling in multi-walled carbon nanotube-triggered lung fibrosis in mice. J Nanobiotechnology 2019; 17:45. [PMID: 30922349 PMCID: PMC6440149 DOI: 10.1186/s12951-019-0478-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/15/2019] [Indexed: 12/13/2022] Open
Abstract
Background Multi-walled carbon nanotube (MWCNT)-induced lung fibrosis leads to health concerns in human. However, the mechanisms underlying fibrosis pathogenesis remains unclear. The adenosine (ADO) is produced in response to injury and serves a detrimental role in lung fibrosis. In this study, we aimed to explore the ADO signaling in the progression of lung fibrosis induced by MWCNT. Results MWCNT exposure markedly increased A2B adenosine receptor (A2BAR) expression in the lungs and ADO level in bronchoalveolar lavage fluid, combined with elevation of blood neutrophils, collagen fiber deposition, and activation of myeloperoxidase (MPO) activity in the lungs. Furthermore, MWCNT exposure elicited an activation of transforming growth factor (TGF)-β1 and follistatin-like 1 (Fstl1), leading to fibroblasts recruitment and differentiation into myofibroblasts in the lungs in an A2BAR-dependent manner. Conversely, treatment of the selective A2BAR antagonist CVT-6883 exhibited a significant reduction in levels of fibrosis mediators and efficiently decreased cytotoxicity and inflammatory in MWCNT treated mice. Conclusion Our results reveal that accumulation of extracellular ADO promotes the process of the fibroblast-to-myofibroblast transition via A2BAR/TGF-β1/Fstl1 signaling in MWCNT-induced lung fibrosis.
Collapse
Affiliation(s)
- Biying Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China
| | - Qizheng Bing
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China
| | - Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Jingjing Lu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Ruiqi Baiyun
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Xiaoya Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Yueying Lv
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Hao Wu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China. .,Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin, 150030, China.
| |
Collapse
|
21
|
Liu H, Wu Q, Chu T, Mo Y, Cai S, Chen M, Zhu G. High-dose acute exposure of paraquat induces injuries of swim bladder, gastrointestinal tract and liver via neutrophil-mediated ROS in zebrafish and their relevance for human health risk assessment. CHEMOSPHERE 2018; 205:662-673. [PMID: 29723724 DOI: 10.1016/j.chemosphere.2018.04.151] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 06/08/2023]
Abstract
The exact toxicological mechanisms of paraquat (PQ) poisoning are not entirely clear, especially on the high-level acute exposure. To assess the health risk of PQ, especially to suicidal individuals, accidental ingestion eaters, occupational groups, and special multitude, firstly we explored the acute toxic effect and the possible mechanisms of high-level exposure of PQ using zebrafish. The mainly target organs of PQ were swim bladder which is the homolog of the mammalian lung, followed by gastrointestinal tract and liver. Morphological malformations which were further defined by histopathologic examination include smaller size, fibrosis and inflammatory cell invasion for swim bladder; irregularly arranged or dissolved epithelial folds, loss of villous architecture, and ecclasis of mucosal cells in a smaller lumen for gastrointestinal tract; as well as smaller size, degeneration, fibrous proliferation, atrophy for liver. In addition, PQ enhanced leukocyte recruitment (neutrophil migrated first, followed by macrophage) into swim bladder and induced ROS which can be scavenged by glutathione. Moreover, qRT-PCR results showed that PQ increased the expression level of genes involved in the inflammatory response, such as L-1β, IL-6, IL-8, TNF-α, TNF-β, IFN-1, TGF-β, and NF-kB. For the first time, our results demonstrated that acute exposure of PQ induced pulmonary toxicity which was followed by gastrointestinal and hepatic toxicity via neutrophil-mediated ROS in zebrafish. In summary, these findings generated here will contribute to our better understanding of characteristics of PQ acute poisoning and can provide valuable information on better PQ poisoning treatments, occupational disease prevention, and providing theoretical foundation for risk management measures.
Collapse
Affiliation(s)
- Hongcui Liu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Qiong Wu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Tianyi Chu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Yinyuan Mo
- Department of Pharmacology/Toxicology and Cancer Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | - Shuyang Cai
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mengli Chen
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China.
| | - Guonian Zhu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
22
|
Fan H, Huang H, Hu L, Zhu W, Yu Y, Lou J, Hu L, Chen F. The activation of STIM1 mediates S-phase arrest and cell death in paraquat induced acute lung intoxication. Toxicol Lett 2018; 292:123-135. [DOI: 10.1016/j.toxlet.2018.04.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/18/2018] [Accepted: 04/24/2018] [Indexed: 12/11/2022]
|
23
|
Wang Y, Wu H, Niu W, Chen J, Liu M, Sun X, Li Z. Tanshinone IIA attenuates paraquat‑induced acute lung injury by modulating angiotensin‑converting enzyme 2/angiotensin‑(1‑7) in rats. Mol Med Rep 2018; 18:2955-2962. [PMID: 30015919 DOI: 10.3892/mmr.2018.9281] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/20/2018] [Indexed: 11/06/2022] Open
Abstract
Tanshinone IIA (TIIA) is an active compound that can be isolated from the Chinese herb, Salvia miltiorrhizae Bunge, also known as danshen. Previous studies have demonstrated that TIIA can effectively attenuate bleomycin‑induced pulmonary fibrosis in rats. However, it has not been determined whether TIIA can attenuate paraquat (PQ)‑induced acute lung injury (ALI). In the present study, the protective effects exhibited by TIIA on PQ‑induced ALI, as well as its underlying mechanisms, were investigated using Sprague‑Dawley (SD) rats. ALI animal models using rats were established via administration of PQ. Adult male SD rats were randomly divided into three groups: A control group, a PQ group and a PQ + TIIA group. Total cell count, total protein levels and lactic dehydrogenase (LDH) levels in bronchoalveolar lavage fluid (BALF), as well as myeloperoxidase (MPO) activity in lung tissues were determined. Lung histological alterations were also investigated. Angiotensin converting enzyme 2 (ACE2) and Angiotensin 1‑7 [Ang‑(1‑7)] expression levels in the lung were also analyzed. The results demonstrated that administration of PQ induced marked histological alterations, and markedly increased neutrophil infiltration, lung wet/dry weight ratio, total cell count, protein content and LDH levels in BALF. In addition, PQ was revealed to significantly decrease ACE2 and Ang‑(1‑7) expression levels in lung tissues. However, it was demonstrated that TIIA attenuated these effects. Therefore, the results of the present study suggest that that TIIA may exhibit a therapeutic effect regarding PQ‑induced ALI in rats, and that ACE2 and Ang‑(1‑7) may be involved in the underlying mechanisms of this effect.
Collapse
Affiliation(s)
- Yanxia Wang
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Huajie Wu
- Department of Pediatrics of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wen Niu
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jian Chen
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Manlin Liu
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xin Sun
- Department of Pediatrics of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhichao Li
- Department of Pathology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
24
|
Dong X, Li X, Li M, Chen M, Fan Q, Wei W. Antiinflammation and Antioxidant Effects of Thalidomide on Pulmonary Fibrosis in Mice and Human Lung Fibroblasts. Inflammation 2018; 40:1836-1846. [PMID: 28730510 DOI: 10.1007/s10753-017-0625-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this study, the potential effects of thalidomide (Thal) on bleomycin (BLM)-induced pulmonary fibrosis were investigated. BALB/C mice model of pulmonary fibrosis induced by an intratracheal instillation of BLM was adopted, and then was intraperitoneally injected with Thal (10, 20, 50 mg/kg) daily for 8 days, while the control and BLM-treated mouse groups were injected with a saline solution. The effects of Thal on pulmonary injury were evaluated by the lung wet/dry weight ratios and histopathological examination. Inflammation of lung tissues was assessed by measuring the levels of interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and transforming growth factor (TGF)-β in bronchoalveolar lavage fluid. Oxidative stress was evaluated by detecting the levels of reactive oxygen species (ROS), superoxide dismutase (SOD), total antioxidant capacity (T-AOC), and malondialdehyde (MDA) in lung tissue. The results indicated that Thal treatment remarkably attenuated pulmonary fibrosis, oxidative stress, and inflammation in mouse lungs. The antiinflammatory and antioxidant effects of Thal were also found in human lung fibroblasts. Thal administration significantly enhanced the activity of thioredoxin reductase; however, the other enzymes or proteins involved in biologic oxidation-reduction equilibrium were not affected. Our findings indicate that Thal-mediated suppression of pulmonary fibrosis is related to the inhibition of oxidative stress and inflammatory response. In summary, these results may provide a rationale to explore clinical application of Thal for the prevention of pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiaoying Dong
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Xin Li
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Minghui Li
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Ming Chen
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Qian Fan
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Wei Wei
- Department of Rheumatism and Immunity, General Hospital of Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
25
|
Abass AO, Kamel NN, Khalifa WH, Gouda GF, El-Manylawi MAF, Mehaisen GMK, Mashaly MM. Propolis supplementation attenuates the negative effects of oxidative stress induced by paraquat injection on productive performance and immune function in turkey poults. Poult Sci 2018; 96:4419-4429. [PMID: 29053856 PMCID: PMC7107162 DOI: 10.3382/ps/pex248] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 08/09/2017] [Indexed: 01/16/2023] Open
Abstract
Paraquat (PQ) is used as a herbicide in agriculture and causes oxidative and inflammatory damage to animal tissues. The current study was conducted to investigate the positive effects of dietary propolis (PR), as a potent naturally produced antioxidant, on growth performance and immune function of turkey poults exposed to oxidative stress induced by PQ injection. Native male turkey poults (n = 120, 49-d-old) were randomly assigned into 4 groups: poults received a basal diet with a daily subcutaneous PQ injection of 5 mg/kg BW for 7 consecutive days (PQ group), an experimental diet containing 1 g/kg PR with a daily subcutaneous PQ injection for 7 days (PR+PQ group), or received the experimental PR diet with a daily subcutaneous injection of 0.5 mL sterile saline for 7 days (PR group); while the control poults received a basal diet with a daily subcutaneous saline injection for 7 consecutive days (C group). The productive performance in the PQ group showed a significant (P < 0.05) reduction in the weight gain (WG) and feed intake (FI), and impaired feed conversion ratio (FCR). Propolis supplementation in the PR+PQ group significantly ameliorated the PQ effects on WG and FCR. Turkey poults of the PQ and PR+PQ groups had a significant augmentation in the blood malondialdehyde (MDA), tumor necrosis factor-α (TNFα), and corticosterone levels, and in contrast, a significant reduction in the triiodothyronine (T3), when compared to the C group. While propolis significantly reduced the MDA and corticosterone, and increased the T3 levels in the PR+PQ group compared to the PQ group. Furthermore, the dietary PR supplementation significantly limited the PQ-suppressive effects on cell- and humoral-mediated immunity and lymphocyte proliferation of turkey poults. In addition, propolis supplementation in the PR and PR+PQ groups markedly reversed the PQ-induced DNA fragmentation and heat shock protein 70 (Hsp70) over-expression in blood cells. It can be concluded that PR could improve turkey immunity and performance, particularly under inflammation and oxidative stress induced by PQ exposure.
Collapse
Affiliation(s)
- Ahmed O Abass
- Poultry Cellular and Molecular Physiology Laboratory, Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Nancy N Kamel
- Animal Production Department, National Research Centre, Dokki, Giza, Egypt
| | - Walaa H Khalifa
- Animal Production Department, National Research Centre, Dokki, Giza, Egypt
| | - G F Gouda
- Animal Breeding Section, Department of Animal Production, Faculty of Agriculture, Ain Shams University, Shoubra Al-Kheima, Cairo, Egypt
| | - M A F El-Manylawi
- Poultry Nutrition Section, Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Gamal M K Mehaisen
- Poultry Cellular and Molecular Physiology Laboratory, Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Magdi M Mashaly
- Poultry Cellular and Molecular Physiology Laboratory, Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| |
Collapse
|
26
|
Yang W, Liu W, Yu W, Fei D, Meng X, Yang S, Meng S, Zhao M. Angptl2 deficiency attenuates paraquat (PQ)-induced lung injury in mice by altering inflammation, oxidative stress and fibrosis through NF-κB pathway. Biochem Biophys Res Commun 2018; 503:94-101. [PMID: 29852175 DOI: 10.1016/j.bbrc.2018.05.186] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/28/2018] [Indexed: 12/13/2022]
Abstract
Paraquat (PQ) is one of the most extensively used herbicides, possessing high toxicity for humans and animals. The lung is the main target organ by the poisoning of PQ resulting in acute lung injury. Nonetheless, molecular mechanisms underlying PQ-induced lung injury remain unclear. Here, we ask if angiopoietin-like protein 2 (Angptl2), a pro-inflammatory protein, contributes to inflammation that accelerates acute lung injury. The results indicated that abundant Angptl2 expression was observed in lung tissues of PQ-treated mice. Histological analysis revealed that PQ-induced histological changes were alleviated by Angptl2 knockout (Angptl2-/-). Angptl2-/- in PQ-treated mice attenuated acute lung injury progression by reducing the number of total cells, total leukocytes, neutrophils and macrophages in bronchoalveolar lavage fluid (BALF) and reducing inflammatory response through the inactivation of nuclear factor kappa B (NF-κB) pathway. Angptl2-/- reduced oxidative stress in PQ-treated mice, as evidenced by the enhanced superoxide dismutase (SOD) activity and reduced malondialdehyde (MDA) levels in serum or lung tissue samples, which was accompanied with increased expressions of nuclear respiratory factor 2 (Nrf-2), heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase 1 (NQO-1). PQ-induced fibrosis was also improved in Angptl2-/- mice by decreasing pulmonary transforming growth factor (TGF)-β1 expressions. In vitro, we found that Angptl2 knockdown-suppressed inflammation, oxidative stress and fibrosis was restored by increasing NF-κB activation in PQ-incubated A549 cells; however, the results above were significantly reversed by inactivating NF-κB using its inhibitor, Bay 11-7085 or LY2409881. Therefore, Angptl2 could provide therapeutic effects on PQ-induced acute lung injury through inhibiting inflammation, oxidative stress and fibrosis by regulating NF-κB pathway.
Collapse
Affiliation(s)
- Wei Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China
| | - Wen Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China; Department of General Surgery, Xinxiang Medical University, No. 601, New Yan Road, Xinxiang 453000, China
| | - Wei Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China
| | - Dongsheng Fei
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China
| | - Xianglin Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China
| | - Songlin Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China
| | - Shishuai Meng
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23, YouZheng Street, Nangang District, Harbin, 150001, China.
| |
Collapse
|
27
|
Therapeutic Effect of Gallic Acid Against Paraquat-Induced Lung Injury in Rats. Jundishapur J Nat Pharm Prod 2018. [DOI: 10.5812/jjnpp.12450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
28
|
Thalidomide Inhibits TGF-β1-induced Epithelial to Mesenchymal Transition in Alveolar Epithelial Cells via Smad-Dependent and Smad-Independent Signaling Pathways. Sci Rep 2017; 7:14727. [PMID: 29116196 PMCID: PMC5677010 DOI: 10.1038/s41598-017-15239-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/24/2017] [Indexed: 12/18/2022] Open
Abstract
Recent evidence indicates that the epithelial to mesenchymal transition (EMT) in primary alveolar cells (AECs) plays an important role in idiopathic pulmonary fibrosis (IPF). In vivo models have suggested that thalidomide (THL) has anti-fibrotic effects against pulmonary fibrosis, but the underlying mechanism of this effect is not clear. This study investigated whether THL regulates alveolar EMT and the possible mechanisms underlying this process. CCL-149 cells were treated with TGF-β1 in the presence of THL at the indicated concentrations. EMT was assessed by changes in cell morphology and in phenotypic markers. Signaling pathways involved in EMT were characterized by western blot analysis. THL inhibited the TGF-β1 induction of α-SMA, vimentin, MMP-2/-9 and collagen type IV expression and restored the morphological changes in primary alveolar epithelial cells caused by TGF-β1. TGF-β1 induction of α-SMA expression was partially dependent on the activation of p38, JNK, ERK, Akt, Smad 2 and Smad3. Moreover, THL inhibited TGF-β1-induced phosphorylation of p38, JNK, ERK, Akt, GSK3β, Smad 2 and Smad3 without altering the total expression levels of those proteins. These findings indicate that TGF-β1-induced EMT in alveolar epithelial cells is inhibited by THL via both Smad-dependent and non-Smad-dependent signaling pathways and suggests therapeutic approaches for targeting this process in pulmonary fibrosis.
Collapse
|
29
|
Mojiri-Forushani H, Hemmati AA, Dehghani MA, Malayeri AR, Pour HH. Effects of herbal extracts and compounds and pharmacological agents on pulmonary fibrosis in animal models: a review. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2017; 15:433-441. [DOI: 10.1016/s2095-4964(17)60363-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
30
|
Study of the Interactions of Bovine Serum Albumin with the New Anti-Inflammatory Agent 4-(1,3-Dioxo-1,3-dihydro-2H-isoindol-2-yl)-N'-[(4-ethoxy-phenyl)methylidene]benzohydrazide Using a Multi-Spectroscopic Approach and Molecular Docking. Molecules 2017; 22:molecules22081258. [PMID: 28749443 PMCID: PMC6152112 DOI: 10.3390/molecules22081258] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/15/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022] Open
Abstract
The lipophilic derivative of thalidomide (4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-N′-[(4-ethoxyphenyl)methylidene]benzohydrazide, 6P) was synthesized to enhance its characteristics and efficacy. Earlier studies have proved the immunomodulatory and anti-inflammatory effects of 6P. In this study the interaction between bovine serum albumin (BSA) and 6P was studied using a multi-spectroscopic approach which included UV spectrophotometry, spectrofluorimetry and three dimensional spectrofluorometric and molecular docking studies. Static quenching was involved in quenching the fluorescence of BSA by 6P, because a complex formation occurred between the 6P and BSA. The binding constant decreased with higher temperature and was in the range of 2.5 × 105–4.8 × 103 L mol−1 suggesting an unstable complex at higher temperatures. A single binding site was observed and the the site probe experiments showed site II (sub-domain IIIA) of BSA as the binding site for 6P. The negative values of ∆G0, ∆H0 and ∆S0 at (298/303/308 K) indicated spontaneous binding between 6P and BSA as well as the interaction was enthalpy driven and van der Waals forces and hydrogen bonding were involved in the interaction. The docking results and the results from the experimental studies are complimentary to each other and confirm that 6P binds at site II (sub-domain IIIA) of BSA.
Collapse
|
31
|
Effects of hemoperfusion and continuous renal replacement therapy on patient survival following paraquat poisoning. PLoS One 2017; 12:e0181207. [PMID: 28704509 PMCID: PMC5509301 DOI: 10.1371/journal.pone.0181207] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Mortality in patients with paraquat (PQ) poisoning is related to plasma PQ levels. Concentrations lower than 5,000 ng/mL are considered critical but curable. This study assessed the effects of hemoperfusion (HP) and continuous renal replacement therapy (CRRT) on the survival of PQ-poisoned patients with plasma PQ levels below 5,000ng/mL. We analyzed the records of 164 patients with PQ poisoning who were treated at the First Affiliated Hospital of Wenzhou Medical University in China between January 2011 and May 2015. We divided these patients into six sub-groups based on baseline plasma PQ levels and treatment, compared their clinical characteristics, and analyzed their survival rates. Patient sub-groups did not differ in terms of age, sex, time between poisoning and hospital admission, or time to first gavage. Biochemical indicators improved over time in all sub-groups following treatment, and the combined HP and CRRT treatment yielded better results than HP or CRRT alone. Fatality rates in the three treatment sub-groups did not differ among patients with baseline plasma PQ levels of 50–1,000 ng/mL, but in patients with 1,000–5,000 ng/mL levels, the mortality rate was 59.2% (HP treatment group), 48% (CRRT treatment group), and 37.9% (combined treatment group). Mortality rates were higher 10–30 days after hospitalization than in the first 10 days after admission. In the early stages of PQ poisoning, CRRT is effective in reducing patient fatality rates, particularly when combined with HP. Our data could be useful in increasing survival in acute PQ poisoning patients.
Collapse
|
32
|
Iqbal M, Bhat MA, Raish M, Ezzeldin E. Determination of (4-(1,3-dioxo-1,3-dihydro-2H-isoindol- 2-yl)-N'-[(4-ethoxyphenyl) methylidene] benzohydrazide, a novel anti-inflammatory agent, in biological fluids by UPLC-MS/MS: Assay development, validation and in vitro metabolic stability study. Biomed Chromatogr 2017; 31. [PMID: 28370149 DOI: 10.1002/bmc.3981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/24/2017] [Accepted: 03/27/2017] [Indexed: 11/11/2022]
Abstract
A thalidomide analog, (4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-N'-[(4-ethoxyphenyl) methylidene] benzohydrazide), has been identified as a promising broad-spectrum anti-inflammatory agent in previous study. In this study, a sensitive and selective UPLC-MS/MS assay was developed and validated for its determination in rat plasma samples. The chromatographic separation was performed on an Aquity BEH C18 column using mobile phase comprising of acetonitrile and 10 mm ammonium acetate in the ratio of 85: 15, at flow rate of 0.3 mL/min. The detection and quantification were performed in positive multiple reaction monitoring mode by parent to daughter ion transition of 414.06 ˃ 148.05 for analyte and 411.18 ˃ 191.07 for internal standard (risperidone), respectively using electrospray ionization source. The sample extraction process consisted of liquid-liquid extraction method using diethyl ether as the extracting solvent. The assay was validated by following FDA guidelines and all parameters were found to be within acceptable limits. The linearity was between 10.1 and 2500 ng/mL and the lower limit of quantification was 10.1 ng/mL. The reported results indicate that the assay could meet the requirement for analysis of this compound in amounts expected to the present in actual samples. Further, in vitro metabolic stability study was performed in rat liver microsomes by using the validated assay.
Collapse
Affiliation(s)
- Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Bioavailability Laboratory, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mashooq A Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Essam Ezzeldin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Bioavailability Laboratory, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
33
|
Satpute RM, Pawar PP, Puttewar S, Sawale SD, Ambhore PD. Effect of resveratrol and tetracycline on the subacute paraquat toxicity in mice. Hum Exp Toxicol 2017; 36:1303-1314. [PMID: 28090784 DOI: 10.1177/0960327116688070] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Paraquat (PQ) is a nonselective bipyridyl herbicide widely used in agriculture to control weeds, but its accidental, occupational, or intentional exposure in humans is known to cause pneumo- and neurotoxicity which may proves fatal. Oxidative stress is reported as an underlined mechanism of PQ-induced toxicity in alveolar cells, neurons, and astroglia. PQ generates superoxides both through electron transport reaction (ETC) with nicotinamide adenine dinucleotide-dependent oxidoreductase and by the redox cycling via reaction with molecular oxygen. In lungs, it causes edema and inflammation resulting in neutrophils infiltration and subsequent activation of pro-inflammatory cytokines. In the present study, toxicity of subacute oral PQ exposure and effect of resveratrol (Res) and/or tetracycline (TC) on oxidative stress and inflammatory markers in lungs, brain, and liver was studied. Levels of glutathione and malondialdehyde and activities of myeloperoxidase, glutathione peroxidase, and catalase were measured in lungs, brain, and liver. PQ interferes in the function of mitochondrial ETC complexes causing decreased adenosine triphosphate levels, and hence the activities of complexes I and IV were studied in brain tissues. Res, a natural antioxidant, and TC, an antibiotic with its antimicrobial and anti-inflammatory properties, offered significant protection from severe oxidative stress and inflammation and ameliorated the general well-being of mice against the toxic outcome of PQ.
Collapse
Affiliation(s)
- R M Satpute
- 1 Toxicology Laboratory, Defence Research and Development Establishment, Nagpur, Maharashtra, India
| | - P P Pawar
- 2 Department of Biotechnology, S.F.S. College, Seminari Hills, Nagpur, India
| | - S Puttewar
- 2 Department of Biotechnology, S.F.S. College, Seminari Hills, Nagpur, India
| | - S D Sawale
- 3 Department of Biotechnology, G.H. Raisoni Institute of Information Technology, Nagpur, India
| | - P D Ambhore
- 4 Department of Biotechnology, Rajarshi Shahu Mahavidyalaya, Latur, India
| |
Collapse
|
34
|
Sengupta A, Manna K, Datta S, Das U, Biswas S, Chakrabarti N, Dey S. Herbicide exposure induces apoptosis, inflammation, immune modulation and suppression of cell survival mechanism in murine model. RSC Adv 2017. [DOI: 10.1039/c6ra27883c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The present study demonstrates paraquat induced cellular toxicity in spleen and associated ROS generation, mitochondria dependent cellular apoptosis, inflammation and splenomegaly inSwiss Albinomice.
Collapse
Affiliation(s)
- Aaveri Sengupta
- Department of Physiology
- DST-PURSE & UGC-CPEPA Supported Department
- Centre for Research in Nanoscience & Nanotechnology (CRNN)
- University of Calcutta
- Kolkata – 700009
| | - Krishnendu Manna
- Department of Physiology
- DST-PURSE & UGC-CPEPA Supported Department
- Centre for Research in Nanoscience & Nanotechnology (CRNN)
- University of Calcutta
- Kolkata – 700009
| | - Siddhartha Datta
- Department of Physiology
- DST-PURSE & UGC-CPEPA Supported Department
- Centre for Research in Nanoscience & Nanotechnology (CRNN)
- University of Calcutta
- Kolkata – 700009
| | - Ujjal Das
- Department of Physiology
- DST-PURSE & UGC-CPEPA Supported Department
- Centre for Research in Nanoscience & Nanotechnology (CRNN)
- University of Calcutta
- Kolkata – 700009
| | - Sushobhan Biswas
- Department of Physiology
- DST-PURSE & UGC-CPEPA Supported Department
- Centre for Research in Nanoscience & Nanotechnology (CRNN)
- University of Calcutta
- Kolkata – 700009
| | - Nilkanta Chakrabarti
- Department of Physiology
- DST-PURSE & UGC-CPEPA Supported Department
- Centre for Research in Nanoscience & Nanotechnology (CRNN)
- University of Calcutta
- Kolkata – 700009
| | - Sanjit Dey
- Department of Physiology
- DST-PURSE & UGC-CPEPA Supported Department
- Centre for Research in Nanoscience & Nanotechnology (CRNN)
- University of Calcutta
- Kolkata – 700009
| |
Collapse
|
35
|
Alizadeh-Tabrizi N, Malekinejad H, Varasteh S, Cheraghi H. Atorvastatin protected from paraquat-induced cytotoxicity in alveolar macrophages via down-regulation of TLR-4. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 49:8-13. [PMID: 27883937 DOI: 10.1016/j.etap.2016.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 06/06/2023]
Abstract
The current study designed to clarify the mechanism of paraquat-induced cytotoxicity and protective effects of Atorvastatin on freshly isolated alveolar macrophages (AMs). AMs were collected via bronchoalveolar lavage and exposed to various concentrations of paraquat in the presence and absence of atorvastatin for 24h. Cell viability, myeloperoxidase activity; nitric oxide generation and total antioxidant capacity were assessed. Expression of TLR-4 at mRNA and protein levels were studied by using PCR and western blot methods Atorvastatin enhanced the paraquat-reduced cell viability and reduced the paraquat-induced myeloperoxidase activity and nitric oxide production. Moreover, atorvastatin down-regulated by 60% the paraquat up-regulated expression of TLR-4 at protein and mRNA level. Our results suggest that, AMs in vitro model could be a novel cytological tool for studies on paraquat poisoning and therapy regimens. Additionally, atorvastatin cytoprotective effects on paraquat-induced cytotoxicity partly attribute to its anti-myeloperoxidase, antioxidant properties, which might be regulated via TLR-4 expression.
Collapse
Affiliation(s)
- Nazli Alizadeh-Tabrizi
- Department of Pharmacology & Toxicology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Hassan Malekinejad
- Department of Pharmacology & Toxicology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia Medical University of Sciences, Urmia, Iran.
| | - Soheil Varasteh
- Faculty of Science, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Hadi Cheraghi
- Department of Pharmacology & Toxicology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
36
|
Amirshahrokhi K, Khalili AR. Carvedilol attenuates paraquat-induced lung injury by inhibition of proinflammatory cytokines, chemokine MCP-1, NF-κB activation and oxidative stress mediators. Cytokine 2016; 88:144-153. [PMID: 27619518 DOI: 10.1016/j.cyto.2016.09.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 11/16/2022]
Abstract
Paraquat is a highly toxic herbicide that selectively accumulates in the lungs and causes pulmonary damage through the oxidative and inflammatory processes. Carvedilol is a nonselective beta and alpha-adrenergic blocking agent that has been shown to possess powerful antioxidant and anti-inflammatory properties. In the present study, we evaluated the protective effects and the underlying mechanisms of carvedilol on paraquat-induced lung injury in a mouse model. Mice were injected with a single dose of paraquat (20mg/kg, ip), and treated with carvedilol (10 and 20mg/kg/day, orally) for eight days. At the end of the experiment, lung tissue and blood samples were collected for histological and biochemical analysis. The results showed that carvedilol treatment improved the histopathological changes in the lung tissue of mice exposed to paraquat. Carvedilol significantly decreased the levels of malondialdehyde (MDA), carbonyl protein, myeloperoxidase (MPO), and nitric oxide (NO), while increased the levels of glutathione (GSH), superoxide dismutase (SOD), catalase and glutathione reductase compared with paraquat group. Carvedilol treatment also significantly reduced the levels of proinflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, transforming growth factor (TGF)-β1 and monocyte chemoattractant protein (MCP)-1 in the lung tissue. Treatment of mice with carvedilol decreased paraquat-induced expression of nuclear factor kappa B (NF-κB). In addition the plasma levels of matrix metalloproteinase (MMP)-9 and the lung hydroxyproline content significantly reduced by carvedilol treatment. Taken together, these results indicate that carvedilol is able to decrease the severity of paraquat-induced lung injury through inhibition of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali-Reza Khalili
- Division of Pathology, Imam Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
37
|
Wang Q, Liu S, Hu D, Wang Z, Wang L, Wu T, Wu Z, Mohan C, Peng A. Identification of apoptosis and macrophage migration events in paraquat-induced oxidative stress using a zebrafish model. Life Sci 2016; 157:116-124. [PMID: 27288846 DOI: 10.1016/j.lfs.2016.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/31/2016] [Accepted: 06/07/2016] [Indexed: 12/30/2022]
Abstract
AIMS Paraquat (PQ) is a pesticide highly toxic to human beings, and a well-known trigger of oxidative stress. Although several animal models of PQ poisoning have been developed, some disadvantages limit their application in vivo. A zebrafish model was used in the present study to better define mechanisms of oxidative stress injury induced by PQ. MAIN METHODS The toxicity of PQ was evaluated in the AB strain of zebrafish, and apoptosis was assessed by acridine orange staining. Macrophage migration was identified using the TG (zlyz:EGFP) transgenic strain, and angiogenesis was observed using the fli1a-EGFP casper strain. Following the validation of gene changes by zebrafish-based in vivo quantitative real-time PCR, network analysis was performed using the Ingenuity Pathway Analysis software. KEY FINDINGS We first established the LC50 of PQ in the zebrafish model, and then found that robust oxidative stress and antioxidant genes were activated after PQ exposure. Moreover, apoptosis and distinct macrophage activation and migration were identified for the first time in PQ-exposed zebrafish. Utilizing this model, both extrinsic and intrinsic pathways involved in PQ-induced apoptosis were elucidated. We further demonstrated that macrophage migration was specifically induced by PQ, and that Rho family members and JNK-MMP13 signaling participated in this process. SIGNIFICANCE Zebrafish is a promising tool for investigating the mechanisms of oxidative stress injury induced by PQ, and for screening effective anti-oxidant drugs.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Shuai Liu
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Dayong Hu
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Zhen Wang
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Ling Wang
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Tianfu Wu
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China; Department of Biomedical Engineering, University of Houston, TX, USA
| | - Zhuanbin Wu
- Shanghai Research Center for Model Organisms, Shanghai, PR China
| | - Chandra Mohan
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China; Department of Biomedical Engineering, University of Houston, TX, USA
| | - Ai Peng
- Department of Nephrology & Rheumatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China.
| |
Collapse
|
38
|
Hong G, Hu L, Tang Y, Zhang T, Kang X, Zhao G, Lu Z. Prognosis and survival analysis of paraquat poisoned patients based on improved HPLC-UV method. J Pharmacol Toxicol Methods 2016; 80:75-81. [PMID: 27216136 DOI: 10.1016/j.vascn.2016.05.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/29/2016] [Accepted: 05/18/2016] [Indexed: 02/08/2023]
Abstract
UNLABELLED Paraquat (PQ) has caused deaths of numerous people around the world. In order to assess the lethal plasma concentration, the patients who acquired acute PQ intoxication were analyzed by plasma concentration monitoring. The plasma PQ concentrations were determined by high performance liquid chromatography (HPLC) which used 5-bromopyrimidine as internal standard and trichloroacetic acid-methanol (1:9) as protein precipitant. The liver, kidney and coagulation function were determined by automatic biochemical analyzer. According to plasma PQ concentration, 90 patients were divided into four groups: trace PQ group (<50ng/mL), low PQ group (<1000ng/mL), medium PQ group (1000-5000ng/mL) and high PQ group (>5000ng/mL). The clinical data from the four groups was statistically analyzed. The results showed the developed HPLC methods exhibited a high degree of accuracy and good linearity within 50-25000ng/mL (R=0.9998). The Spearman's correlation analysis showed PQ concentration had a strong relationship to total bilirubin, direct bilirubin, aspartic transaminase, urea nitrogen, prothrombin time, prothrombin activity, and international normalized ratio (P<0.01). The cured or survival PQ poisoned patients among the trace PQ group, the low PQ group, the medium PQ group, and the high PQ group were 19/19 (100%), 19/21 (90.47%), 11/25 (44.0%), and 0/25 (0%) respectively. The mean hospital days were (10.37±8.04), (18.76±12.06), (16.76±14.44), and (4.04±5.41) days respectively. The Cox regression analysis indicated that plasma PQ concentration was highly related to prognosis (P<0.05). In conclusion, no patient presenting with a PQ concentration over 5000ng/mL survived. The plasma PQ level is related to liver, kidney and coagulation function, which can be used as an important clinical index to judge the prognosis of PQ poisoned patients. CHEMICAL COMPOUNDS Paraquat (PubChem CID: 15938), 5-bromopyrimidine (PubChem CID: 78344), acetonitrile (PubChem CID: 6342), sodium dihydrogen phosphate (PubChem CID: 23672064), sodium heptanesulfonate (PubChem CID: 23672332), methylprednisolone (PubChem CID: 6741), cyclophosphamide (PubChem CID: 2907).
Collapse
Affiliation(s)
- Guangliang Hong
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lufeng Hu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yahui Tang
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Tao Zhang
- Department of Intensive Care Unit, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Xiaowen Kang
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Guangju Zhao
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhongqiu Lu
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
39
|
Amirshahrokhi K, Khalili AR. Gastroprotective effect of 2-mercaptoethane sulfonate against acute gastric mucosal damage induced by ethanol. Int Immunopharmacol 2016; 34:183-188. [PMID: 26967742 DOI: 10.1016/j.intimp.2016.03.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 02/28/2016] [Accepted: 03/04/2016] [Indexed: 01/07/2023]
Abstract
Gastric mucosal damage induced by ethanol is a serious medical problem. Recent evidences suggest that reactive oxygen species and inflammatory mediators play a key role in the destruction of gastric mucosa. The present study was aimed to evaluate the potential beneficial effect of MESNA (2-mercaptoethane sulfonate) against ethanol-induced gastric mucosal damage in mice. The animals were orally pretreated with vehicle or MESNA and then treated with acidified ethanol to induce gastric mucosal damage. One hour after ethanol ingestion mice were euthanized and stomach samples were collected for biochemical analysis. Macroscopic and histopathological evaluation of gastric mucosa showed that pretreatment with MESNA attenuated gastric lesions induced by ethanol. Administration of MESNA significantly increased glutathione content and superoxide dismutase and catalase activity in the gastric tissues. In addition, MESNA markedly reduced ethanol-induced lipid peroxidation, myeloperoxidase activity, tumor necrosis factor-alpha, interleukin (IL)-1β, IL-6, and monocyte chemotactic protein-1 levels. These findings suggest that the thiol-containing compound MESNA is able to decrease alcohol-induced oxidative stress and inflammation in the gastric tissue. It seems that MESNA may have a protective effect against ethanol-induced gastric mucosal damage.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali-Reza Khalili
- Division of Pathology, Imam Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
40
|
Diss L, Dyball S, Ghela T, Golding J, Morris R, Robinson S, Tucker R, Walter T, Young P, Allen M, Fidalgo S, Gard P, Mabley J, Patel B, Chatterjee P, Yeoman M. Acute paraquat exposure impairs colonic motility by selectively attenuating nitrergic signalling in the mouse. Auton Neurosci 2016; 195:8-15. [PMID: 26853977 DOI: 10.1016/j.autneu.2016.01.005] [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: 09/03/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 01/06/2023]
Abstract
Paraquat, a common herbicide, is responsible for large numbers of deaths worldwide through both deliberate and accidental ingestion. Previous studies have eluded that the bioavailability of paraquat increases substantially with increasing dose and that these changes may in part be due to the effects that these high concentrations have on the gastrointestinal tract (GI tract). To date, the actions of acute, high concentrations (20mM for 60 min) of paraquat on the GI tract, particularly the colon which is a major site of paraquat absorption, are unknown. This study examined the effects of acute paraquat administration on colonic motility in the C57BL/6 mouse. Acute paraquat exposure decreased colonic motility and the amplitude of colonic migrating motor complexes (CMMCs), which are major motor patterns involved in faecal pellet propulsion. In isolated segments of distal colon, paraquat increased resting tension and markedly attenuated electrical field stimulation-evoked relaxations. Pharmacological dissection of paraquat's mechanism of action on both the CMMCs and field stimulated tissue using the nitric oxide synthase inhibitor NG-nitro-L-arginine and direct measurement of NO release from the myenteric plexus, demonstrated that paraquat selectively attenuates nitrergic signalling pathways. These changes did not appear to be due to alterations in colonic oxidative stress, inflammation or complex 1 activity, but were most likely caused by paraquat's ability to act as a redox couple. In summary, these data demonstrate that acute paraquat exposure attenuates colonic transit. These changes may facilitate the absorption of paraquat into the circulation and so facilitate its toxicity.
Collapse
Affiliation(s)
- Lucy Diss
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Sarah Dyball
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Tina Ghela
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Jonathan Golding
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Rachel Morris
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Stephen Robinson
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Rosemary Tucker
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Talia Walter
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Paul Young
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Marcus Allen
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Sara Fidalgo
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Paul Gard
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Jon Mabley
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Bhavik Patel
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Prabal Chatterjee
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK
| | - Mark Yeoman
- School of Pharmacy and Biomolecular Sciences, Huxley Building, College of Life, Health and Physical Sciences, University of Brighton, Brighton, East Sussex BN2 4GJ, UK.
| |
Collapse
|
41
|
Bhat MA, Al-Omar MA, Ansari MA, Zoheir KMA, Imam F, Attia SM, Bakheet SA, Nadeem A, Korashy HM, Voronkov A, Berishvili V, Ahmad SF. Design and Synthesis of N-Arylphthalimides as Inhibitors of Glucocorticoid-Induced TNF Receptor-Related Protein, Proinflammatory Mediators, and Cytokines in Carrageenan-Induced Lung Inflammation. J Med Chem 2015; 58:8850-67. [PMID: 26456029 DOI: 10.1021/acs.jmedchem.5b00934] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
N-Arylphthalimides (1-10P) derived from thalidomide by insertion of hydrophobic groups were evaluated for anti-inflammatory activity, and (4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)-N'-[(4-ethoxyphenyl)methylidene]benzohydrazide 6P was identified as a promising anti-inflammatory agent. Further testing confirmed that compared with the control, 6P treatment resulted in a considerable decrease in CD4(+), NF-κB p65(+), TNF-α(+), IL-6(+), GITR(+), and IL-17(+) cell populations and an increase in the Foxp3(+), CD4(+)Foxp3(+), and IκBα(+) populations in whole blood and pleural fluid of a mouse model of lung inflammation. Moreover, treatment with compound 6P decreased the proteins associated with inflammation including TNF-α, IL-6, IL-17, GITR, NF-κB, COX-2, STAT-3, and iNOS and increased the anti-inflammatory mediators such as IL-10 and IL-4. Further, histopathological examination confirmed the potent anti-inflammatory effects of compound 6P. Thus, the N-arylphthalimide derivative 6P acts as a potent anti-inflammatory agent in the carrageenan-induced lung inflammation model, suggesting that this compound may be useful for the treatment of inflammation in a clinical setting.
Collapse
Affiliation(s)
- Mashooq A Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Mohamed A Al-Omar
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Khairy M A Zoheir
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Hesham M Korashy
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| | - Andrey Voronkov
- Department of Chemistry, Lomonosov Moscow State University , Leninskie Gory, 1/3, Moscow 119991, Russia
- Digital Bio Pharm Ltd. , 145-157 St. John Street, London, EC1V 4PW, U.K
- Moscow Institute of Physics and Technology (State University) , 9 Institutskiy Lane, Dolgoprudny, Moscow Oblast 141700, Russia
| | - Vladimir Berishvili
- Department of Chemistry, Lomonosov Moscow State University , Leninskie Gory, 1/3, Moscow 119991, Russia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University , 2457 Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
42
|
Protective effects of thalidomide on pulmonary injuries in a rat model of paraquat intoxication. JOURNAL OF INFLAMMATION-LONDON 2015. [PMID: 26221080 PMCID: PMC4517355 DOI: 10.1186/s12950-015-0093-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Background This study was designed to evaluate the protective effects of thalidomide on paraquat (PQ)-induced lung injuries in a rat model and to explore the underlying mechanisms. Methods Rats were exposed to 50 mg/kg PQ by oral gavage, and treated with thalidomide through oral administration at 60 mg/kg once a day, 6 days/week for 2 weeks. Serum levels of IL-6, TNF-alpha, TGFbeta1 and COL1A1 were detected at different time points after paraquat exposure. At the end of the study, lung tissues were collected for pathological inspection as well as analyses of water content and expression levels of IL-6, TNF-alpha, TGFbeta1 and COL1A1 mRNA. Results The results showed that thalidomide treatment could significantly alleviate PQ-induced pathological changes in lung tissue and severity of lung edema. Thalidomide treatment after PQ exposure resulted in significantly reduced serum levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1, as compared to PQ group. PCR analysis demonstrated that expression levels of IL-6, TNF-alpha, TGF-beta1 and COL1A1 in lung tissue were significantly increased after PQ exposure but reduced by thalidomide, which were confirmed by immunohistochemistry staining. Conclusions Our results indicated that inflammatory factors played important roles in PQ-induced lung injuries and thalidomide could protect rats from PQ-induced lung injuries by inhibiting the upregulation of inflammatory factors.
Collapse
|
43
|
Zhu H, Shi X, Ju D, Huang H, Wei W, Dong X. Anti-inflammatory effect of thalidomide on H1N1 influenza virus-induced pulmonary injury in mice. Inflammation 2015; 37:2091-8. [PMID: 24912813 DOI: 10.1007/s10753-014-9943-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The purpose of this study is to investigate the anti-inflammatory effect of thalidomide (Thd) on H1N1-induced acute lung injury in mice. BALB/C mice were infected intranasally with influenza A virus (H1N1) and then treated with Thd at a dose of 100 or 200 mg/kg/day for 7 days. Weight loss and survival of mice were monitored for 14 days after virus challenge, and the serum and lung tissues were collected at 4 days for histological and biochemical analysis. The results showed that Thd significantly improved the survival rate, reduced the infiltration of inflammatory cells and cytokine (e.g., IL-6, TNF-α) and chemokine (e.g., RANTES, IP-10) levels, and inhibited activated p-NFκB p65 in infected mice. These findings suggested that Thd may attenuate H1N1-induced pulmonary injury and thus may find use in the treatment of viral diseases.
Collapse
Affiliation(s)
- Haiyan Zhu
- Department of Biosynthesis, School of Pharmacy, Fudan University, 826 Zhangheng Road, 201203, Shanghai, China
| | | | | | | | | | | |
Collapse
|
44
|
Lv P, Li HY, Ji SS, Li W, Fan LJ. Thalidomide Inhibits Adhesion Molecules in Experimental Acute Pancreatitis-Associated Lung Injury. Drug Dev Res 2015; 76:24-30. [PMID: 25620023 DOI: 10.1002/ddr.21237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/10/2014] [Indexed: 12/30/2022]
Abstract
Preclinical Research The study evaluated the effect of thalidomide on adhesion molecule expression in acute pancreatitis-associated lung injury in rats. Acute pancreatitis was induced in rats by retrograde infusion of 5% sodium taurocholate into the bile-pancreatic duct, and thalidomide (100 mg/kg) was given daily by intragastric route for 8 days before this treatment. Serum lipase (LPS), protein levels in bronchoalveolar lavage fluid (BALF), superoxide dismutase (SOD), glutathione peroxidase (GSHpx), and malondialdehyde (MDA) levels in lung were measured. Compared with the acute pancreatitis- group, lung histopathology, serum LPS, protein levels in BALF, SOD, GSHpx, and MDA levels, and the expression levels of intercellular adhesion molecule-1 and E-selectin mRNA and protein in rats given thalidomide were improved (P < 0.01). Thus, thalidomide may reduce the expression of adhesion molecules via inhibition of oxidative stress to alleviate acute pancreatitis-associated lung injury in a rat model. Drug Dev Res, 2014. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peng Lv
- Department of Gastroenterology, Jining First People's Hospital, Jining, 272111, China
| | | | | | | | | |
Collapse
|
45
|
Carvalho TT, Borghi SM, Pinho-Ribeiro FA, Mizokami SS, Cunha TM, Ferreira SH, Cunha FQ, Casagrande R, Verri WA. Granulocyte-colony stimulating factor (G-CSF)-induced mechanical hyperalgesia in mice: Role for peripheral TNFα, IL-1β and IL-10. Eur J Pharmacol 2015; 749:62-72. [PMID: 25584775 DOI: 10.1016/j.ejphar.2014.12.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 12/17/2014] [Accepted: 12/22/2014] [Indexed: 01/28/2023]
Abstract
Granulocyte-colony stimulating factor (G-CSF) is a therapeutic approach to increase peripheral neutrophil counts after anti-tumor therapies. Pain is the major side effect of G-CSF. Intraplantar administration of G-CSF in mice induces mechanical hyperalgesia. However, the peripheral mechanisms involved in this effect were not elucidated. Therefore, the participation of pronociceptive cytokines tumor necrosis factor (TNF) alpha (TNFα), interleukin (IL)-1 beta (IL-1β) and antinociceptive cytokine IL-10 in G-CSF-induced mechanical hyperalgesia in mice was investigated. G-CSF-induced mechanical hyperalgesia was inhibited by systemic and local treatment with etanercept and IL-1 receptor antagonist (IL-1ra) or TNF receptor 1 (TNFR1) deficiency and increased in IL-10 deficient mice. In agreement, G-CSF injection induced significant TNFα, IL-1β and IL-10 production in paw tissue. G-CSF-induced hyperalgesia was dose-dependently inhibited by thalidomide (5-45mg/kg) and pentoxifylline (0.5-13.5mg/kg), and treatment with these drugs inhibited G-CSF-induced TNFα, IL-1β and IL-10 production. The combined treatment with pentoxifylline or thalidomide with morphine, at doses that are ineffective as single treatment, diminished G-CSF-induced hyperalgesia through inhibiting cytokine production. Indomethacin also reduces G-CSF hyperalgesia alone or combined with pentoxifylline or thalidomide. Thus, G-CSF-induced hyperalgesia might be mediate by peripheral production of pronociceptive cytokines TNFα and IL-1β and down-regulated by IL-10. Systemic IL-1ra reduced G-CSF-induced increase of peripheral neutrophil counts. However, local treatment with morphine, IL-1ra or etanercept, and systemic treatment with indomethacin, etanercept, thalidomide and pentoxifylline did not alter G-CSF-induced mobilization of neutrophils. Therefore, this study advances in the understanding of G-CSF-induced hyperalgesia and suggests therapeutic approaches for its control.
Collapse
Affiliation(s)
- Thacyana T Carvalho
- Department of Pathology, Center of Biological Science, Londrina State University, Rod. Celso Garcia Cid KM480 PR445, CEP 86057-970, Cx Postal 10.011, Londrina, Paraná, Brazil.
| | - Sergio M Borghi
- Department of Pathology, Center of Biological Science, Londrina State University, Rod. Celso Garcia Cid KM480 PR445, CEP 86057-970, Cx Postal 10.011, Londrina, Paraná, Brazil.
| | - Felipe A Pinho-Ribeiro
- Department of Pathology, Center of Biological Science, Londrina State University, Rod. Celso Garcia Cid KM480 PR445, CEP 86057-970, Cx Postal 10.011, Londrina, Paraná, Brazil.
| | - Sandra S Mizokami
- Department of Pathology, Center of Biological Science, Londrina State University, Rod. Celso Garcia Cid KM480 PR445, CEP 86057-970, Cx Postal 10.011, Londrina, Paraná, Brazil.
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Avenida Bandeirantes, 3900, CEP 14049-900 Ribeirao Preto, Sao Paulo, Brazil.
| | - Sergio H Ferreira
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Avenida Bandeirantes, 3900, CEP 14049-900 Ribeirao Preto, Sao Paulo, Brazil.
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Avenida Bandeirantes, 3900, CEP 14049-900 Ribeirao Preto, Sao Paulo, Brazil.
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, University Hospital (Health Science Centre), Londrina State University, Avenida Robert Koch, 60, Hospital Universitário, 86038-350 Londrina, Paraná, Brazil.
| | - Waldiceu A Verri
- Department of Pathology, Center of Biological Science, Londrina State University, Rod. Celso Garcia Cid KM480 PR445, CEP 86057-970, Cx Postal 10.011, Londrina, Paraná, Brazil.
| |
Collapse
|
46
|
Liu MW, Su MX, Zhang W, Wang YQ, Chen M, Wang L, Qian CY. Protective effect of Xuebijing injection on paraquat-induced pulmonary injury via down-regulating the expression of p38 MAPK in rats. Altern Ther Health Med 2014; 14:498. [PMID: 25511395 PMCID: PMC4301062 DOI: 10.1186/1472-6882-14-498] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 12/10/2014] [Indexed: 01/08/2023]
Abstract
Background Exposure to paraquat results in acute lung injury. A systemic inflammatory response has been widely established as a contributor to paraquat-induced acute lung injury. Recent studies have reported that consumption of Xuebijing prevents inflammatory response-induced diseases. This study investigated whether consumption of Xuebijing protected rats against paraquat-induced acute lung injury. Methods Adult male Sprague Dawley rats were randomly divided into four groups: control group; paraquat group; paraquat + Xuebijing group; and paraquat + dexamethasone group. Rats in the paraquat, paraquat + Xuebijing and paraquat + dexamethasone groups were intraperitoneally injected with paraquat (30 mg/kg) or administered paraquat and Xuebijing at 8 mL/kg or dexamethasone at 5 mg/kg, respectively, via an injection into the tail vein. Lung p38 MAPK, NF-κB65, IkB, p-IκB-α, HIF-1α, Nrf2 and TGF-β1 expression were essayed using western blotting. IL-6, TNF-α, IL-1β, IL-10, TGF-β1 and PIIIP were measured using ELISA. ROS, oxidised glutathione and glutathione activity were measured. Results After inducing acute lung injury with paraquat for 24 h, Xuebijing was observed to block lung p-p38 MAPK, NF-κB65, HIF-1α, p-IκB-α and TGF-β1 expression, and increased Nrf2 and IkB expression. The numbers of neutrophils and lymphocytes and total number of cells were significantly lower in the Xuebijing group compared with the control group. IL-6, TNF-α, IL-1β, TGF-β1 and PIIIP levels were significantly decreased in the Xuebijing group. ROS and oxidised glutathione activity were markedly inhibited by Xuebijing. Histological evaluation showed attenuation of the effects of Xuebijing on paraquat-induced lung injury. Compared with the paraquat + dexamethasone group, the Xuebijing + paraquat group showed no significant differences. Conclusions Inhibiting the expression of p38 MAPK and NF-κB65 was crucial for the protective effects of Xuebijing on paraquat-induced acute lung injury. The findings suggest that Xuebijing could effectively ameliorate paraquat-induced acute lung injury in rats. Xuebijing was as effective as dexamethasone at improving paraquat-induced lung injury by regulating lung inflammation, lung function and oxidative stress responses.
Collapse
|
47
|
Amirshahrokhi K, Khalili AR. The effect of thalidomide on ethanol-induced gastric mucosal damage in mice: involvement of inflammatory cytokines and nitric oxide. Chem Biol Interact 2014; 225:63-9. [PMID: 25478868 DOI: 10.1016/j.cbi.2014.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/06/2014] [Accepted: 11/25/2014] [Indexed: 12/21/2022]
Abstract
Excessive ethanol ingestion causes gastric mucosal damage through the inflammatory and oxidative processes. The present study was aimed to evaluate the protective effect of thalidomide on ethanol-induced gastric mucosal damage in mice. The animals were pretreated with vehicle or thalidomide (30 or 60 mg/kg, orally), and one hour later, the gastric mucosal injury was induced by oral administration of acidified ethanol. The animals were euthanized one hour after ethanol ingestion, and gastric tissues were collected to biochemical analyzes. The gastric mucosal lesions were assessed by macroscopic and histopathological examinations. The results showed that treatment of mice with thalidomide prior to the administration of ethanol dose-dependently reduced the gastric ulcer index. Thalidomide pretreatment significantly reduced the levels of pro-inflammatory cytokines [tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6], malondialdehyde (MDA) and myeloperoxidase (MPO) activity. In addition, thalidomide significantly inhibited ethanol-induced nitric oxide (NO) overproduction in gastric tissue. Histological observations showed that ethanol-induced gastric mucosal damage was attenuated by thalidomide pretreatment. It seems that thalidomide as an anti-inflammatory agent may have a protective effect against alcohol-induced mucosal damage by inhibition of neutrophil infiltration and reducing the production of nitric oxide and inflammatory cytokines in gastric tissue.
Collapse
Affiliation(s)
- Keyvan Amirshahrokhi
- Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali-Reza Khalili
- Division of Pathology, Imam Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
48
|
Tyagi N, Kumari A, Dash D, Singh R. Protective effects of intranasal curcumin on paraquot induced acute lung injury (ALI) in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:913-921. [PMID: 25461551 DOI: 10.1016/j.etap.2014.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 10/06/2014] [Accepted: 10/10/2014] [Indexed: 06/04/2023]
Abstract
Paraquot (PQ) is widely and commonly used as herbicide and has been reported to be hazardous as it causes lung injury. However, molecular mechanism underlying lung toxicity caused by PQ has not been elucidated. Curcumin, a known anti-inflammatory molecule derived from rhizomes of Curcuma longa has variety of pharmacological activities including free-radical scavenging properties but the protective effects of curcumin on PQ-induced acute lung injury (ALI) have not been studied. In this study, we aimed to study the effects of curcumin on ALI caused by PQ in male parke's strain mice which were challenged acutely by PQ (50mg/kg, i.p.) with or without curcumin an hour before (5mg/kg, i.n.) PQ intoxication. Lung specimens and the bronchoalveolar lavage fluid (BALF) were isolated for pathological and biochemical analysis after 48h of PQ exposure. Curcumin administration has significantly enhanced superoxide dismutase (SOD) and catalase activities. Lung wet/dry weight ratio, malondialdehyde (MDA) and lactate dehydrogenase (LDH) content, total cell number and myeloperoxidase (MPO) levels in BALF as well as neutrophil infiltration were attenuated by curcumin. Pathological studies also revealed that intranasal curcumin alleviate PQ-induced pulmonary damage and pro-inflammatory cytokine levels like tumor necrosis factor-α (TNF-α) and nitric oxide (NO). These results suggest that intranasal curcumin may directly target lungs and curcumin inhalers may prove to be effective in PQ-induced ALI treatment in near future.
Collapse
Affiliation(s)
- Namitosh Tyagi
- Department of Zoology, MMV, Banaras Hindu University, Varanasi, 221005, India
| | - Asha Kumari
- Department of Zoology, MMV, Banaras Hindu University, Varanasi, 221005, India
| | - D Dash
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Rashmi Singh
- Department of Zoology, MMV, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
49
|
Chen D, Zhang Z, Yao H, Cao Y, Xing H, Xu S. Pro- and anti-inflammatory cytokine expression in immune organs of the common carp exposed to atrazine and chlorpyrifos. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2014; 114:8-15. [PMID: 25175644 DOI: 10.1016/j.pestbp.2014.07.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 07/26/2014] [Accepted: 07/27/2014] [Indexed: 06/03/2023]
Abstract
Atrazine (ATR) and chlorpyrifos (CPF) are toxic and subject to long-term in vivo accumulation in different aquatic species throughout the world. The purpose of the present study was to examine the effect of ATR, CPF and combined ATR/CPF exposure on cytokines in the head kidney and spleen of common carp (Cyprinus carpio L.). The carp were sampled after a 40-d exposure to CPF and ATR, individually or in combination, followed by a 40-d recovery to measure the mRNA expression of IL-6fam (IL-6), IL-8, TNF-α, IL-10 and TGF-β1 (TGF-β) in the head kidney and spleen tissues. These results showed that the expression of cytokines IL-6, IL-8 and TNF-α in the head kidney and spleen was upregulated following ATR, CPF and mixed ATR/CPF exposure compared with the control group. The expression of IL-10 and TGF-β mRNA was significantly inhibited in both head kidney and spleen of carp exposed to ATR, CPF and the ATR/CPF mixture. The results suggested that long-term exposure of ATR, CPF and the ATR/CPF mixture in aquatic environments can induce the dysregulation of pro-/anti-inflammatory cytokine expression. The information regarding the effects of ATR and CPF on cytokine mRNA expression generated in this study will be important information for pesticides toxicology evaluation.
Collapse
Affiliation(s)
- Dechun Chen
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China; Department of Biological Engineering, Jilin Engineering Vocational College, 1299 Changfa Road, Siping 136001, China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Haidong Yao
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Ye Cao
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China
| | - Houjuan Xing
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China; Animal Health Supervision Institute of Heilongjiang Province, 243 Haping Road, Xiangfang District, Harbin 150069, China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, 59 Mucai Street, Harbin 150030, China.
| |
Collapse
|
50
|
Thalidomide alleviates acute pancreatitis-associated lung injury via down-regulation of NFκB induced TNF-α. Pathol Res Pract 2014; 210:558-64. [DOI: 10.1016/j.prp.2014.04.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 04/21/2014] [Accepted: 04/29/2014] [Indexed: 12/20/2022]
|