1
|
Dong S, Han J, Sun XY, Zhang B, Wang W. A novel 2D g-C 3N 4 material applied for Paraquat adsorbing and detoxifying in vitro and in vivo. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115594. [PMID: 37856982 DOI: 10.1016/j.ecoenv.2023.115594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
In the environmental safety area, the widespread use of the herbicide Paraquat (PQ) poses a great threat to hydrobionts and mammals. Due to the lack of specific antidote, it may lead to irreversible pulmonary fibrosis with a mortality rate of 60%. Therefore, it is necessary to develop an effective and specific PQ antidote. The g-C3N4 (HPCN) with excellent surface physicochemical properties was prepared by a two-step calcination method using urea and dicyandiamide as raw materials, showing a significant photocatalyst against environmental PQ pollution. The SEM results showed that HPCN possesses a porous layered structure. X-ray diffraction and infrared spectroscopy indicated that the conjugated aromatic rings were orderly stacked, forming a 2D layered structure of g-C3N4. The HPCN had a larger specific surface area (56.84 m2 g-1) and pore volume (0.2718 cm3 g-1), which enhanced its adsorption capacity and photocatalytic activity. HPCN exhibited an effective adsorption rate of 38.25% for PQ in water under light. Compared with the PQ group (54.8%), the cell viability of the HPCN group (91.4%) significantly increased by 36.6%, and the SEM observation revealed the restoration of normal cell morphology. The HPCN effectively reduced PQ content in zebrafish and mice in vivo, resulting in an approximately 70% increase in survival rate. The UV-Vis results indicated that the adsorption rate of HPCN for PQ in zebrafish was 43.5%. The enhanced catalytic performance of HPCN provides a promising solution for the detoxification of PQ and of other environmental pollutants.
Collapse
Affiliation(s)
- Shi Dong
- School of Chemical Engineering, Shandong Institute of Petroleum and Chemical Technology, Shandong 257061, PR China; Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, PR China; Key Laboratory of Xinjiang Phytomedicine Resources and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832002, PR China.
| | - Jun Han
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, PR China.
| | - Xi-Yin Sun
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, PR China.
| | - Bo Zhang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, PR China.
| | - Wei Wang
- School of Chemical Engineering, Shandong Institute of Petroleum and Chemical Technology, Shandong 257061, PR China; School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, PR China.
| |
Collapse
|
2
|
Dolivo DM, Reed CR, Gargiulo KA, Rodrigues AE, Galiano RD, Mustoe TA, Hong SJ. Anti-fibrotic effects of statin drugs: a review of evidence and mechanisms. Biochem Pharmacol 2023:115644. [PMID: 37321414 DOI: 10.1016/j.bcp.2023.115644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
Fibrosis is a pathological repair process common among organs, that responds to damage by replacement of tissue with non-functional connective tissue. Despite the widespread prevalence of tissue fibrosis, manifesting in numerous disease states across myriad organs, therapeutic modalities to prevent or alleviate fibrosis are severely lacking in quantity and efficacy. Alongside development of new drugs, repurposing of existing drugs may be a complementary strategy to elect anti-fibrotic compounds for pharmacologic treatment of tissue fibrosis. Drug repurposing can provide key advantages to de novo drug discovery, harnessing the benefits of previously elucidated mechanisms of action and already existing pharmacokinetic profiles. One class of drugs a wealth of clinical data and extensively studied safety profiles is the statins, a class of antilipidemic drugs widely prescribed for hypercholesterolemia. In addition to these widely utilized lipid-lowering effects, increasing data from cellular, pre-clinical mammalian, and clinical human studies have also demonstrated that statins are able to alleviate tissue fibrosis originating from a variety of pathological insults via lesser-studied, pleiotropic effects of these drugs. Here we review literature demonstrating evidence for direct effects of statins antagonistic to fibrosis, as well as much of the available mechanistic data underlying these effects. A more complete understanding of the anti-fibrotic effects of statins may enable a clearer picture of their anti-fibrotic potential for various clinical indications. Additionally, more lucid comprehension of the mechanisms by which statins exert anti-fibrotic effects may aid in development of novel therapeutic agents that target similar pathways but with greater specificity or efficacy.
Collapse
Affiliation(s)
- David M Dolivo
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States.
| | - Charlotte R Reed
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Kristine A Gargiulo
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Adrian E Rodrigues
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Robert D Galiano
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Thomas A Mustoe
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States
| | - Seok Jong Hong
- Department of Surgery-Northwestern University Feinberg School of Medicine, United States.
| |
Collapse
|
3
|
Matsubayashi S, Ito S, Araya J, Kuwano K. Drugs against metabolic diseases as potential senotherapeutics for aging-related respiratory diseases. Front Endocrinol (Lausanne) 2023; 14:1079626. [PMID: 37077349 PMCID: PMC10106576 DOI: 10.3389/fendo.2023.1079626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Recent advances in aging research have provided novel insights for the development of senotherapy, which utilizes cellular senescence as a therapeutic target. Cellular senescence is involved in the pathogenesis of various chronic diseases, including metabolic and respiratory diseases. Senotherapy is a potential therapeutic strategy for aging-related pathologies. Senotherapy can be classified into senolytics (induce cell death in senescent cells) and senomorphics (ameliorate the adverse effects of senescent cells represented by the senescence-associated secretory phenotype). Although the precise mechanism has not been elucidated, various drugs against metabolic diseases may function as senotherapeutics, which has piqued the interest of the scientific community. Cellular senescence is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), which are aging-related respiratory diseases. Large-scale observational studies have reported that several drugs, such as metformin and statins, may ameliorate the progression of COPD and IPF. Recent studies have reported that drugs against metabolic diseases may exert a pharmacological effect on aging-related respiratory diseases that can be different from their original effect on metabolic diseases. However, high non-physiological concentrations are needed to determine the efficacy of these drugs under experimental conditions. Inhalation therapy may increase the local concentration of drugs in the lungs without exerting systemic adverse effects. Thus, the clinical application of drugs against metabolic diseases, especially through an inhalation treatment modality, can be a novel therapeutic approach for aging-related respiratory diseases. This review summarizes and discusses accumulating evidence on the mechanisms of aging, as well as on cellular senescence and senotherapeutics, including drugs against metabolic diseases. We propose a developmental strategy for a senotherapeutic approach for aging-related respiratory diseases with a special focus on COPD and IPF.
Collapse
|
4
|
Signaling pathways involved in paraquat-induced pulmonary toxicity: Molecular mechanisms and potential therapeutic drugs. Int Immunopharmacol 2022; 113:109301. [DOI: 10.1016/j.intimp.2022.109301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
|
5
|
Yildirim M, Kayalar O, Atahan E, Oztay F. Atorvastatin attenuates pulmonary fibrosis in mice and human lung fibroblasts, by the regulation of myofibroblast differentiation and apoptosis. J Biochem Mol Toxicol 2022; 36:e23074. [PMID: 35416377 DOI: 10.1002/jbt.23074] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 03/02/2022] [Accepted: 04/01/2022] [Indexed: 12/25/2022]
Abstract
Statins have anti-inflammatory and antifibrotic effects in addition to cholesterol-lowering effect. We aimed to investigate the effect of atorvastatin (ATR) in fibrotic mouse lung and human lung fibroblasts (MRC5s). Pulmonary fibrosis was induced by a single dose of bleomycin by intratracheal instillation in adult mice. ATR was administered (20 mg/kg ip) to mice with healthy and pulmonary fibrosis for 10 days from Day 7 of the experiment. Mice were dissected on the 21st day. The levels of alpha-smooth muscle actin (α-SMA), pSMAD2/3, LOXL2, and p-Src were determined by Western blot analysis in the lungs. Furthermore, a group of MRC5 was differentiated into myofibroblasts by transforming growth factor-beta (TGF-β). Another group of MRC5s was treated with 10 µM ATR at 24 h after TGF-β stimulation. Cells were collected at 0, 24, 48, and 72 h. The effects of ATR on myofibroblast differentiation, apoptosis, and TGF-β and Wnt/β-catenin signaling activations were examined by Western blot analysis and flow cytometry in MRC5s. ATR attenuated pulmonary fibrosis by regulating myofibroblast differentiation and interstitial accumulation of collagen, by acting on LOXL2, p-Src, and pSMAD2/3 in mice lungs. Additionally, it blocked myofibroblast differentiation via reduced TGF-β and Wnt/β-catenin signaling and decreased α-SMA in MRC5s stimulated with TGF-β. Moreover, ATR caused myofibroblast apoptosis via caspase-3 activation. ATR treatment attenuates pulmonary fibrosis in mice treated with bleomycin. It also inhibits fibroblast/myofibroblast activation, by both reducing myofibroblasts differentiation and inducing myofibroblast apoptosis.
Collapse
Affiliation(s)
- Merve Yildirim
- Department of Biology, Science Faculty, Istanbul University, Istanbul, Turkey
| | - Ozgecan Kayalar
- Department of Biology, Science Faculty, Istanbul University, Istanbul, Turkey.,Koç University School of Medicine Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Ersan Atahan
- Department of Chest Diseases, Cerrahpasa School of Medicine, Istanbul University Cerrahpasa, Istanbul, Turkey
| | - Fusun Oztay
- Department of Biology, Science Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
6
|
Shi X, Chen Y, Liu Q, Mei X, Liu J, Tang Y, Luo R, Sun D, Ma Y, Wu W, Tu W, Zhao Y, Xu W, Ke Y, Jiang S, Huang Y, Zhang R, Wang L, Chen Y, Xia J, Pu W, Zhu H, Zuo X, Li Y, Xu J, Gao F, Wei D, Chen J, Yin W, Wang Q, Dai H, Yang L, Guo G, Cui J, Song N, Zou H, Zhao S, Distler JH, Jin L, Wang J. LDLR dysfunction induces LDL accumulation and promotes pulmonary fibrosis. Clin Transl Med 2022; 12:e711. [PMID: 35083881 PMCID: PMC8792399 DOI: 10.1002/ctm2.711] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/27/2021] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
Treatments for pulmonary fibrosis (PF) are ineffective because its molecular pathogenesis and therapeutic targets are unclear. Here, we show that the expression of low-density lipoprotein receptor (LDLR) was significantly decreased in alveolar type II (ATII) and fibroblast cells, whereas it was increased in endothelial cells from systemic sclerosis-related PF (SSc-PF) patients and idiopathic PF (IPF) patients compared with healthy controls. However, the plasma levels of low-density lipoprotein (LDL) increased in SSc-PF and IPF patients. The disrupted LDL-LDLR metabolism was also observed in four mouse PF models. Upon bleomycin (BLM) treatment, Ldlr-deficient (Ldlr-/-) mice exhibited remarkably higher LDL levels, abundant apoptosis, increased fibroblast-like endothelial and ATII cells and significantly earlier and more severe fibrotic response compared to wild-type mice. In vitro experiments revealed that apoptosis and TGF-β1 production were induced by LDL, while fibroblast-like cell accumulation and ET-1 expression were induced by LDLR knockdown. Treatment of fibroblasts with LDL or culture medium derived from LDL-pretreated endothelial or epithelial cells led to obvious fibrotic responses in vitro. Similar results were observed after LDLR knockdown operation. These results suggest that disturbed LDL-LDLR metabolism contributes in various ways to the malfunction of endothelial and epithelial cells, and fibroblasts during pulmonary fibrogenesis. In addition, pharmacological restoration of LDLR levels by using a combination of atorvastatin and alirocumab inhibited BLM-induced LDL elevation, apoptosis, fibroblast-like cell accumulation and mitigated PF in mice. Therefore, LDL-LDLR may serve as an important mediator in PF, and LDLR enhancing strategies may have beneficial effects on PF.
Collapse
Affiliation(s)
- Xiangguang Shi
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Yahui Chen
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Xueqian Mei
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Jing Liu
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
- Division of RheumatologyHuashan hospital, Fudan UniversityShanghaiP. R. China
| | - Yulong Tang
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Ruoyu Luo
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Dayan Sun
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Yanyun Ma
- MOE Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life SciencesFudan UniversityShanghaiP. R. China
- Institute for Six‐sector EconomyFudan UniversityShanghaiP. R. China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Wenzhen Tu
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Yinhuan Zhao
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Weihong Xu
- The Clinical Laboratory of Tongren HosipitalShanghai Jiaotong UniversityShanghaiP. R. China
| | - Yuehai Ke
- Department of Pathology and PathophysiologyZhejiang University School of MedicineHangzhouZhejiang ProvinceP. R. China
| | - Shuai Jiang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Yan Huang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Rui Zhang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
- Institute for Six‐sector EconomyFudan UniversityShanghaiP. R. China
| | - Lei Wang
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Yuanyuan Chen
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Jingjing Xia
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Weilin Pu
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Honglin Zhu
- Department of Internal Medicine 3 and Institute for Clinical ImmunologyUniversity of ErlangenNurembergGermany
- Department of Rheumatology, Xiangya HospitalCentral South UniversityChangshaHunan ProvinceP. R. China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya HospitalCentral South UniversityChangshaHunan ProvinceP. R. China
| | - Yisha Li
- Department of Rheumatology, Xiangya HospitalCentral South UniversityChangshaHunan ProvinceP. R. China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Fei Gao
- Wuxi Lung Transplant CenterWuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiP. R. China
| | - Dong Wei
- Wuxi Lung Transplant CenterWuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiP. R. China
| | - Jingyu Chen
- Wuxi Lung Transplant CenterWuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiP. R. China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongP. R. China
| | - Qingwen Wang
- Rheumatology and Immunology DepartmentPeking University Shenzhen HospitalShenzhenP. R. China
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory MedicineChinese Academy of Medical ScienceBeijingP. R. China
| | - Libing Yang
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory MedicineChinese Academy of Medical ScienceBeijingP. R. China
- School of MedicineTsinghua UniversityBeijingP. R. China
| | - Gang Guo
- Department of Rheumatology and ImmunologyYiling Hospital Affiliated to Hebei Medical UniversityShijiazhuangHebei ProvinceP. R. China
| | - Jimin Cui
- Department of Rheumatology and ImmunologyYiling Hospital Affiliated to Hebei Medical UniversityShijiazhuangHebei ProvinceP. R. China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityFudan Zhangjiang InstituteShanghaiP. R. China
| | - Hejian Zou
- Division of RheumatologyHuashan hospital, Fudan UniversityShanghaiP. R. China
- Institute of Rheumatology, Immunology and AllergyFudan UniversityShanghaiP. R. China
| | - Shimin Zhao
- Institute of Metabolism and Integrative BiologyFudan UniversityShanghaiP. R. China
| | - Jörg H.W. Distler
- Department of Internal Medicine 3 and Institute for Clinical ImmunologyUniversity of ErlangenNurembergGermany
| | - Li Jin
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058)Chinese Academy of Medical SciencesShanghaiP. R. China
| | - Jiucun Wang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
- Institute of Rheumatology, Immunology and AllergyFudan UniversityShanghaiP. R. China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058)Chinese Academy of Medical SciencesShanghaiP. R. China
| |
Collapse
|
7
|
Yang Z, Wang M, Ren Y, Li L, Cao L, Zhang W, Lv K, Sun Z, Nie S. Inhibition of Wnt10b/β-catenin signaling alleviates pulmonary fibrogenesis induced by paraquat in vivo and in vitro. Life Sci 2021; 286:120027. [PMID: 34627778 DOI: 10.1016/j.lfs.2021.120027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/23/2021] [Accepted: 10/01/2021] [Indexed: 01/04/2023]
Abstract
Pulmonary fibrosis (PF) caused by paraquat remains a critical issue, and the molecular mechanisms are still unclear. Epithelial-mesenchymal transition (EMT) is regarded as a hallmark of PF, conferring alveolar epithelial cells partial mesenchymal characteristics, facilitating migration, expressing excessive extracellular matrix components, and participating in lung parenchyma remodeling and stiffening. Aberration of Wnt signaling has been identified in EMT and PF, and Wnt protein family consists of 19 ligands. The relationship of the specific Wnt ligands and fibrogenesis induced by PQ was not well defined. In current study, PQ-induced lung fibrosis rat model and EMT cell model were utilized to investigate the underlying molecular mechanisms both in vivo and in vitro. The results demonstrated that canonical Wnt/β-catenin signaling was highly activated and Wnt10b was the most affected. Additionally, suppression of Wnt10b by RNA interference could reverse EMT in vitro and detain the process of PF in vivo. These data establish Wnt10b as the key regulator of EMT and lung fibrogenesis, and suggest the potential of targeted interference against Wnt10b as a promising therapeutic strategy for lung fibrosis.
Collapse
Affiliation(s)
- Zhizhou Yang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China; Department of Emergency Medicine, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, PR China
| | - Mengmeng Wang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China; Department of Emergency Medicine, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, PR China
| | - Yi Ren
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Liang Li
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Liping Cao
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Kongbo Lv
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Zhaorui Sun
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China; Department of Emergency Medicine, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, PR China.
| | - Shinan Nie
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China; Department of Emergency Medicine, the First School of Clinical Medicine, Southern Medical University, Nanjing, 210002, PR China.
| |
Collapse
|
8
|
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: 2.3] [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
|
9
|
Subbiah R, Tiwari RR. The herbicide paraquat-induced molecular mechanisms in the development of acute lung injury and lung fibrosis. Crit Rev Toxicol 2021; 51:36-64. [PMID: 33528289 DOI: 10.1080/10408444.2020.1864721] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The herbicide paraquat (PQ; 1,1'-dimethyl-4,4'-bipyridylium dichloride) is a highly toxic organic heterocyclic herbicide that has been widely used in agricultural settings. Since its commercial introduction in the early 1960s, numerous cases of fatal PQ poisonings attributed to accidental and/or intentional ingestion of PQ concentrated formulations have been reported. The clinical manifestations of the respiratory system during the acute phase of PQ poisoning mainly include acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), followed by pulmonary fibrosis in a later phase. The focus of this review is to summarize the most recent publications related to PQ-induced lung toxicity as well as the underlying molecular mechanisms for PQ-mediated pathologic processes. Growing sets of data from in vitro and in vivo models have demonstrated the involvement of the PQ in regulating lung oxidative stress, inflammatory response, epigenetics, apoptosis, autophagy, and the progression of lung fibrosis. The article also summarizes novel therapeutic avenues based on a literature review, which can be explored as potential means to combat PQ-induced lung toxicity. Finally, we also presented clinical studies on the association of PQ exposure with the incidence of lung injury and pulmonary fibrosis.
Collapse
Affiliation(s)
- Rajasekaran Subbiah
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Rajnarayan R Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
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: 2.0] [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
|
Hypoxia-Inducible Factor-1: A Potential Target to Treat Acute Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8871476. [PMID: 33282113 PMCID: PMC7685819 DOI: 10.1155/2020/8871476] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is an acute hypoxic respiratory insufficiency caused by various intra- and extrapulmonary injury factors. Presently, excessive inflammation in the lung and the apoptosis of alveolar epithelial cells are considered to be the key factors in the pathogenesis of ALI. Hypoxia-inducible factor-1 (HIF-1) is an oxygen-dependent conversion activator that is closely related to the activity of reactive oxygen species (ROS). HIF-1 has been shown to play an important role in ALI and can be used as a potential therapeutic target for ALI. This manuscript will introduce the progress of HIF-1 in ALI and explore the feasibility of applying inhibitors of HIF-1 to ALI, which brings hope for the treatment of ALI.
Collapse
|
12
|
Buendía JA, Justinico Castro JA, Vela LJT, Sinisterra D, Sánchez Villamil JP, Zuluaga Salazar AF. Comparison of four pharmacological strategies aimed to prevent the lung inflammation and paraquat-induced alveolar damage. BMC Res Notes 2019; 12:584. [PMID: 31533801 PMCID: PMC6749662 DOI: 10.1186/s13104-019-4598-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/31/2019] [Indexed: 11/17/2022] Open
Abstract
Objective The aim of this study was to compare in vivo effect of five pharmacological options on inflammation and pulmonary fibrosis induced by paraquat. Methods 54 Wistar SPF rats were used. After 2 h post-intoxication with paraquat ion, groups of 9 animals were randomly assigned to (1) cyclophosphamide plus dexamethasone (2) low molecular weight heparin (3) unfractionated heparin (4) vitamin C every 24 h, (5) atorvastatin or (6) placebo with intraperitoneal saline. Lung inflammation, alveolar injury, hepatocyte damage, hepatic regeneration, acute tubular necrosis and kidney congestion were evaluated. Results In the control group 100% of animals presented moderate and severe lung inflammation, while in the groups with atorvastatin and intratracheal heparin this proportion was lower (55.5%; CI 26.6–81.3%) (p = 0.025). A lower degree of moderate or severe hepatic regeneration was evident in the treatment groups with atorvastatin (p = 0.009). In this study was demonstrated that statins and heparin might have a protective effect in the paraquat-induced destructive phase. More evidence is needed to evaluated of dose–response effects of these drugs before to study in clinical trials.
Collapse
Affiliation(s)
- Jefferson Antonio Buendía
- Grupo de Investigación en Farmacología y Toxicología, Centro de Información y Estudio de Medicamentos y Tóxicos (CIEMTO), Facultad de Medicina, Universidad de Antioquia, Carrera 51D #62-29, Medellín, Colombia.
| | | | - Laura Joanna Tapia Vela
- Departamento de Patología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Denis Sinisterra
- Grupo de Investigación en Farmacología y Toxicología, Centro de Información y Estudio de Medicamentos y Tóxicos (CIEMTO), Facultad de Medicina, Universidad de Antioquia, Carrera 51D #62-29, Medellín, Colombia
| | | | - Andrés Felipe Zuluaga Salazar
- Grupo de Investigación en Farmacología y Toxicología, Centro de Información y Estudio de Medicamentos y Tóxicos (CIEMTO), Facultad de Medicina, Universidad de Antioquia, Carrera 51D #62-29, Medellín, Colombia
| |
Collapse
|
13
|
Jendzjowsky NG, Kelly MM. The Role of Airway Myofibroblasts in Asthma. Chest 2019; 156:1254-1267. [PMID: 31472157 DOI: 10.1016/j.chest.2019.08.1917] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/14/2019] [Accepted: 08/11/2019] [Indexed: 12/17/2022] Open
Abstract
Airway remodeling is a characteristic feature of asthma and is thought to play an important role in the pathogenesis of airway hyperresponsiveness. Myofibroblasts are key structural cells involved in injury and repair, and there is evidence that dysregulation of their normal function contributes to airway remodeling. Despite the importance of myofibroblasts, a lack of specific cellular markers and inconsistent nomenclature have limited recognition of their key role in airway remodeling. Myofibroblasts are increased several-fold in the airways in asthma, in proportion to the severity of the disease. Myofibroblasts are postulated to be derived from both tissue-resident and bone marrow-derived cells, depending on the stage of injury and the tissue. A small number of studies have demonstrated attenuation of myofibroblast numbers and also reversal of established myofibroblast populations in asthma and other inflammatory processes. In this article, we review what is currently known about the biology of myofibroblasts in the airways in asthma and identify potential targets to reduce or reverse the remodeling process. However, further translational research is required to better understand the mechanistic role of the myofibroblast in asthma.
Collapse
Affiliation(s)
- Nicholas G Jendzjowsky
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Margaret M Kelly
- Airway Inflammation Research Group, Snyder Institute for Chronic Disease, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|