1
|
Li J, Ma W, Tang Z, Li Y, Zheng R, Xie Y, Li G. Macrophage‑driven pathogenesis in acute lung injury/acute respiratory disease syndrome: Harnessing natural products for therapeutic interventions (Review). Mol Med Rep 2025; 31:16. [PMID: 39513609 PMCID: PMC11551695 DOI: 10.3892/mmr.2024.13381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a common respiratory disease characterized by hypoxemia and respiratory distress. It is associated with high morbidity and mortality. Due to the complex pathogenesis of ALI, the clinical management of patients with ALI/ARDS is challenging, resulting in numerous post‑treatment sequelae and compromising the quality of life of patients. Macrophages, as a class of innate immune cells, play an important role in ALI/ARDS. In recent years, the functions and phenotypes of macrophages have been better understood due to the development of flow cytometry, immunofluorescence, single‑cell sequencing and spatial genomics. However, no macrophage‑targeted drugs for the treatment of ALI/ARDS currently exist in clinical practice. Natural products are important for drug development, and it has been shown that numerous natural compounds from herbal medicine can alleviate ALI/ARDS caused by various factors by modulating macrophage abnormalities. In the present review, the natural products from herbal medicine that can modulate macrophage abnormalities in ALI/ARDS to treat ALI/ARDS are introduced, and their mechanisms of action, discovered in the previous five years (2019‑2024), are presented. This will provide novel ideas and directions for further research, to develop new drugs for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Jincun Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wenyu Ma
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Zilei Tang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yingming Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Ruiyu Zheng
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuhuan Xie
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Gang Li
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
2
|
Gagnani R, Srivastava M, Suri M, Singh H, Shanker Navik U, Bali A. A focus on c-Jun-N-terminal kinase signaling in sepsis-associated multiple organ dysfunction: Mechanisms and therapeutic strategies. Int Immunopharmacol 2024; 143:113552. [PMID: 39536486 DOI: 10.1016/j.intimp.2024.113552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Sepsis is a life-threatening condition characterized by a widespread inflammatory response to infection, inevitably leading to multiple organ dysfunctions. Extensive research, both in vivo and in vitro, has revealed key factors contributing to sepsis, such as apoptosis, inflammation, cytokine release, oxidative stress, and systemic stress. The changes observed during sepsis-induced conditions are mainly attributed to altered signal transduction pathways, which play a critical role in cell proliferation, migration, and apoptosis. C-Jun N-terminal kinases, JNKs, and serine/threonine protein kinases in the mitogen-activated super family have gained considerable interest for their contribution to cellular events under sepsis conditions. JNK1 and JNK2 are present in various tissues like the lungs, liver, and intestine, while JNK3 is found in neurons. The JNK pathway plays a crucial role in the signal transduction of cytokines related to sepsis development, notably TNF-α and IL-1β. Activated JNK leads to apoptosis, causing tissue damage and organ dysfunction. Further, JNK activation is significant in several inflammatory conditions. Pharmacologically inhibiting JNK has been shown to prevent sepsis-associated damage across multiple organs, including the lungs, liver, intestines, heart, and kidneys. Multiple signaling pathways have been implicated in sepsis, including JNK/c-Myc, Mst1-JNK, MKK4-JNK, JNK-dependent autophagy, and Sirt1/FoxO3a. The review examines the role of JNK signaling in the development of sepsis-induced multiple-organ dysfunction through specific mechanisms. It also discusses different therapeutic approaches to target JNK. This review emphasizes the potential of JNKs as targets for the development of therapeutic agents for sepsis and the associated specific organ damage.
Collapse
Affiliation(s)
- Riya Gagnani
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| | - Mukul Srivastava
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Manisha Suri
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Harshita Singh
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Uma Shanker Navik
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Bali
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
3
|
Liu D, Weng S, Fu C, Guo R, Chen M, Shi B, Weng J. Autophagy in Acute Lung Injury. Cell Biochem Biophys 2024:10.1007/s12013-024-01604-2. [PMID: 39527232 DOI: 10.1007/s12013-024-01604-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Acute lung injury (ALI) is a critical condition marked by rapid-onset respiratory failure due to extensive inflammation and increased pulmonary vascular permeability, often progressing to acute respiratory distress syndrome (ARDS) with high mortality. Autophagy, a cellular degradation process essential for removing damaged organelles and proteins, plays a crucial role in regulating lung injury and repair. This review examines the protective role of autophagy in maintaining cellular function and reducing inflammation and oxidative stress in ALI. It underscores the necessity of precise regulation to fully harness the therapeutic potential of autophagy in this context. We summarize the mechanisms by which autophagy influences lung injury and repair, discuss the interplay between autophagy and apoptosis, and examine potential therapeutic strategies, including autophagy inducers, targeted autophagy signaling pathways, antioxidants, anti-inflammatory drugs, gene editing, and stem cell therapy. Understanding the role of autophagy in ALI could lead to novel interventions for improving patient outcomes and reducing mortality rates associated with this severe condition.
Collapse
Affiliation(s)
- Danjuan Liu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Shuoyun Weng
- School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, China
| | - Chunjin Fu
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Rongjie Guo
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Min Chen
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Bingbing Shi
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China
| | - Junting Weng
- Department of Critical Care Medicine, the Affiliated Hospital of Putian University, Putian, 351100, China.
| |
Collapse
|
4
|
Wang Q, Ning Y, Zhang J, Du X, Xu Z, Hu Y, Gao F, Chen Y. Rapamycin and Hyperoside-Co-loaded Macrophage Delivery System Enhanced Pulmonary Fibrosis Therapy by Autophagy Upregulation and Epithelial-to-Mesenchymal Transition Inhibition. ACS APPLIED MATERIALS & INTERFACES 2024; 16:48993-49002. [PMID: 39225760 DOI: 10.1021/acsami.4c07604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pulmonary fibrosis is a lethal interstitial lung disease, for which current treatments are inadequate in halting its progression. A significant factor contributing to the development of fibrosis is insufficient autophagy, which leads to increased fibroblast proliferation and collagen deposition. However, treatments aimed at upregulating autophagy often cause further lung pathology due to the disruption of epithelial cell balance. In response, we have developed a novel macrophage delivery system loaded with an epithelial-to-mesenchymal transition inhibitor, hyperoside (HYP), and an autophagy inducer, rapamycin (RAP). This system targets the fibrotic areas of the lung through chemotaxis, releases liposomes via macrophage extracellular traps, and effectively inhibits fibroblast proliferation while restoring the alveolar structure through the combined effects of RAP and HYP, ultimately reducing lung pathology without causing systemic toxicity. Our findings not only highlight a promising method to enhance autophagy-based treatments for pulmonary fibrosis but also demonstrate the potential of macrophages as effective nanocarriers for drug delivery.
Collapse
Affiliation(s)
- Qi Wang
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yuanmeng Ning
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jinru Zhang
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xu Du
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zihan Xu
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yongcheng Hu
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Gao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yanzuo Chen
- Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
5
|
Xiang Y, Zhong Y, Lai X, Fang Z, Su G, Lv Y, Tang X, Ouyang L, Gao X, Zheng H, He L, Chen J, Huang J, Lai T. Myeloid-specific Hdac10 deletion protects against LPS-induced acute lung injury via P62 acetylation at lysine 165. Respir Res 2024; 25:263. [PMID: 38956592 PMCID: PMC11221109 DOI: 10.1186/s12931-024-02891-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Aberrant activation of macrophages is associated with pathogenesis of acute lung injury (ALI). However, the potential pathogenesis has not been explored. OBJECTIVES We aimed to identify whether histone deacetylase (HDAC) 10 is involved in lipopolysaccharide (LPS)-exposed ALI and reveal the underlying pathogenesis by which it promotes lung inflammation in LPS-exposed ALI via modifying P62 with deacetylation. METHODS We constructed an ALI mice model stimulated with LPS to determine the positive effect of Hdac10 deficiency. Moreover, we cultured murine alveolar macrophage cell line (MH-S cells) and primary bone marrow-derived macrophages (BMDMs) to explore the pro-inflammatory activity and mechanism of HDAC10 after LPS challenge. RESULTS HDAC10 expression was increased both in mice lung tissues and macrophage cell lines and promoted inflammatory cytokines production exposed to LPS. Hdac10 deficiency inhibited autophagy and inflammatory response after LPS stimulation. In vivo, Hdac10fl/fl-LysMCre mice considerably attenuated lung inflammation and inflammatory cytokines release exposed to LPS. Mechanistically, HDAC10 interacts with P62 and mediates P62 deacetylation at lysine 165 (K165), by which it promotes P62 expression and increases inflammatory cytokines production. Importantly, we identified that Salvianolic acid B (SAB), an HDAC10 inhibitor, reduces lung inflammatory response in LPS-stimulated ALI. CONCLUSION These results uncover a previously unknown role for HDAC10 in regulating P62 deacetylation and aggravating lung inflammation in LPS-induced ALI, implicating that targeting HDAC10 is an effective therapy for LPS-exposed ALI.
Collapse
Affiliation(s)
- Yuanyuan Xiang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Yu Zhong
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Xianwen Lai
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Zhenfu Fang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Guomei Su
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Yingying Lv
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Xiantong Tang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Lihuan Ouyang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China
| | - Xiao Gao
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Hongying Zheng
- The Department of Pulmonary and Critical Care Medicine, The Third People' s Hospital of Xining City, Qinghai, 810005, China
| | - Lilin He
- The Department of Pulmonary and Critical Care Medicine, The Third People' s Hospital of Xining City, Qinghai, 810005, China
| | - Jialong Chen
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523121, China.
| | - Jiewen Huang
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China.
- Dongguan Key Laboratory of Immune Inflammation and Metabolism, Dongguan, 523121, China.
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, Dongguan Institute of Respiratory Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523121, China.
- Dongguan Key Laboratory of Immune Inflammation and Metabolism, Dongguan, 523121, China.
| |
Collapse
|
6
|
Al-Bari MAA, Peake N, Eid N. Tuberculosis-diabetes comorbidities: Mechanistic insights for clinical considerations and treatment challenges. World J Diabetes 2024; 15:853-866. [PMID: 38766427 PMCID: PMC11099355 DOI: 10.4239/wjd.v15.i5.853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/08/2024] [Accepted: 03/21/2024] [Indexed: 05/10/2024] Open
Abstract
Tuberculosis (TB) remains a leading cause of death among infectious diseases, particularly in poor countries. Viral infections, multidrug-resistant and ex-tensively drug-resistant TB strains, as well as the coexistence of chronic illnesses such as diabetes mellitus (DM) greatly aggravate TB morbidity and mortality. DM [particularly type 2 DM (T2DM)] and TB have converged making their control even more challenging. Two contemporary global epidemics, TB-DM behaves like a syndemic, a synergistic confluence of two highly prevalent diseases. T2DM is a risk factor for developing more severe forms of multi-drug resistant-TB and TB recurrence after preventive treatment. Since a bidirectional relationship exists between TB and DM, it is necessary to concurrently treat both, and promote recommendations for the joint management of both diseases. There are also some drug-drug interactions resulting in adverse treatment outcomes in TB-DM patients including treatment failure, and reinfection. In addition, autophagy may play a role in these comorbidities. Therefore, the TB-DM comorbidities present several health challenges, requiring a focus on multidisciplinary collaboration and integrated strategies, to effectively deal with this double burden. To effectively manage the comorbidity, further screening in affected countries, more suitable drugs, and better treatment strategies are required.
Collapse
Affiliation(s)
| | - Nicholas Peake
- Biosciences and Chemistry and Biomolecular Research Centre, Sheffield Hallam University, Sheffield S1 1WB, United Kingdom
| | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
7
|
Wang S, Yu J, Liu Y, Yu J, Ma Y, Zhou L, Liu X, Liu L, Li W, Niu X. Bletilla striata polysaccharide attenuated the progression of pulmonary fibrosis by inhibiting TGF-β1/Smad signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117680. [PMID: 38171465 DOI: 10.1016/j.jep.2023.117680] [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: 10/22/2023] [Revised: 12/09/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bletilla striata, a traditional medicinal plant, has been utilized as a folk medicine for many years because of its superior biological activity in China. However, Bletilla striata polysaccharide (BSP) has received less attention, and its specific mechanism for ameliorating pulmonary fibrosis is completely unclear. AIMS OF THE STUDY In this study, we aim to assess BSP on the treatment of PF and explore potential mechanisms. MATERIALS AND METHODS BSP was successfully extracted and purified from Bletilla striata. The mechanisms were assessed in bleomycin-induced pulmonary fibrosis model and lung fibroblasts activated by transforming growth factor-β1 (TGF-β1). Histological analysis, immunofluorescence, Western blot and flow cytometry were used to explore the alterations after BSP intervention. RESULTS The results in vivo showed an anti-PF effect of BSP treatment, which reduced pathogenic damages. Furthermore, TGF-β1-induced abnormal migration and upregulated expression of collagen I (COL1A1), vimentin and α-smooth muscle actin (α-SMA) were suppressed by BSP in L929 cells. Moreover, the abnormal proliferation was retarded by inhibiting the cell cycle of G1 to S phase. Immunofluorescence assay showed that BSP activated autophagy and played an antifibrotic role by inhibiting the expression of p62 and phospho-mammalian target of rapamycin (p-mTOR). Last but not least, the suppression of TGF-β1/Smad signaling pathway was critical for BSP to perform therapeutic effects in vitro and in vivo. CONCLUSION The possible mechanisms were involved in improving ECM deposition, regulating cell migration and proliferation, and promoting cellular autophagy. Briefly, all of the above revealed that BSP might be a novel therapy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Siqi Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yang Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Yajing Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lili Zhou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Xinyao Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Lingyi Liu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| | - Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
8
|
Xu B, Huang M, Qi H, Xu H, Cai L. Tomatidine activates autophagy to improve lung injury and inflammation in sepsis by inhibiting NF-κB and MAPK pathways. Mol Genet Genomics 2024; 299:14. [PMID: 38400847 DOI: 10.1007/s00438-024-02109-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/29/2023] [Indexed: 02/26/2024]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening medical condition with high mortality and morbidity. Autophagy is involved in the pathophysiological process of sepsis-induced ALI, including inflammation, which indicates that regulating autophagy may be beneficial for this disease. Tomatidine, a natural compound abundant in unripe tomatoes, has been reported to have anti-inflammatory, anti-tumorigenic, and lipid-lowering effects. However, the biological functions and mechanisms of tomatidine in sepsis-induced ALI remain unknown. The principal objective of this study was to investigate the effect of tomatidine on sepsis-induced ALI. Cecal ligation and puncture (CLP) was used to induce septic lung injury in mice, and 10 mg/kg tomatidine was intraperitoneally injected into mice 2 h after the operation. The results of hematoxylin and eosin staining and assessment of lung edema and total protein levels in bronchoalveolar lavage fluid (BALF) demonstrated that tomatidine alleviated CLP-induced severe lung injuries such as hemorrhage, infiltration of inflammatory cells, and interstitial and alveolar edema in mice. Additionally, the levels of proinflammatory cytokines in BALF and lung tissues were measured by enzyme-linked immunosorbent assay (ELISA), and the results showed that tomatidine inhibited CLP-induced inflammatory damage to lungs. Moreover, the results of western blotting showed that tomatidine promoted autophagy during CLP-induced ALI. Mechanistically, immunofluorescence staining and western blotting were used to measure the protein levels of TLR4, phosphorylated NF-κB, phosphorylated IκBα, and phosphorylated MAPKs, showing that tomatidine inactivated NF-κB and MAPK signaling in lung tissues of CLP-induced ALI mice. In conclusion, tomatidine exerts protective effects against sepsis-induced severe damage to the lungs by inhibiting inflammation and activating autophagy in CLP-treated mice through inactivating the NF-κB and MAPK pathways, which may be an effective candidate for treating septic ALI.
Collapse
Affiliation(s)
- Bo Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui, 230000, China.
| | - Min Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 230000, China
| | - Hang Qi
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui, 230000, China
| | - Hongzhou Xu
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui, 230000, China
| | - Liang Cai
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui, 230000, China
| |
Collapse
|
9
|
Shi F, Cao J, Zhou D, Wang X, Yang H, Liu T, Chen Z, Zeng J, Du S, Yang L, Jia R, Zhang S, Zhang M, Guo Y, Lin X. Revealing the clinical effect and biological mechanism of acupuncture in COPD: A review. Biomed Pharmacother 2024; 170:115926. [PMID: 38035864 DOI: 10.1016/j.biopha.2023.115926] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND To provide new ideas for the clinical and mechanism research of acupuncture in the treatment of chronic obstructive pulmonary disease (COPD), this study systematically reviews clinical research and the progress of basic research of acupuncture in the treatment of COPD. METHODS PubMed and Web of Science databases were searched using acupuncture and COPD as keywords in the last 10 years, and the included literature was determined according to exclusion criteria. FINDINGS Acupuncture can relieve clinical symptoms, improve exercise tolerance, anxiety, and nutritional status, as well as hemorheological changes (blood viscosity), reduce the inflammatory response, and reduce the duration and frequency of COPD in patients with COPD. Mechanistically, acupuncture inhibits M1 macrophage activity, reduces neutrophil infiltration, reduces inflammatory factor production in alveolar type II epithelial cells, inhibits mucus hypersecretion of airway epithelial cells, inhibits the development of chronic inflammation in COPD, and slows tissue structure destruction. Acupuncture may control pulmonary COPD inflammation through the vagal-cholinergic anti-inflammatory, vagal-adrenomedullary-dopamine, vagal-dual-sensory nerve fiber-pulmonary, and CNS-hypothalamus-orexin pathways. Furthermore, acupuncture can increase endogenous cortisol levels by inhibiting the HPA axis, thus improving airway antioxidant capacity and reducing airway inflammation in COPD. In conclusion, the inhibition of the chronic inflammatory response is the key mechanism of acupuncture treatment for COPD.
Collapse
Affiliation(s)
- Fangyuan Shi
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiaojiao Cao
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Dan Zhou
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xue Wang
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haitao Yang
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tingting Liu
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihan Chen
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiaming Zeng
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Simin Du
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Yang
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruo Jia
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Siqi Zhang
- Ministry of Education, and State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, China
| | - Mingxing Zhang
- School of Intergrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xiaowei Lin
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research Center of Experimental Acupuncture Science, School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
10
|
Jiang Y, Gao S, Chen Z, Zhao X, Gu J, Wu H, Liao Y, Wang J, Chen W. Pyroptosis in septic lung injury: Interactions with other types of cell death. Biomed Pharmacother 2023; 169:115914. [PMID: 38000360 DOI: 10.1016/j.biopha.2023.115914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 11/26/2023] Open
Abstract
Sepsis is a life-threatening systemic inflammatory response syndrome caused by the host imbalanced response to infection. Lung injury is the most common complication of sepsis and one of the leading causes of patient death. Pyroptosis is a specific programmed cell death characterized by the release of inflammatory cytokines. Appropriate pyroptosis can reduce tissue damage and exert a protective effect against infection during sepsis. However, overactivated pyroptosis results in massive cell death, leading to septic shock, multiple organ dysfunction syndrome, and even an increased risk of secondary infection. Recent studies suggest that pyroptosis can interact with and cross-regulate other types of cell death programs to establish a complex network of cell death, which participates in the occurrence and development of septic lung injury. This review will focus on the interactions between pyroptosis and other types of cell death, including apoptosis, necroptosis, PANoptosis, NETosis, autophagy, and ferroptosis, to summarize the role of pyroptosis in sepsis-induced lung injury, and will discuss the potential therapeutic strategies of targeting pyroptosis during sepsis treatment.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai 201104, China; Department of Anesthesiology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201799, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 200032, China
| | - Shenjia Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai 201104, China; Department of Anesthesiology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201799, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 200032, China
| | - Zhaoyuan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai 201104, China; Department of Anesthesiology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201799, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 200032, China
| | - Xiaoqiang Zhao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiahui Gu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai 201104, China; Department of Anesthesiology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201799, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 200032, China
| | - Han Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai 201104, China; Department of Anesthesiology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201799, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 200032, China
| | - Yun Liao
- Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jun Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai 201104, China; Department of Anesthesiology, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai 201799, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai 200032, China.
| |
Collapse
|
11
|
Albano GD, Montalbano AM, Gagliardo R, Profita M. Autophagy/Mitophagy in Airway Diseases: Impact of Oxidative Stress on Epithelial Cells. Biomolecules 2023; 13:1217. [PMID: 37627282 PMCID: PMC10452925 DOI: 10.3390/biom13081217] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Autophagy is the key process by which the cell degrades parts of itself within the lysosomes. It maintains cell survival and homeostasis by removing molecules (particularly proteins), subcellular organelles, damaged cytoplasmic macromolecules, and by recycling the degradation products. The selective removal or degradation of mitochondria is a particular type of autophagy called mitophagy. Various forms of cellular stress (oxidative stress (OS), hypoxia, pathogen infections) affect autophagy by inducing free radicals and reactive oxygen species (ROS) formation to promote the antioxidant response. Dysfunctional mechanisms of autophagy have been found in different respiratory diseases such as chronic obstructive lung disease (COPD) and asthma, involving epithelial cells. Several existing clinically approved drugs may modulate autophagy to varying extents. However, these drugs are nonspecific and not currently utilized to manipulate autophagy in airway diseases. In this review, we provide an overview of different autophagic pathways with particular attention on the dysfunctional mechanisms of autophagy in the epithelial cells during asthma and COPD. Our aim is to further deepen and disclose the research in this direction to stimulate the develop of new and selective drugs to regulate autophagy for asthma and COPD treatment.
Collapse
Affiliation(s)
- Giusy Daniela Albano
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Section of Palermo, Via Ugo La Malfa 153, 90146 Palermo, Italy; (A.M.M.); (R.G.); (M.P.)
| | | | | | | |
Collapse
|
12
|
Feng L, Chen X, Huang Y, Zhang X, Zheng S, Xie N. Immunometabolism changes in fibrosis: from mechanisms to therapeutic strategies. Front Pharmacol 2023; 14:1243675. [PMID: 37576819 PMCID: PMC10412938 DOI: 10.3389/fphar.2023.1243675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Immune cells are essential for initiating and developing the fibrotic process by releasing cytokines and growth factors that activate fibroblasts and promote extracellular matrix deposition. Immunometabolism describes how metabolic alterations affect the function of immune cells and how inflammation and immune responses regulate systemic metabolism. The disturbed immune cell function and their interactions with other cells in the tissue microenvironment lead to the origin and advancement of fibrosis. Understanding the dysregulated metabolic alterations and interactions between fibroblasts and the immune cells is critical for providing new therapeutic targets for fibrosis. This review provides an overview of recent advances in the pathophysiology of fibrosis from the immunometabolism aspect, highlighting the altered metabolic pathways in critical immune cell populations and the impact of inflammation on fibroblast metabolism during the development of fibrosis. We also discuss how this knowledge could be leveraged to develop novel therapeutic strategies for treating fibrotic diseases.
Collapse
Affiliation(s)
- Lixiang Feng
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xingyu Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yujing Huang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaodian Zhang
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shaojiang Zheng
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Department of Pathology, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
13
|
Zhao Y, Wu J, Guan S, Xue T, Wei X, Cao D, Kong P, Zhang X. PIF1 Promotes Autophagy to Inhibit Chronic Hypoxia Induced Apoptosis of Pulmonary Artery Endothelial Cells. Int J Chron Obstruct Pulmon Dis 2023; 18:1319-1332. [PMID: 37396201 PMCID: PMC10312211 DOI: 10.2147/copd.s406453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/14/2023] [Indexed: 07/04/2023] Open
Abstract
Purpose Pulmonary artery hypertension (PAH) is a common complication of chronic obstructive pulmonary disease and obstructive sleep apnea/hypopnea syndrome worldwide. Pulmonary vascular alterations associated with PAH have multifactorial causes, in which endothelial cells play an important role. Autophagy is closely related to endothelial cell injury and the development of PAH. PIF1 is a multifunctional helicase crucial for cell survival. The present study investigated the effect of PIF1 on autophagy and apoptosis in human pulmonary artery endothelial cells (HPAECs) under chronic hypoxia stress. Methods Chronic hypoxia Gene expression profiling chip-assays identified the PIF1 gene as differentially expressed, which was verified by RT-qPCR analysis. Electron microscopy, immunofluorescence, and Western blotting were used to analyze autophagy and the expression of LC3 and P62. Apoptosis was analyzed using flow cytometry. Results Our study found that chronic hypoxia induces autophagy in HPAECs, and apoptosis was exacerbated by inhibiting autophagy. Levels of the DNA helicase PIF1 were increased in HPAECs after chronic hypoxia. PIF1 knockdown inhibited autophagy and promoted the apoptosis of HPAECs under chronic hypoxia stress. Conclusion Based on these findings, we conclude that PIF1 inhibits the apoptosis of HPAECs by accelerating the autophagy pathway. Therefore, PIF1 plays a crucial role in HPAEC dysfunction in chronic hypoxia-induced PAH and may be a potential target for the treatment of PAH.
Collapse
Affiliation(s)
- Yujing Zhao
- Department of the First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Juan Wu
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Shuai Guan
- Department of Pediatric Infectious Diseases, The First People’s Hospital of Datong, Datong, Shanxi, People’s Republic of China
| | - Ting Xue
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xiaolei Wei
- Department of the First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Dawei Cao
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Pengzhou Kong
- Department of Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xinri Zhang
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
14
|
Mao M, Song S, Li X, Lu J, Li J, Zhao W, Liu H, Liu J, Zeng B. Advances in epigenetic modifications of autophagic process in pulmonary hypertension. Front Immunol 2023; 14:1206406. [PMID: 37398657 PMCID: PMC10313199 DOI: 10.3389/fimmu.2023.1206406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Pulmonary hypertension is characterized by pulmonary arterial remodeling that results in increased pulmonary vascular resistance, right ventricular failure, and premature death. It is a threat to public health globally. Autophagy, as a highly conserved self-digestion process, plays crucial roles with autophagy-related (ATG) proteins in various diseases. The components of autophagy in the cytoplasm have been studied for decades and multiple studies have provided evidence of the importance of autophagic dysfunction in pulmonary hypertension. The status of autophagy plays a dynamic suppressive or promotive role in different contexts and stages of pulmonary hypertension development. Although the components of autophagy have been well studied, the molecular basis for the epigenetic regulation of autophagy is less understood and has drawn increasing attention in recent years. Epigenetic mechanisms include histone modifications, chromatin modifications, DNA methylation, RNA alternative splicing, and non-coding RNAs, which control gene activity and the development of an organism. In this review, we summarize the current research progress on epigenetic modifications in the autophagic process, which have the potential to be crucial and powerful therapeutic targets against the autophagic process in pulmonary hypertension development.
Collapse
Affiliation(s)
- Min Mao
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- National Health Commission (NHC) Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shasha Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Xin Li
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- National Health Commission (NHC) Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayao Lu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Jie Li
- Marketing Department, Shenzhen Reyson Biotechnology Co., Ltd, Shenzhen, China
- Nanjing Evertop Electronics Ltd., Nanjing, China
| | - Weifang Zhao
- Quality Management Department International Registration, North China Pharmaceutical Co., Ltd. (NCPC), Hebei Huamin Pharmaceutical Co., Ltd., Shijiazhuang, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- National Health Commission (NHC) Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jingxin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Bin Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
15
|
Yousefi P, Tabibzadeh A, Keyvani H, Esghaei M, Karampoor S, Razizadeh MH, Mousavizadeh L. The potential importance of autophagy genes expression profile dysregulation and ATG polymorphisms in COVID-19 pathogenesis. APMIS 2023; 131:161-169. [PMID: 36478304 PMCID: PMC9877785 DOI: 10.1111/apm.13286] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Autophagy is one of the important mechanisms in cell maintenance, which is considered associated with different pathological conditions such as viral infections. In this current study, the expression level and polymorphisms in some of the most important genes in the autophagy flux in COVID-19 patients were evaluated. This cross-sectional study was conducted among 50 confirmed COVID-19 patients and 20 healthy controls. The COVID-19 patients were divided into a severe group and a mild group according to their clinical features. The expression levels of ATG5, ATG16L1, LC3, and BECN1 were evaluated by the 2-∆∆CT method and beta-actin as the internal control. The polymorphisms of the ATG5 (rs506027, rs510432) and ATG16L1 (rs2241880 or T300A) were evaluated by the Sanger sequencing following the conventional PCR. The mean age of the included patients was 58.3 ± 17.9 and 22 (44%) were female. The expression levels of the LC3 were downregulated, while BECN1 and ATG16L1 genes represent an upregulation in COVID-19 patients. The polymorphism analysis revealed the ATG16L1 rs2241880 and AGT5 rs506027 polymorphism frequencies are statistically significantly different between COVID-19 and Healthy controls. The autophagy alteration represents an association with COVID-19 pathogenesis and severity. The current study is consistent with the alteration of autophagy elements in COVID-19 patients by mRNA expression-level evaluation. Furthermore, ATG16L1 rs2241880 and AGT5 rs506027 polymorphisms seem to be important in COVID-19 and are highly suggested for further investigations.
Collapse
Affiliation(s)
- Parastoo Yousefi
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Tabibzadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Keyvani
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Esghaei
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Leila Mousavizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Xiao J, Shen X, Kou R, Wang K, Zhai L, Ding L, Chen H, Mao C. Kirenol inhibits inflammation challenged by lipopolysaccharide through the AMPK-mTOR-ULK1 autophagy pathway. Int Immunopharmacol 2023; 116:109734. [PMID: 36706589 DOI: 10.1016/j.intimp.2023.109734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/03/2023] [Accepted: 01/11/2023] [Indexed: 01/27/2023]
Abstract
Kirenol is a bioactive substance isolated from Herba Siegesbeckiae. Although the anti-inflammatory activity of kirenol has been well documented, its role in autophagy remains unknown. The present study aimed to investigate the protective role of kirenol on inflammation challenged by lipopolysaccharide (LPS) in acute lung injury (ALI) cell and mouse models and unravel the underlying mechanisms, with a particular focus on autophagy. For this purpose, an ALI cell and mouse models were established, and the effects of kirenol on the expression of molecules related to inflammation and autophagy were examined. The present results revealed that kirenol could significantly inhibit inflammatory cytokines secretion in cells and in the mice injured by LPS; this effect may be attributed to enhanced autophagy as evidenced by the up-regulation of LC3-II and the down-regulation of p62 both in vitro and in vivo. Phosphorylated AMPK and ULK1 increased, while phosphorylated mTOR decreased in the kirenol-treated ALI cell model. Moreover, inhibition of autophagy using AMPK inhibitor or 3-MA or chloroquine (CQ) reversed the anti-inflammatory and autophagy-enhancement effects of kirenol exposure in vitro, indicating that kirenol could enhance autophagy by activating the AMPK-mTOR-ULK1 pathway. The results of RNA sequencing suggested that kirenol was strongly related to the biological functions of acute inflammatory response and the AMPK signaling pathway. Further in vivo ALI mouse model studies demonstrated the protective role of kirenol against lung inflammation, such as improved histopathology, decreased lung edema, and leukocyte infiltration were abolished by 3-MA. These findings implicate that kirenol can inhibit LPS-induced inflammation via the AMPK-mTOR-ULK1 autophagy pathway.
Collapse
Affiliation(s)
- Juan Xiao
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China
| | - Xiaofang Shen
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China
| | - Ruiming Kou
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China
| | - Ke Wang
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China
| | - Lihong Zhai
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China
| | - Lu Ding
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China
| | - Huabo Chen
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China.
| | - Chun Mao
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China; Medical College, Hubei University of Arts and Science, Xiangyang 441053, China.
| |
Collapse
|
17
|
A zebrafish HCT116 xenograft model to predict anandamide outcomes on colorectal cancer. Cell Death Dis 2022; 13:1069. [PMID: 36564370 PMCID: PMC9789132 DOI: 10.1038/s41419-022-05523-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Colon cancer is one of the leading causes of death worldwide. In recent years, cannabinoids have been extensively studied for their potential anticancer effects and symptom management. Several in vitro studies reported anandamide's (AEA) ability to block cancer cell proliferation and migration, but evidence from in vivo studies is still lacking. Thus, in this study, the effects of AEA exposure in zebrafish embryos transplanted with HCT116 cells were evaluated. Totally, 48 hpf xenografts were exposed to 10 nM AEA, 10 nM AM251, one of the cannabinoid 1 receptor (CB1) antagonist/inverse agonists, and to AEA + AM251, to verify the specific effect of AEA treatment. AEA efficacy was evaluated by confocal microscopy, which demonstrated that these xenografts presented a smaller tumor size, reduced tumor angiogenesis, and lacked micrometastasis formation. To gain deeper evidence into AEA action, microscopic observations were completed by molecular analyses. RNA seq performed on zebrafish transcriptome reported the downregulation of genes involved in cell proliferation, angiogenesis, and the immune system. Conversely, HCT116 cell transcripts resulted not affected by AEA treatment. In vitro HCT116 culture, in fact, confirmed that AEA exposure did not affect cell proliferation and viability, thus suggesting that the reduced tumor size mainly depends on direct effects on the fish rather than on the transplanted cancer cells. AEA reduced cell proliferation and tumor angiogenesis, as suggested by socs3 and pcnp mRNAs and Vegfc protein levels, and exerted anti-inflammatory activity, as indicated by the reduction of il-11a, mhc1uba, and csf3b mRNA. Of note, are the results obtained in groups exposed to AM251, which presence nullifies AEA's beneficial effects. In conclusion, this study promotes the efficacy of AEA in personalized cancer therapy, as suggested by its ability to drive tumor growth and metastasis, and strongly supports the use of zebrafish xenograft as an emerging model platform for cancer studies.
Collapse
|
18
|
Zhang Y, Zhang J, Fu Z. Role of autophagy in lung diseases and ageing. Eur Respir Rev 2022; 31:31/166/220134. [PMID: 36543345 PMCID: PMC9879344 DOI: 10.1183/16000617.0134-2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022] Open
Abstract
The lungs face ongoing chemical, mechanical, biological, immunological and xenobiotic stresses over a lifetime. Advancing age progressively impairs lung function. Autophagy is a "housekeeping" survival strategy involved in numerous physiological and pathological processes in all eukaryotic cells. Autophagic activity decreases with age in several species, whereas its basic activity extends throughout the lifespan of most animals. Dysregulation of autophagy has been proven to be closely related to the pathogenesis of several ageing-related pulmonary diseases. This review summarises the role of autophagy in the pathogenesis of pulmonary diseases associated with or occurring in the context of ageing, including acute lung injury, chronic obstructive pulmonary disease, asthma and pulmonary fibrosis, and describes its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China,Corresponding author: Zhiling Fu ()
| |
Collapse
|
19
|
TM9SF1 knockdown decreases inflammation by enhancing autophagy in a mouse model of acute lung injury. Heliyon 2022; 8:e12092. [PMID: 36561687 PMCID: PMC9763745 DOI: 10.1016/j.heliyon.2022.e12092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/06/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
TM9SF1 is a member of the TM9SF (Transmembrane 9 Superfamily Member) family, which usually has a long N-terminal extracellular region and nine transmembrane domains. TM9SF1's biological function and mechanisms in inflammation are yet unknown. Tm9sf1 was shown to be upregulated in the lung tissues of mice suffering from LPS-induced acute lung injury (ALI). Tm9sf1 knockout mice were studied, and it was shown that Tm9sf1 knockout significantly alleviated LPS-induced ALI, as evidenced by higher survival rate, improved pulmonary vascular permeability, decreased inflammatory cell infiltration, and downregulated inflammatory cytokines. TM9SF1 was also demonstrated to be a negative regulator of autophagy in the LPS-induced ALI model in vitro and in vivo. The autophagy inhibitor 3-MA could counteract the beneficial effects of Tm9sf1 knockout on ALI. Therefore, we discover for the first time the role and mechanism of TM9SF1 in LPS-induced ALI and establish a relationship between TM9SF1 regulated autophagy and ALI progression, which may provide novel targets for the treatment of ALI.
Collapse
|
20
|
Wei S, Xu T, Chen Y, Zhou K. Autophagy, cell death, and cytokines in K. pneumoniae infection: Therapeutic Perspectives. Emerg Microbes Infect 2022; 12:2140607. [DOI: 10.1080/22221751.2022.2140607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Sha Wei
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Tingting Xu
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
21
|
Millar MW, Fazal F, Rahman A. Therapeutic Targeting of NF-κB in Acute Lung Injury: A Double-Edged Sword. Cells 2022; 11:3317. [PMID: 36291185 PMCID: PMC9601210 DOI: 10.3390/cells11203317] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a devastating disease that can be caused by a variety of conditions including pneumonia, sepsis, trauma, and most recently, COVID-19. Although our understanding of the mechanisms of ALI/ARDS pathogenesis and resolution has considerably increased in recent years, the mortality rate remains unacceptably high (~40%), primarily due to the lack of effective therapies for ALI/ARDS. Dysregulated inflammation, as characterized by massive infiltration of polymorphonuclear leukocytes (PMNs) into the airspace and the associated damage of the capillary-alveolar barrier leading to pulmonary edema and hypoxemia, is a major hallmark of ALI/ARDS. Endothelial cells (ECs), the inner lining of blood vessels, are important cellular orchestrators of PMN infiltration in the lung. Nuclear factor-kappa B (NF-κB) plays an essential role in rendering the endothelium permissive for PMN adhesion and transmigration to reach the inflammatory site. Thus, targeting NF-κB in the endothelium provides an attractive approach to mitigate PMN-mediated vascular injury, not only in ALI/ARDS, but in other inflammatory diseases as well in which EC dysfunction is a major pathogenic mechanism. This review discusses the role and regulation of NF-κB in the context of EC inflammation and evaluates the potential and problems of targeting it as a therapy for ALI/ARDS.
Collapse
Affiliation(s)
| | | | - Arshad Rahman
- Department of Pediatrics (Neonatology), Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
22
|
Cheng X, Chen Q, Sun P. Natural phytochemicals that affect autophagy in the treatment of oral diseases and infections: A review. Front Pharmacol 2022; 13:970596. [PMID: 36091810 PMCID: PMC9461701 DOI: 10.3389/fphar.2022.970596] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/03/2022] [Indexed: 01/01/2023] Open
Abstract
Autophagy is a critical factor in eukaryotic evolution. Cells provide nutrition and energy during autophagy by destroying non-essential components, thereby allowing intracellular material conversion and managing temporary survival stress. Autophagy is linked to a variety of oral disorders, including the type and extent of oral malignancies. Furthermore, autophagy is important in lymphocyte formation, innate immunity, and the regulation of acquired immune responses. It is also required for immunological responses in the oral cavity. Knowledge of autophagy has aided in the identification and treatment of common oral disorders, most notably cancers. The involvement of autophagy in the oral immune system may offer a new understanding of the immune mechanism and provide a novel approach to eliminating harmful bacteria in the body. This review focuses on autophagy creation, innate and acquired immunological responses to autophagy, and the status of autophagy in microbial infection research. Recent developments in the regulatory mechanisms of autophagy and therapeutic applications in oral illnesses, particularly oral cancers, are also discussed. Finally, the relationship between various natural substances that may be used as medications and autophagy is investigated.
Collapse
Affiliation(s)
| | | | - Ping Sun
- *Correspondence: Ping Sun, ; Qianming Chen,
| |
Collapse
|
23
|
Fu Z, Wu X, Zheng F, Zhang Y. Activation of the AMPK-ULK1 pathway mediated protective autophagy by sevoflurane anesthesia restrains LPS-induced acute lung injury (ALI). Int Immunopharmacol 2022; 108:108869. [DOI: 10.1016/j.intimp.2022.108869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 11/05/2022]
|
24
|
Hu J, Ge S, Sun B, Ren J, Xie J, Zhu G. Comprehensive Analysis of Potential ceRNA Network and Different Degrees of Immune Cell Infiltration in Acute Respiratory Distress Syndrome. Front Genet 2022; 13:895629. [PMID: 35719385 PMCID: PMC9198558 DOI: 10.3389/fgene.2022.895629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/04/2022] [Indexed: 11/15/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a leading cause of death in critically ill patients due to hypoxemic respiratory failure. The specific pathogenesis underlying ARDS has not been fully elucidated. In this study, we constructed a triple regulatory network involving competing endogenous RNA (ceRNA) to investigate the potential mechanism of ARDS and evaluated the immune cell infiltration patterns in ARDS patients. Overall, we downloaded three microarray datasets that included 60 patients with sepsis-induced ARDS and 79 patients with sepsis alone from the public Gene Expression Omnibus (GEO) database and identified differentially expressed genes (DEGs, including 9 DElncRNAs, 9 DEmiRNAs, and 269 DEmRNAs) by R software. The DEGs were subjected to the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for functional enrichment analysis, and a protein–protein interaction (PPI) network was generated for uncovering interactive relationships among DEmRNAs. Then, a ceRNA network that contained 5 DElncRNAs, 7 DEmiRNAs, and 71 DEmRNAs was established according to the overlapping genes in both DEGs and predicted genes by public databases. Finally, we identified the TUG1/miR-140-5p/NFE2L2 pathway as the hub pathway in the whole network through Cytoscape. In addition, we evaluated the distribution of 22 subtypes of immune cells and recognized three differentially expressed immune cells in patients with sepsis-induced ARDS by “Cell Type Identification by Estimating Relative Subsets of Known RNA Transcripts (CIBERSORT)” algorithm, namely, naive B cells, regulatory T cells, and eosinophils. Correlations between differentially expressed immune cells and hub genes in the ceRNA network were also performed. In conclusion, we demonstrated a new potential regulatory mechanism underlying ARDS (the TUG1/miR-140-5p/NFE2L2 ceRNA regulatory pathway), which may help in further exploring the pathogenesis of ARDS.
Collapse
Affiliation(s)
- Jiaxin Hu
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shanhui Ge
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Borui Sun
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jianwei Ren
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jiang Xie
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Guangfa Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Alsayed HA, Mohammad HMF, Khalil CM, El-Kherbetawy MK, Elaidy SM. Autophagy modulation by irbesartan mitigates the pulmonary fibrotic alterations in bleomycin challenged rats: Comparative study with rapamycin. Life Sci 2022; 303:120662. [PMID: 35636582 DOI: 10.1016/j.lfs.2022.120662] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 01/07/2023]
Abstract
AIMS In pulmonary fibrosis, autophagy handles the maintenance of alveolar epithelial cells, prevents epithelial-mesenchymal transition (EMT), and controls collagen turnover. The mammalian target of rapamycin (mTOR) and its translational-dependent proteins are essential regulators of autophagy. Irbesartan (IRB) has earlier ameliorative effects in experimental pulmonary fibrosis. The current study aimed to explore therapeutic autophagy-modulated pulmonary fibrotic changes by IRB versus rapamycin (RAPA) in bleomycin (BLM)-challenged rats. MATERIALS AND METHODS A single intratracheal BLM dose at day (0), IRB in different doses (10, 20, and 40 mg/kg) or RAPA (2.5 mg/kg) was given daily for 14 continuous days. KEY FINDINGS IRB significantly diminished the fibrotic lung scores. Pulmonary levels of transforming growth factor (TGF)-β1 and hydroxyproline exhibited marked attenuation in IRB (40 mg/kg)-treated rats compared to other treated groups. IRB (40 mg/kg) was not significantly different from RAPA. It downregulated the fibrotic lung phosphorylated mammalian target of rapamycin (p-mTOR) levels and augmented lung Unc-51-like autophagy activating kinase 1 (ULK1), LC3-I and LC3-II more than IRB (10 and 20 mg/kg)-treated fibrotic groups. SIGNIFICANCE Autophagic effects via the mTOR signalling pathway may play a role in IRB's antifibrotic effects. Consideration of IRB as a therapeutic antifibrotic agent in pulmonary fibrosis needs further experimental and clinical long-term validation, especially in comorbid with primary hypertension, heart failure, and diabetic renal insults.
Collapse
Affiliation(s)
- Hadeer A Alsayed
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Hala M F Mohammad
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt; Central Laboratory, Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | - Cherine M Khalil
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt
| | | | - Samah M Elaidy
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, 41522 Ismailia, Egypt.
| |
Collapse
|
26
|
Shu Q, Zhou Y, Zhu Z, Chen X, Fang Q, Zhong L, Chen Z, Fang L. A Novel Risk Model Based on Autophagy-Related LncRNAs Predicts Prognosis and Indicates Immune Infiltration Landscape of Patients With Cutaneous Melanoma. Front Genet 2022; 13:885391. [PMID: 35571053 PMCID: PMC9101482 DOI: 10.3389/fgene.2022.885391] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
Cutaneous melanoma (CM) is a malignant tumor with a high incidence rate and poor prognosis. Autophagy plays an essential role in the development of CM; however, the role of autophagy-related long noncoding RNAs (lncRNAs) in this process remains unknown. Human autophagy-related genes were extracted from the Human Autophagy Gene Database and screened for autophagy-related lncRNAs using Pearson correlation. Multivariate Cox regression analysis was implemented to identify ten autophagy-related lncRNAs associated with prognosis, and a risk model was constructed. The Kaplan-Meier survival curve showed that the survival probability of the high-risk group was lower than that of the low-risk group. A novel predictive model was constructed to investigate the independent prognostic value of the risk model. The nomogram results showed that the risk score was an independent prognostic signature that distinguished it from other clinical characteristics. The immune infiltration landscape of the low-risk and high-risk groups was further investigated. The low-risk groups displayed higher immune, stromal, and ESTIMATE scores and lower tumor purity. The CIBERSORT and single sample gene set enrichment analysis (ssGSEA) algorithms indicated a notable gap in immune cells between the low- and high-risk groups. Ten autophagy-related lncRNAs were significantly correlated with immune cells. Finally, Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) results demonstrated that autophagy-related lncRNA-mediated and immune-related signaling pathways are crucial factors in regulating CM. Altogether, these data suggest that constructing a risk model based on ten autophagy-related lncRNAs can accurately predict prognosis and indicate the tumor microenvironment of patients with CM. Thus, our study provides a new perspective for the future clinical treatment of CM.
Collapse
Affiliation(s)
- Qi Shu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yi Zhou
- Department of Pharmacy, First People’s Hospital of Linping District, Hangzhou, China
| | - Zhengjie Zhu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Xi Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qilu Fang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Like Zhong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Zhuo Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Luo Fang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
27
|
Zhang Y, Zhang J, Fu Z. Molecular hydrogen is a potential protective agent in the management of acute lung injury. Mol Med 2022; 28:27. [PMID: 35240982 PMCID: PMC8892414 DOI: 10.1186/s10020-022-00455-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome, which is a more severe form of ALI, are life-threatening clinical syndromes observed in critically ill patients. Treatment methods to alleviate the pathogenesis of ALI have improved to a great extent at present. Although the efficacy of these therapies is limited, their relevance has increased remarkably with the ongoing pandemic caused by the novel coronavirus disease 2019 (COVID-19), which causes severe respiratory distress syndrome. Several studies have demonstrated the preventive and therapeutic effects of molecular hydrogen in the various diseases. The biological effects of molecular hydrogen mainly involve anti-inflammation, antioxidation, and autophagy and cell death modulation. This review focuses on the potential therapeutic effects of molecular hydrogen on ALI and its underlying mechanisms and aims to provide a theoretical basis for the clinical treatment of ALI and COVID-19.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
28
|
Zheng L, Wen L, Lei W, Ning Z. Added value of systemic inflammation markers in predicting pulmonary infection in stroke patients: A retrospective study by machine learning analysis. Medicine (Baltimore) 2021; 100:e28439. [PMID: 34967381 PMCID: PMC8718201 DOI: 10.1097/md.0000000000028439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 12/07/2021] [Indexed: 01/05/2023] Open
Abstract
Exploring candidate markers to predict the clinical outcomes of pulmonary infection in stroke patients have a high unmet need. This study aimed to develop machine learning (ML)-based predictive models for pulmonary infection.Between January 2008 and April 2021, a retrospective analysis of 1397 stroke patients who had CT angiography from skull to diaphragm (including CT of the chest) within 24 hours of symptom onset. A total of 21 variables were included, and the prediction model of pulmonary infection was established by multiple ML-based algorithms. Risk factors for pulmonary infection were determined by the feature selection method. Area under the curve (AUC) and decision curve analysis were used to determine the model with the best resolution and to assess the net clinical benefits associated with the use of predictive models, respectively.A total of 889 cases were included in this study as a training group, while 508 cases were as a validation group. The feature selection indicated the top 6 predictors were procalcitonin, C-reactive protein, soluble interleukin-2 receptor, consciousness disorder, dysphagia, and invasive procedure. The AUCs of the 5 models ranged from 0.78 to 0.87 in the training cohort. When the ML-based models were applied to the validation set, the results also remained reconcilable, and the AUC was between 0.891 and 0.804. The decision curve analysis also showed performed better than positive line and negative line, indicating the favorable predictive performance and clinical values of the models.By incorporating clinical characteristics and systemic inflammation markers, it is feasible to develop ML-based models for the presence and consequences of signs of pulmonary infection in stroke patients, and the use of the model may be greatly beneficial to clinicians in risk stratification and management decisions.
Collapse
Affiliation(s)
- Lv Zheng
- Department of Rehabilitation, Shenzhen Longgang Central Hospital, Shenzhen, China
| | - Lv Wen
- Department of Rehabilitation, Shenzhen Longgang Central Hospital, Shenzhen, China
| | - Wang Lei
- Department of Rehabilitation, Shenzhen Longgang Central Hospital, Shenzhen, China
| | - Zhang Ning
- Department of Rehabilitation, First Affiliated Hospital of Heilongjiang University of Chinese medicine, Harbin, China
| |
Collapse
|
29
|
Xue X, Meng L, Cai H, Sun Y, Zhang Y, Li H, Kang Y, Zhou B, Shang F, Guan W, Zhang L, Chen X, Zhang L. Xuanfei Pingchuan Capsules Ameliorate Autophagy in Human Bronchial Epithelial Cells by Inhibiting p38 Phosphorylation. Front Pharmacol 2021; 12:748234. [PMID: 34925010 PMCID: PMC8678282 DOI: 10.3389/fphar.2021.748234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Background: This study aimed to investigate the protective effect of Xuanfei Pingchuan Capsules (XFPC) on autophagy and p38 phosphorylation in human bronchial epithelial (HBE) cells induced by cigarette smoke extract (CSE). Methods: HBE cells were divided into five groups: blank, CSE, low XFPC dose (XFPC-L), medium XFPC dose (XFPC-M), and high XFPC dose (XFPC-H). HBE cells were induced by CSE to establish a cell model for chronic obstructive pulmonary disease, and different doses of XFPC medicated serum were used to treat the cells. The Cell Counting Kit-8 was used to detect cell viability. Flow cytometry was used to detect cell apoptosis. Fluorescence microscopy and the expression level of microtubule-associated protein light chain 3 (LC3)-II in immunohistochemical method were used to observe autophagy in cells. Western blot was used to detect the protein expression level of p38, phospho-p38 (p-p38), LC3-I, LC3-II and Beclin 1. Real-time polymerase chain reaction was used to detect the expression of LC3-I, LC3-II and Beclin 1 on mRNA level. Results: Compared with the blank group, the cell viability of the CSE group was significantly decreased, and apoptosis and the level of autophagy in cells were significantly increased. The mRNA and protein expression of LC3-I, LC3-II, Beclin 1 and the protein level of p-p38 were significantly increased in the CSE-HBE cells. Compared to the CSE group, the different doses of XFPC medicated serum increased cell viability, decreased cell apoptosis, and inhibited mRNA and protein expression of LC3-I, LC3-II, Beclin 1 and protein level of p-p38. These results were especially observed in the group XFPC-H. After adding a p38 agonist, the therapeutic effect of XFPC on cell viability and autophagy was suppressed. Conclusion: XFPC significantly increased cell viability in a CSE-induced HBE cell model for chronic obstructive pulmonary disease through inhibiting the level of autophagy mediated by phosphorylation of p38.
Collapse
Affiliation(s)
- Xiaoming Xue
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Lihong Meng
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Hongyu Cai
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Yaoqin Sun
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Ye Zhang
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Hao Li
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Yu Kang
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Bobo Zhou
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Fang Shang
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Wei Guan
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Li Zhang
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Xu Chen
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| | - Luodan Zhang
- Department of Respiration, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, China
| |
Collapse
|
30
|
Hu Y, He T, Zhu J, Wang X, Tong J, Li Z, Dong J. The Link between Circadian Clock Genes and Autophagy in Chronic Obstructive Pulmonary Disease. Mediators Inflamm 2021; 2021:2689600. [PMID: 34733115 PMCID: PMC8560276 DOI: 10.1155/2021/2689600] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/08/2021] [Indexed: 11/29/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a progressive respiratory disease, is characterized by the alveolar epithelium injury and persistent airway inflammation. It is documented that oscillation and dysregulated expression of circadian clock genes, like Bmal1, Per1, and Per2, involved in COPD pathogenies, including chronic inflammation and imbalanced autophagy level, and targeting the associations of circadian rhythm and autophagy is promising strategies in the management and treatment of COPD. Herein, we reviewed the mechanisms of the circadian clock and the unbalance of the autophagic level in COPD, as well as the link between the two, so as to provide further theoretical bases for the study on the pathogenesis of COPD.
Collapse
Affiliation(s)
- Yuedi Hu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1, Qianjiang Road, Hefei City, Anhui Province, China
| | - Tiantian He
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1, Qianjiang Road, Hefei City, Anhui Province, China
| | - Jie Zhu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, No. 1, Qianjiang Road, Hefei City, Anhui Province, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
- Institute of Traditional Chinese Medicine Prevention and Control on Respiratory Disease, Anhui Academy of Chinese Medicine, No. 117, Meishan Road, Hefei City, Anhui Province, China
| | - Xiaole Wang
- Institute of Traditional Chinese Medicine Prevention and Control on Respiratory Disease, Anhui Academy of Chinese Medicine, No. 117, Meishan Road, Hefei City, Anhui Province, China
| | - Jiabing Tong
- Institute of Traditional Chinese Medicine Prevention and Control on Respiratory Disease, Anhui Academy of Chinese Medicine, No. 117, Meishan Road, Hefei City, Anhui Province, China
- Department of Respiratory Medicine, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Meishan Road, Hefei City, Anhui Province, China
| | - Zegeng Li
- Institute of Traditional Chinese Medicine Prevention and Control on Respiratory Disease, Anhui Academy of Chinese Medicine, No. 117, Meishan Road, Hefei City, Anhui Province, China
- Department of Respiratory Medicine, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Meishan Road, Hefei City, Anhui Province, China
| | - Jingcheng Dong
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
31
|
lncRNA-SNHG14 Plays a Role in Acute Lung Injury Induced by Lipopolysaccharide through Regulating Autophagy via miR-223-3p/Foxo3a. Mediators Inflamm 2021; 2021:7890288. [PMID: 34539244 PMCID: PMC8443345 DOI: 10.1155/2021/7890288] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/05/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
lncRNAs play important roles in lipopolysaccharide- (LPS-) induced acute lung injury. But the mechanism still needs further research. In the present study, we investigate the functional role of the lncRNA-SNHG14/miR-223-3p/Foxo3a pathway in LPS-induced ALI and tried to confirm its regulatory effect on autophagy. Transcriptomic profile changes were identified by RNA-seq in LPS-treated alveolar type II epithelial cells. The expression changes of lncRNA-SNHG14/miR-223-3p/Foxo3a were confirmed using qRT-PCR and west blot. The binding relationship of lncRNA-SNHG14/miR-223-3p/and miR-223-3p/Foxo3a was verified using dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays. Using gain-of-function or loss-of-function approaches, the effect of lncRNA-SNHG14/miR-223-3p/Foxo3a was investigated in LPS-induced acute lung injury mice model and in vitro. Increasing of lncRNA-SNHG14 and Foxo3a with reducing miR-223-3p was found in LPS-treated A549 cells and lung tissue collected from the LPS-induced ALI model. lncRNA-SNHG14 inhibited miR-223-3p but promoted Foxo3a expression as a ceRNA. Artificially changes of lncRNA-SNHG14/miR-223-3p/Foxo3a pathway promoted or protected cell injury from LPS in vivo and in vitro. Autophagy activity could be influenced by lncRNA-SNHG14/miR-223-3p/Foxo3a pathway in cells with or without LPS treatment. In conclusion, aberrant expression changes of lncRNA-SNHG14 participated alveolar type II epithelial cell injury and acute lung injury induced by LPS through regulating autophagy. One underlying mechanism is that lncRNA-SNHG14 regulated autophagy by controlling miR-223-3p/Foxo3a as a ceRNA. It suggested that lncRNA-SNHG14 may serve as a potential therapeutic target for patients with sepsis-induced ALI.
Collapse
|
32
|
Rivero CV, Martínez SJ, Novick P, Cueto JA, Salassa BN, Vanrell MC, Li X, Labriola CA, Polo LM, Engman DM, Clos J, Romano PS. Repurposing Carvedilol as a Novel Inhibitor of the Trypanosoma cruzi Autophagy Flux That Affects Parasite Replication and Survival. Front Cell Infect Microbiol 2021; 11:657257. [PMID: 34476220 PMCID: PMC8406938 DOI: 10.3389/fcimb.2021.657257] [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: 01/22/2021] [Accepted: 05/20/2021] [Indexed: 11/13/2022] Open
Abstract
T. cruzi, the causal agent of Chagas disease, is a parasite able to infect different types of host cells and to persist chronically in the tissues of human and animal hosts. These qualities and the lack of an effective treatment for the chronic stage of the disease have contributed to the durability and the spread of the disease around the world. There is an urgent necessity to find new therapies for Chagas disease. Drug repurposing is a promising and cost-saving strategy for finding new drugs for different illnesses. In this work we describe the effect of carvedilol on T. cruzi. This compound, selected by virtual screening, increased the accumulation of immature autophagosomes characterized by lower acidity and hydrolytic properties. As a consequence of this action, the survival of trypomastigotes and the replication of epimastigotes and amastigotes were impaired, resulting in a significant reduction of infection and parasite load. Furthermore, carvedilol reduced the whole-body parasite burden peak in infected mice. In summary, in this work we present a repurposed drug with a significant in vitro and in vivo activity against T. cruzi. These data in addition to other pharmacological properties make carvedilol an attractive lead for Chagas disease treatment.
Collapse
Affiliation(s)
- Cynthia Vanesa Rivero
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET- Universidad Nacional de Cuyo, Mendoza, Argentina.,Leishmaniasis Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Santiago José Martínez
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET- Universidad Nacional de Cuyo, Mendoza, Argentina.,Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Paul Novick
- Department of Chemistry, Stanford University, San Francisco, CA, United States
| | - Juan Agustín Cueto
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Betiana Nebaí Salassa
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Cristina Vanrell
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Xiaomo Li
- Leishmaniasis Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Carlos Alberto Labriola
- Laboratorio de Biología estructural y celular, Fundación Instituto Leloir (FIL-CONICET), Buenos Aires, Argentina
| | - Luis Mariano Polo
- Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET- Universidad Nacional de Cuyo, Mendoza, Argentina
| | - David M Engman
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Joachim Clos
- Leishmaniasis Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Patricia Silvia Romano
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora - Instituto de Histología y Embriología "Dr. Mario H. Burgos", IHEM-CONICET- Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
33
|
Htwe YM, Wang H, Belvitch P, Meliton L, Bandela M, Letsiou E, Dudek SM. Group V Phospholipase A 2 Mediates Endothelial Dysfunction and Acute Lung Injury Caused by Methicillin-Resistant Staphylococcus Aureus. Cells 2021; 10:1731. [PMID: 34359901 PMCID: PMC8304832 DOI: 10.3390/cells10071731] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/25/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022] Open
Abstract
Lung endothelial dysfunction is a key feature of acute lung injury (ALI) and clinical acute respiratory distress syndrome (ARDS). Previous studies have identified the lipid-generating enzyme, group V phospholipase A2 (gVPLA2), as a mediator of lung endothelial barrier disruption and inflammation. The current study aimed to determine the role of gVPLA2 in mediating lung endothelial responses to methicillin-resistant Staphylococcus aureus (MRSA, USA300 strain), a major cause of ALI/ARDS. In vitro studies assessed the effects of gVPLA2 inhibition on lung endothelial cell (EC) permeability after exposure to heat-killed (HK) MRSA. In vivo studies assessed the effects of intratracheal live or HK-MRSA on multiple indices of ALI in wild-type (WT) and gVPLA2-deficient (KO) mice. In vitro, HK-MRSA increased gVPLA2 expression and permeability in human lung EC. Inhibition of gVPLA2 with either the PLA2 inhibitor, LY311727, or with a specific monoclonal antibody, attenuated the barrier disruption caused by HK-MRSA. LY311727 also reduced HK-MRSA-induced permeability in mouse lung EC isolated from WT but not gVPLA2-KO mice. In vivo, live MRSA caused significantly less ALI in gVPLA2 KO mice compared to WT, findings confirmed by intravital microscopy assessment in HK-MRSA-treated mice. After targeted delivery of gVPLA2 plasmid to lung endothelium using ACE antibody-conjugated liposomes, MRSA-induced ALI was significantly increased in gVPLA2-KO mice, indicating that lung endothelial expression of gVPLA2 is critical in vivo. In summary, these results demonstrate an important role for gVPLA2 in mediating MRSA-induced lung EC permeability and ALI. Thus, gVPLA2 may represent a novel therapeutic target in ALI/ARDS caused by bacterial infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Steven M. Dudek
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (Y.M.H.); (H.W.); (P.B.); (L.M.); (M.B.); (E.L.)
| |
Collapse
|
34
|
Sefiani A, Geoffroy CG. The Potential Role of Inflammation in Modulating Endogenous Hippocampal Neurogenesis After Spinal Cord Injury. Front Neurosci 2021; 15:682259. [PMID: 34220440 PMCID: PMC8249862 DOI: 10.3389/fnins.2021.682259] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Currently there are approximately 291,000 people suffering from a spinal cord injury (SCI) in the United States. SCI is associated with traumatic changes in mobility and neuralgia, as well as many other long-term chronic health complications, including metabolic disorders, diabetes mellitus, non-alcoholic steatohepatitis, osteoporosis, and elevated inflammatory markers. Due to medical advances, patients with SCI survive much longer than previously. This increase in life expectancy exposes them to novel neurological complications such as memory loss, cognitive decline, depression, and Alzheimer's disease. In fact, these usually age-associated disorders are more prevalent in people living with SCI. A common factor of these disorders is the reduction in hippocampal neurogenesis. Inflammation, which is elevated after SCI, plays a major role in modulating hippocampal neurogenesis. While there is no clear consensus on the mechanism of the decline in hippocampal neurogenesis and cognition after SCI, we will examine in this review how SCI-induced inflammation could modulate hippocampal neurogenesis and provoke age-associated neurological disorders. Thereafter, we will discuss possible therapeutic options which may mitigate the influence of SCI associated complications on hippocampal neurogenesis.
Collapse
|
35
|
Bo L, Li Y, Liu W, Jin F, Li C. Selective inhibition of JNK mitochondrial location is protective against seawater inhalation‑induced ALI/ARDS. Mol Med Rep 2021; 24:515. [PMID: 34013361 PMCID: PMC8138518 DOI: 10.3892/mmr.2021.12154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/21/2021] [Indexed: 02/04/2023] Open
Abstract
Localization of phosphorylated (p)‑JNK to the mitochondria can lead to functional mitochondrial disorder, resulting in a decrease in energy supply and membrane potential, as well as an increase in reactive oxygen species production and apoptosis. JNK is involved in the occurrence of acute lung injury (ALI), and activation of the JNK pathway is one of the crucial factors resulting in injury. The aim of the present study was to investigate whether the JNK‑mitochondria (mitoJNK) location participated in the occurrence of ALI and acute respiratory distress syndrome (ALI/ARDS). The present study examined the activation of the JNK pathway, the content of JNK located on the mitochondria and the treatment effects of a cell‑permeable peptide Tat‑SabKIM1, which can selectively inhibit the location of JNK on mitochondria. The expression levels of proteins were detected by western blot analysis. Lung injuries were evaluated by histological examination, wet‑to‑dry weight ratios, and H2O2 and malondialdehyde concentrations in the lung tissues. Lung cells apoptosis was evaluated using TUNEL assay. The results demonstrated that JNK was phosphorylated and activated during seawater inhalation‑induced ALI/ARDS, not only in the routine JNK pathway but also in the mitoJNK pathway. It was also found that Tat‑SabKIM1 could specifically inhibit JNK localization to mitochondria and the activation of mitoJNK signaling. Furthermore, Tat‑SabKIM1 could inhibit Bcl‑2‑regulated autophagy and mitochondria‑mediated apoptosis. In conclusion, mitoJNK localization disrupted the normal physiological functions of the mitochondria during ALI/ARDS, and selective inhibition of JNK and mitochondrial SH3BP5 (also known as Sab) binding with Tat‑SabKIM1 can block deterioration from ALI/ARDS.
Collapse
Affiliation(s)
- Liyan Bo
- Department of Respiratory and Critical Care Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Yanyan Li
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Wei Liu
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Faguang Jin
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Congcong Li
- Department of Respiratory and Critical Care Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
36
|
Ding L, Yang J, Zhang C, Zhang X, Gao P. Neutrophils Modulate Fibrogenesis in Chronic Pulmonary Diseases. Front Med (Lausanne) 2021; 8:616200. [PMID: 33987189 PMCID: PMC8110706 DOI: 10.3389/fmed.2021.616200] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammatory pulmonary diseases are characterized by recurrent and persistent inflammation of the airways, commonly associated with poor clinical outcomes. Although their etiologies vary tremendously, airway neutrophilia is a common feature of these diseases. Neutrophils, as vital regulators linking innate and adaptive immune systems, are a double-edged sword in the immune response of the lung involving mechanisms such as phagocytosis, degranulation, neutrophil extracellular trap formation, exosome secretion, release of cytokines and chemokines, and autophagy. Although neutrophils serve as strong defenders against extracellular pathogens, neutrophils and their components can trigger various cascades leading to inflammation and fibrogenesis. Here, we review current studies to elucidate the versatile roles of neutrophils in chronic pulmonary inflammatory diseases and describe the common pathogenesis of these diseases. This may provide new insights into therapeutic strategies for chronic lung diseases.
Collapse
Affiliation(s)
- Lili Ding
- Department of Intensive Care Unit, The First Hospital of Jilin University, Changchun, China
| | - Juan Yang
- Department of Intensive Care Unit, The First Hospital of Jilin University, Changchun, China
| | - Chunmei Zhang
- Intensive Care Unit of Emergency Department, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiuna Zhang
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital, Jilin University, Changchun, China
| | - Pujun Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
37
|
Wen X, Xiaoyue D, Longkun D, Yue X, Man Y, Min Z, Liang W, Chengxue Y, Huaxi X. Three main short-chain fatty acids inhibit the activation of THP-1 cells by Mycoplasma pneumoniae. Biosci Biotechnol Biochem 2021; 85:923-930. [PMID: 33590852 DOI: 10.1093/bbb/zbaa110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022]
Abstract
The overactivation of macrophages causes chronic inflammatory diseases. Short-chain fatty acids (SCFAs), potential drugs for clinical treatment, are modulators of macrophage inflammatory reaction. Therefore, the modulation of macrophage-mediated cell activity is expected to become a new therapeutic strategy for inflammatory diseases caused by Mycoplasma pneumoniae. In this study, 2 kinds of SCFAs (propionate and butyrate) were found to have anti-inflammatory effects in M. pneumoniae-stimulated THP-1 cells inflammatory. They inhibited the expressions of IL-4, IL-6, ROS, and NLRP3 inflammasome, while enhancing the expressions of IL-10 and IFN-γ. Our study revealed these 2 agents to repress transcriptional activities of NF-κB, which are important modulators of inflammation. Meanwhile, SCFAs can significantly enhance the autophagy induced by M. pneumoniae. Considering that SCFAs have few side effects, they might be the promising adjuvant therapy for the prevention and/or treatment of various inflammatory diseases.
Collapse
Affiliation(s)
- Xia Wen
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Dai Xiaoyue
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ding Longkun
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xi Yue
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yan Man
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhang Min
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wu Liang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yi Chengxue
- School of Medical Technology, Zhenjiang College, Zhenjiang, China
| | - Xu Huaxi
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
38
|
Rao L, De La Rosa I, Xu Y, Sha Y, Bhattacharya A, Holtzman MJ, Gilbert BE, Eissa NT. Pseudomonas aeruginosa survives in epithelia by ExoS-mediated inhibition of autophagy and mTOR. EMBO Rep 2021; 22:e50613. [PMID: 33345425 PMCID: PMC7857434 DOI: 10.15252/embr.202050613] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
One major factor that contributes to the virulence of Pseudomonas aeruginosa is its ability to reside and replicate unchallenged inside airway epithelial cells. The mechanism by which P. aeruginosa escapes destruction by intracellular host defense mechanisms, such as autophagy, is not known. Here, we show that the type III secretion system effector protein ExoS facilitates P. aeruginosa survival in airway epithelial cells by inhibiting autophagy in host cells. Autophagy inhibition is independent of mTOR activity, as the latter is also inhibited by ExoS, albeit by a different mechanism. Deficiency of the critical autophagy gene Atg7 in airway epithelial cells, both in vitro and in mouse models, greatly enhances the survival of ExoS-deficient P. aeruginosa but does not affect the survival of ExoS-containing bacteria. The inhibitory effect of ExoS on autophagy and mTOR depends on the activity of its ADP-ribosyltransferase domain. Inhibition of mTOR is caused by ExoS-mediated ADP ribosylation of RAS, whereas autophagy inhibition is due to the suppression of autophagic Vps34 kinase activity.
Collapse
Affiliation(s)
- Lang Rao
- Department of MedicineBaylor College of MedicineHoustonTXUSA
- Veterans Administration Long Beach Health Care System and University of California at IrvineIrvineCAUSA
- Southern California Institute for Research and EducationLong BeachCAUSA
| | | | - Yi Xu
- Department of MedicineBaylor College of MedicineHoustonTXUSA
| | - Youbao Sha
- Department of MedicineBaylor College of MedicineHoustonTXUSA
| | | | - Michael J Holtzman
- Department of Internal MedicineWashington University School of MedicineSt. LouisMOUSA
| | - Brian E Gilbert
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTXUSA
| | - N Tony Eissa
- Department of MedicineBaylor College of MedicineHoustonTXUSA
- Veterans Administration Long Beach Health Care System and University of California at IrvineIrvineCAUSA
| |
Collapse
|
39
|
Yu Q, Yang S, Li Z, Zhu Y, Li Z, Zhang J, Li C, Feng F, Wang W, Zhang Q. The relationship between endoplasmic reticulum stress and autophagy in apoptosis of BEAS-2B cells induced by cigarette smoke condensate. Toxicol Res (Camb) 2021; 10:18-28. [PMID: 33613969 DOI: 10.1093/toxres/tfaa095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/02/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022] Open
Abstract
Cigarette smoke (CS) is one of the severe risk factors for the development of the pulmonary disease. However, the underlying mechanisms, especially the CS-induced the human bronchial epithelial cells (BEAS-2B) apoptosis related to endoplasmic reticulum stress (ERS) and autophagy, remains to be studied. This study aims to investigate the relationship between ERS and autophagy in apoptosis induced by CS condensate (CSC). BEAS-2B cells were stimulated with 0.02, 0.04 and 0.08 mg/ml CSC for 24 h to detect the ERS, autophagy and apoptosis. Then, ERS and autophagy of BEAS-2B cells were inhibited, respectively, by using 4-PBA and 3-MA, and followed by CSC treatment. The results showed that CSC decreased cell viability, increased cell apoptosis, elevated cleaved-caspase 3/pro-caspase 3 ratio and Bax expressions, but decreased Bcl-2 expressions. The GRP78 and CHOP expressions and LC3-II/LC3-I ratio were dose-dependently increased. The structure of the endoplasmic reticulum was abnormal and the number of autolysosomes was increased in BEAS-2B cells after CSC stimulation. The LC3-II/LC3-I ratio was decreased after ERS inhibition with 4-PBA, but GRP78 and CHOP expressions were enhanced after autophagy inhibition with 3-MA. CSC-induced apoptosis was further increased, Bax expressions and cleaved-caspase 3/pro-caspase 3 ratio were improved, but Bcl-2 expressions were decreased after 3-MA or 4-PBA treatment. In conclusion, the study indicates that ERS may repress apoptosis of BEAS-2B cells induced by CSC via activating autophagy, but autophagy relieves ERS in a negative feedback. This study provides better understanding and experimental support on the underlying mechanisms of pulmonary disease stimulated by CS.
Collapse
Affiliation(s)
- Qi Yu
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Sa Yang
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Zhongqiu Li
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Yonghang Zhu
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Zhenkai Li
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Jiatong Zhang
- Department of Disease Control and Prevention, Hospital of Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Chunyang Li
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Feifei Feng
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Wei Wang
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| | - Qiao Zhang
- Department of Toxicology, College of Public Health, Zhengzhou University, 100 Kexue Ave, Zhongyuan District, Zhengzhou 450001, China
| |
Collapse
|
40
|
Pehote G, Vij N. Autophagy Augmentation to Alleviate Immune Response Dysfunction, and Resolve Respiratory and COVID-19 Exacerbations. Cells 2020; 9:cells9091952. [PMID: 32847034 PMCID: PMC7565665 DOI: 10.3390/cells9091952] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
The preservation of cellular homeostasis requires the synthesis of new proteins (proteostasis) and organelles, and the effective removal of misfolded or impaired proteins and cellular debris. This cellular homeostasis involves two key proteostasis mechanisms, the ubiquitin proteasome system and the autophagy–lysosome pathway. These catabolic pathways have been known to be involved in respiratory exacerbations and the pathogenesis of various lung diseases, such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and coronavirus disease-2019 (COVID-19). Briefly, proteostasis and autophagy processes are known to decline over time with age, cigarette or biomass smoke exposure, and/or influenced by underlying genetic factors, resulting in the accumulation of misfolded proteins and cellular debris, elevating apoptosis and cellular senescence, and initiating the pathogenesis of acute or chronic lung disease. Moreover, autophagic dysfunction results in an impaired microbial clearance, post-bacterial and/or viral infection(s) which contribute to the initiation of acute and recurrent respiratory exacerbations as well as the progression of chronic obstructive and restrictive lung diseases. In addition, the autophagic dysfunction-mediated cystic fibrosis transmembrane conductance regulator (CFTR) immune response impairment further exacerbates the lung disease. Recent studies demonstrate the therapeutic potential of novel autophagy augmentation strategies, in alleviating the pathogenesis of chronic obstructive or restrictive lung diseases and exacerbations such as those commonly seen in COPD, CF, ALI/ARDS and COVID-19.
Collapse
Affiliation(s)
- Garrett Pehote
- Michigan State University College of Osteopathic Medicine, East Lansing, MI 48823, USA;
| | - Neeraj Vij
- Department of Pediatrics and Pulmonary Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- PRECISION THERANOSTICS INC, Baltimore, MD 21202, USA
- VIJ BIOTECH, Baltimore, MD 21202, USA
- Correspondence: or ; Tel.: +1-240-623-0757
| |
Collapse
|
41
|
Zhang L, Ma C, Wang X, Bai J, He S, Zhang J, Xin W, Li Y, Jiang Y, Li J, Zhu D. MicroRNA-874-5p regulates autophagy and proliferation in pulmonary artery smooth muscle cells by targeting Sirtuin 3. Eur J Pharmacol 2020; 888:173485. [PMID: 32805255 DOI: 10.1016/j.ejphar.2020.173485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 11/29/2022]
Abstract
Autophagy is a major cause of pathological vascular remodeling under hypoxic pulmonary hypertension (PH). Sirtuin 3 (Sirt 3) has recently been reported to be involved in the regulation of autophagy, however, its role as an autophagy regulator during hypoxic PH, particularly the molecular mechanism, remains poorly understood. In the present study, Western blot, immunohistochemistry, immunofluorescence, bromodeoxyuridine incorporation and cell cycle analyses were performed to elucidate the underlying mechanism of hypoxia-induced autophagy and cell proliferation with respect to Sirt 3. We observed that the Sirt 3 expression was decreased under hypoxia and that Sirt 3 overexpression significantly inhibited the effects of hypoxia on autophagy. Next, we investigated the mechanistic role of microRNAs in Sirt 3-associated autophagy under hypoxic conditions, with luciferase reporter, microscale thermophoresis and RNA immunoprecipitation assays, results confirming that Sirt 3 is a direct target of miR-874-5p. Furthermore, miR-874-5p was upregulated following hypoxia, and miR-874-5p depletion in turn inhibited autophagy and consequently suppressed abnormal smooth muscle cell proliferation. These findings provide insight into the contribution of the miR-874-5p/Sirt 3 cascade with regard to changes in autophagy and proliferation associated with PH.
Collapse
Affiliation(s)
- Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Xiaoying Wang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Siyu He
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Junting Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Wei Xin
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Yiying Li
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Yuan Jiang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Jianshuang Li
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China; State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Daqing, 163319, PR China; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin, 150081, PR China.
| |
Collapse
|
42
|
Chen S, Ding R, Hu Z, Yin X, Xiao F, Zhang W, Yan S, Lv C. MicroRNA-34a Inhibition Alleviates Lung Injury in Cecal Ligation and Puncture Induced Septic Mice. Front Immunol 2020; 11:1829. [PMID: 32903604 PMCID: PMC7438583 DOI: 10.3389/fimmu.2020.01829] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/08/2020] [Indexed: 12/29/2022] Open
Abstract
Sepsis is the leading cause of death in intensive care units. MicroRNA-34a (miR-34a) is involved in sepsis progression, while its underlying mechanisms on sepsis-induced lung injury remain obscure. Oxidative stress, pyroptosis, and inhibition of autophagy can result in organ injury. MiR-34a has been reported to regulate oxidative stress and autophagy via inhibiting silent information regulator T1 (SIRT1) and autophagy gene 4B (ATG4B) signaling. This study aimed at identifying the function of miR-34a in oxidative stress, inflammation, pyroptosis, and autophagy in sepsis-induced lung injury. Male 8-week-old C57BL/6 mice were subjected to cecal ligation and puncture and treated with miR-34a antagomir/agomir. Survival (n = 10), histopathological changes (n = 6), and lung wet-to-dry ratio (n = 6) were recorded and assayed. Other detection (n = 6) was performed to investigate the level of oxidative stress, inflammation, pyroptosis, and autophagy in lung tissues. Results showed that miR-34a down-regulation ameliorated lung injury in septic mice as reflected by decreased lung injury scores (decrease from 3.00 ± 0.32 to 2.00 ± 0.32) and wet-to-dry ratio (0.36-fold decrease). MiR-34a down-regulation also decreased reactive oxygen species accumulation (0.36-fold decrease), and promoted superoxide dismutase activity and the expression of SIRT1 (1.24-fold increase), heme oxygenase-1 and nuclear factor erythroid 2 like 2 to inhibit oxidative stress in septic mice. Moreover, miR-34a down-regulation suppressed inflammatory response and pyroptosis in septic mice, as evidenced by decreased level of pro-inflammatory factors including tumor necrosis factor α, interleukin-6 (IL-6), IL-1β, and IL-18, activity of caspase-1 (0.51-fold decrease) and expression of nucleotide-binding domain and leucine-rich repeat protein-3 (0.48-fold decrease), apoptosis-associated speck-like protein containing a CARD, cleaved-caspase-1, and cleaved-gasdermin D (0.36-fold decrease), and increased level of anti-inflammatory factors IL-10. MiR-34a down-regulation also enhanced autophagy in septic mice as evidenced by more autolysosomes and elevated expressions of ATG4B (0.90-fold increase), beclin1, ATG9, and LC3 II/I. Among these experiments, miR-34a up-regulation showed opposite effects on oxidative stress, inflammatory response, pyroptosis, and autophagy in septic mice. Additionally, miR-34a could bind to the 3′-untranslated region of SIRT1 and ATG4B. In conclusion, our findings demonstrated that miR-34a was implicated in oxidative stress, inflammation, pyroptosis, and autophagy in the development of sepsis. MiR-34a inhibition had a potential to alleviate sepsis-induced lung injury.
Collapse
Affiliation(s)
- Song Chen
- Department of Trauma Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Renyu Ding
- Intensive Care Unit, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ziwei Hu
- Intensive Care Unit, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaohan Yin
- Intensive Care Unit, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Feng Xiao
- Department of Critical Care Medicine, The Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Wei Zhang
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China.,Department of Emergency Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.,Emergency and Trauma College, Hainan Medical University, Haikou, China
| | - Shijiao Yan
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China.,School of Public Health, Hainan Medical University, Haikou, China
| | - Chuanzhu Lv
- Department of Trauma Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China.,Department of Emergency Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.,Emergency and Trauma College, Hainan Medical University, Haikou, China
| |
Collapse
|
43
|
Bao YR, Chen JW, Jiang Y, Wang LH, Xue R, Qian JX, Zhang GX. Sodium Tanshinone II Sulfonate A Ameliorates Hypoxia-Induced Pulmonary Hypertension. Front Pharmacol 2020; 11:687. [PMID: 32508639 PMCID: PMC7253651 DOI: 10.3389/fphar.2020.00687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background Pulmonary hypertension (PH) remains a prevalent disease globally. Sodium tanshinone II sulfonate A (STS) has been used in clinical treatment of PH. Aims The aim of the present study was to investigate the effect of sodium STS treatment on hypoxia-induced PH and related mechanisms. Methods Male Sprague-Dawley rats were housed in a hypoxic chamber with an oxygen concentration of 10 ± 1% for 8 h a day over 21 days. Rats were treated with either STS (low-dose: 10 mg/kg or high-dose: 30 mg/kg) or LY294002 (which is an inhibitor of PI3K). Pulmonary arterial pressure (PAP) was measured, right ventricular hypertrophy parameters were monitored, lung edema parameters were measured, and pathological changes were observed by hematoxylin-eosin (HE) staining. Protein expressions of apoptosis, and PI3K/AKT/mTOR/autophagy pathways in rat lung tissue were examined by western blot. Levels of the pro-inflammatory factors IL-6, IL-8, TNF-α in lung tissues of rats were measured using an enzyme linked immunosorbent assay (ELISA). Results Results of our study demonstrate that persistent exposure to hypoxic conditions increased PAP, right ventricular hypertrophy, lung edema, parameters of lung vascular proliferation and decreased the ratio of Bax/Bcl-2. Furthermore, hypoxic conditions activated the PI3K/Akt/mTOR pathway, inhibited autophagy, and elevated abundance of inflammatory factors in rat lung tissue. Treatment with STS resulted in a dose-dependent decrease in PAP, right ventricular hypertrophy, lung edema, lung vascular proliferation and reversed hypoxia induced lung tissue protein expression and pro-inflammatory factors in rat lung tissue. In addition, hypoxia-induced increases in PAP, cardiac hypertrophy, and lung expression of the proteins PI3K/Akt/mTOR/autophagy pathway were partially reversed by treatment with LY294002. Conclusions STS alleviates hypoxia-induced PH by promoting apoptosis, inhibiting PI3K/AKT/mTOR pathway, up-regulating autophagy, and inhibiting inflammatory responses.
Collapse
Affiliation(s)
- Ya-Ru Bao
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Jing-Wei Chen
- Department of Internal Medicine, Suzhou TCM Hospital affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yan Jiang
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Lin-Hui Wang
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Rong Xue
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| | - Jin-Xian Qian
- Department of Respiratory and Critical Care Medicine, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Guo-Xing Zhang
- Department of Physiology, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
44
|
Xu J, Zhou Y, Yang Y, Lv C, Liu X, Wang Y. Involvement of ABC-transporters and acyltransferase 1 in intracellular cholesterol-mediated autophagy in bovine alveolar macrophages in response to the Bacillus Calmette-Guerin (BCG) infection. BMC Immunol 2020; 21:26. [PMID: 32397995 PMCID: PMC7216371 DOI: 10.1186/s12865-020-00356-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Background Understanding pathogenic mechanisms is imperative for developing novel treatment to the tuberculosis, an important public health burden worldwide. Recent studies demonstrated that host cholesterol levels have implications in the establishment of Mycobacterium tuberculosis (M. tuberculosis, Mtb) infection in host cells, in which the intracellular cholesterol-mediated ATP-binding cassette transporters (ABC-transporters) and cholesterol acyltransferase1 (ACAT1) exhibited abilities to regulate macrophage autophagy induced by Mycobacterium bovis bacillus Calmette–Guérin (BCG). Results The results showed that a down-regulated expression of the ABC-transporters and ACAT1 in primary bovine alveolar macrophages (AMs) and murine RAW264.7 cells in response to a BCG infection. The inhibited expression of ABC-transporters and ACAT1 was associated with the reduction of intracellular free cholesterol, which in turn induced autophagy in macrophages upon to the Mycobacterial infection. These results strongly suggest an involvement of ABC-transporters and ACAT1 in intracellular cholesterol-mediated autophagy in AMs in response to BCG infection. Conclusion This study thus provides an insight into into a mechanism by which the cholesterol metabolism regulated the autophagy in macrophages in response to mycobacterial infections.
Collapse
Affiliation(s)
- Jinrui Xu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Yinchuan, China.,College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Yanbing Zhou
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Yinchuan, China.,College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Yi Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Yinchuan, China.,College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Cuiping Lv
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Yinchuan, China.,College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Xiaoming Liu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Yinchuan, China. .,College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China.
| | - Yujiong Wang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Yinchuan, China. .,College of Life Science, Ningxia University, Yinchuan, 750021, Ningxia, China.
| |
Collapse
|
45
|
Zheng Q, Wang YC, Liu QX, Dong XJ, Xie ZX, Liu XH, Gao W, Bai XJ, Li ZF. FK866 attenuates sepsis-induced acute lung injury through c-jun-N-terminal kinase (JNK)-dependent autophagy. Life Sci 2020; 250:117551. [PMID: 32179075 DOI: 10.1016/j.lfs.2020.117551] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022]
Abstract
AIMS Increasing evidence indicates that FK866, a specific noncompetitive nicotinamide phosphoribosyl transferase inhibitor, exhibits a protective effect on acute lung injury (ALI). Autophagy plays a pivotal role in sepsis-induced ALI. However, the contribution of autophagy and the underlying mechanism by which FK866-confered lung protection remains elusive. Herein, we aimed to study whether FK866 could alleviate sepsis-induced ALI via the JNK-dependent autophagy. MAIN METHODS Male C57BL/6 mice were subjected to cecal ligation and puncture (CLP) to establish the polymicrobial sepsis mice model, and treated with FK866 (10 mg/kg) at 24, 12 and 0.5 h before the CLP procedure. The lung protective effects were measured by lung histopathology, tissue edema, vascular leakage, inflammation infiltration, autophagy-related protein expression and JNK activity. A549 cells were stimulated with LPS (1000 ng/ml) to generate the ALI cell model, and pretreated with FK866 or SP600125 for 30 min to measure the autophagy-related protein expression and JNK activity. KEY FINDINGS Our results demonstrated that FK866 reduced lung injury score, tissue edema, vascular leakage, and inflammatory infiltration, and upregulated autophagy. The protective effect of autophagy conferred by FK866 on ALI was further clarified by using 3-methyladenine (3MA) and rapamycin. Additionally, the activity of JNK was suppressed by FK866, and inhibition of JNK promoted autophagy and showed a benefit effect. SIGNIFICANCE Our study indicates that FK866 protects against sepsis-induced ALI by induction of JNK-dependent autophagy. This may provide new insights into the functional mechanism of NAMPT inhibition in sepsis-induced ALI.
Collapse
Affiliation(s)
- Qiang Zheng
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Yu-Chang Wang
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Qin-Xin Liu
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Xi-Jie Dong
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Zhen-Xing Xie
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Xing-Hua Liu
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Wei Gao
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Xiang-Jun Bai
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Zhan-Fei Li
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China.
| |
Collapse
|
46
|
Gon Y, Shimizu T, Mizumura K, Maruoka S, Hikichi M. Molecular techniques for respiratory diseases: MicroRNA and extracellular vesicles. Respirology 2019; 25:149-160. [PMID: 31872560 DOI: 10.1111/resp.13756] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/04/2019] [Accepted: 11/10/2019] [Indexed: 12/11/2022]
Abstract
miRNA are a class of evolutionarily conserved non-coding 19- to 22-nt regulatory RNA. They affect various cellular functions through modulating the transcriptional and post-transcriptional levels of their target mRNA by changing the stability of protein-coding transcripts or attenuating protein translation. miRNA were discovered in the early 1990s, and they have been the focus of new research in both basic and clinical medical sciences. Today, it has become clear that specific miRNA are linked to the pathogenesis of respiratory diseases, as well as cancer and cardiovascular disease. In addition, EV, including exosomes, which are small membrane-bound vesicles secreted by cells, were found to contain various functional miRNA that can be used for diagnostic and therapeutic purposes. As body fluids, such as blood and respiratory secretions, are major miRNA sources in the body, EV carrying extracellular miRNA are considered potentially useful for the diagnosis and assessment of pathological conditions, as well as the treatment of respiratory or other diseases. Although research in the field of lung cancer is actively progressing, studies in other respiratory fields have emerged recently as well. In this review, we provide an update in the topics of miRNA and EV focused on airway inflammatory diseases, such as asthma and COPD, and explore their potential for clinical applications on respiratory diseases.
Collapse
Affiliation(s)
- Yasuhiro Gon
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tetsuo Shimizu
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kenji Mizumura
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Shuichiro Maruoka
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Mari Hikichi
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
47
|
Zhao H, Wang Y, Qiu T, Liu W, Yao P. Autophagy, an important therapeutic target for pulmonary fibrosis diseases. Clin Chim Acta 2019; 502:139-147. [PMID: 31877297 DOI: 10.1016/j.cca.2019.12.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
As an evolutionarily conserved intracellular degradation pathway, autophagy is essential to cellular homeostasis. Several studies have demonstrated that autophagy showed an important effect on some pulmonary fibrosis diseases, including idiopathic pulmonary fibrosis (IPF), cystic fibrosis lung disease, silicosis and smoking-induced pulmonary fibrosis. For example, autophagy mitigates the pathological progression of IPF by regulating the apoptosis of fibroblasts and the senescence of alveolar epithelial cells. In addition, autophagy ameliorates cystic fibrosis lung disease via rescuing transmembrane conductance regulators (CFTRs) to the plasma membrane. Furthermore, autophagy alleviates the silica-induced pulmonary fibrosis by decreasing apoptosis of alveolar epithelial cells in silicosis. However, excessive macrophage autophagy aggravates the pathogenesis of silicosis fibrosis by promoting the proliferation and migration of lung fibroblasts in silicosis. Autophagy is also involved in smoking-induced pulmonary fibrosis, coal workers' pneumoconiosis, ionizing radiation-mediated pulmonary fibrosis and heavy metal nanoparticle-mediated pulmonary fibrosis. In this review, the role and signalling mechanisms of autophagy in the progression of pulmonary fibrosis diseases have been systematically analysed. It has provided a new insight into the therapeutic potential associated with autophagy in pulmonary fibrosis diseases. In conclusion, the targeting of autophagy might prove to be a prospective avenue for the therapeutic intervention of pulmonary fibrosis diseases.
Collapse
Affiliation(s)
- Hong Zhao
- Nursing College, University of South China, Hengyang, 421001, China
| | - Yiqun Wang
- Department of Anesthesiology, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China
| | - Tingting Qiu
- Nursing College, University of South China, Hengyang, 421001, China
| | - Wei Liu
- Department of Intensive Care Units, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China.
| | - Pingbo Yao
- Department of Clinical Technology, Changsha Health Vocational College, Changsha 410100, China.
| |
Collapse
|
48
|
Khan IM, Niazi S, Iqbal Khan MK, Pasha I, Mohsin A, Haider J, Iqbal MW, Rehman A, Yue L, Wang Z. Recent advances and perspectives of aggregation-induced emission as an emerging platform for detection and bioimaging. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.115637] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
Advances in Autophagy, Tissue Injury, and Homeostasis: Cells Special Issue. Cells 2019; 8:cells8070743. [PMID: 31330980 PMCID: PMC6679422 DOI: 10.3390/cells8070743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 01/12/2023] Open
|
50
|
Zhang Z, Wang X, Ma C, Li Z, Chen H, Zhang Z, Li T. Genipin protects rats against lipopolysaccharide-induced acute lung injury by reinforcing autophagy. Int Immunopharmacol 2019; 72:21-30. [PMID: 30959368 DOI: 10.1016/j.intimp.2019.03.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/11/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
Although the protective effects of genipin against acute lung injury (ALI) have been described previously, the associated mechanism remains unclear. We have previously reported that genipin exerts its pharmacological effects by regulating autophagy. Here, we hypothesized that the up-regulation of autophagy may contribute to the protective effects exhibited by genipin against ALI. In the present study, ALI was induced by intratracheal LPS administration in rats. Genipin treatment significantly reduced LPS-induced lung injury as evidenced by improved histopathology, decreased lung edema, total cells, and protein concentration in the bronchoalveolar lavage fluid (BALF). This protection was inhibited by 3-methyladenine (3-MA), an inhibitor of autophagy. Genipin treatment reduced the expression of P62 and increased the expression of Beclin-1 and LC3II, indicating increased autophagy. Genipin treatment also alleviated LPS-induced cell apoptosis (down-regulation of Bax, up-regulation of Bcl-2, and decreased number of terminal deoxynucleotidyl transferase dUTP nick end label-positive cells) and oxidative stress (increased SOD and decreased MDA content) in the lung. Furthermore, genipin attenuated LPS-induced production of TNF-α, IL-1β, and IL-6 in the lung and BALF. These protective effects induced by genipin were reversed by 3-MA treatment, indicating that autophagy was involved in the protective effects exerted by genipin against inflammation and apoptosis in ALI. In A549 cells incubated with LPS for 6 h, genipin treatment increased the number of GFP-LC3 punctae. 3-MA prevented the protective effects of genipin against mitochondrial dysfunction and cell death. These findings suggest that genipin protects against apoptosis and inflammation in LPS-induced ALI by promoting autophagy.
Collapse
Affiliation(s)
- Zhijie Zhang
- Department of Anesthesiology, Yidu Central Hospital of Weifang, Weifang, Shandong Province, China
| | - Xue Wang
- Department of Anesthesiology, Yidu Central Hospital of Weifang, Weifang, Shandong Province, China
| | - Chengzhou Ma
- Department of Anesthesiology, Dongcheng Street Hospital of Linqu, Weifang, Shandong Province, China
| | - Zhiwang Li
- Department of Anesthesiology, The First People's Hospital of Chenzhou/Institute of Translation Medicine, University of South China, Chenzhou 423000, China
| | - Huayong Chen
- Department of Anesthesiology, Yidu Central Hospital of Weifang, Weifang, Shandong Province, China
| | - Zhiming Zhang
- Department of Anesthesiology, The First People's Hospital of Chenzhou/Institute of Translation Medicine, University of South China, Chenzhou 423000, China.
| | - Tao Li
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou/Institute of Translation Medicine, University of South China, Chenzhou 423000, China.
| |
Collapse
|