1
|
Liu X, Song X, Li G, Zhang Y, Liu N, Tang K, Du H, Jie L. HMGB1 regulates the activation of dendritic cells and CD4+ T cell responses through the modulation of autophagy in bleomycin-induced pulmonary fibrosis. Immunobiology 2025; 230:152906. [PMID: 40311346 DOI: 10.1016/j.imbio.2025.152906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND The role of HMGB1 in inflammation and autophagy has garnered increasing attention; however, its impact on the activation of dendritic cells (DCs) and autophagy remains unclear. This study aims to explore the effects of HMGB1 on DC activation, autophagy, and its influence on CD4+ T cell responses in a bleomycin-induced pulmonary fibrosis (PF) mouse model. METHODS Thirty mice were randomly divided into control and model groups. The model group was established by intratracheal injection of bleomycin to induce PF. Flow cytometry was used to detect DC surface markers, and western blot was employed to assess the expression of autophagy-related protein LC3. Lung DCs and peripheral blood CD14+ monocytes were sorted using magnetic beads and differentiated into M0-DCs, which were then subjected to HMGB1 stimulation experiments to assess activation and cytokine secretion. HMGB1-stimulated or untreated M0-DCs were co-cultured with CFSE-labeled naive CD4+ T cells to evaluate T cell proliferation and differentiation. The effects of HMGB1 on DCs activation, cytokine secretion, and autophagy-related protein expression were assessed after treatment with autophagy regulators. RESULTS The model group showed significantly elevated levels of HMGB1 in serum and lung tissues, accompanied by upregulated activation markers of DCs and increased expression of autophagy-related protein LC3. HMGB1 stimulation significantly enhanced the activation of M0-DCs and the secretion of pro-inflammatory cytokines, promoting the proliferation of CD4+ T cells and their differentiation into Th1 and Th17 subsets. Rapamycin, which enhances autophagy, potentiated HMGB1-mediated DC activation, while 3-MA, which inhibits autophagy, suppressed the effects of HMGB1, further influencing CD4+ T cell differentiation. CONCLUSION HMGB1 modulates DC autophagy, thereby affecting their activation and immune responses of CD4+ T cells in bleomycin-induced PF. Targeting HMGB1 and the autophagy pathway may provide new strategies for the treatment of PF.
Collapse
Affiliation(s)
- Xiuhua Liu
- Department of rheumatology and clinical immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, China; Department of Rheumatism and Immunology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, China
| | - Xinghui Song
- Department of Rheumatism and Immunology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, China
| | - Guangting Li
- Department of Rheumatism and Immunology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, China
| | - Yuping Zhang
- Department of rheumatology and clinical immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, China
| | - Nina Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Kaijiang Tang
- Department of Rheumatism and Immunology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545007, China
| | - Hongyan Du
- Department of rheumatology and clinical immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, China; School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Province Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Ligang Jie
- Department of rheumatology and clinical immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
2
|
Li Y, Zheng J, Liu F, Tan X, Jiang H, Wang Y. Discussion of the material basis for prevention and treatment of pulmonary fibrosis using naturally medicinal and edible homologous herbs based on the dynamic process of Nrf2, NF-κB and TGF-β in PF. Biomed Pharmacother 2025; 185:117911. [PMID: 40090283 DOI: 10.1016/j.biopha.2025.117911] [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: 09/27/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 03/18/2025] Open
Abstract
Pulmonary fibrosis (PF) is a progressive chronic lung disease with a high incidence and poor prognosis. Despite extensive research into the mechanisms that initiate and drive the progression of pulmonary fibrosis, developing effective treatments remains challenging due to the multiple etiologies, pathogenic links, and signaling pathways involved in PF. Indeed, nuclear factor erythroid 2-related factor 2 (Nrf2), nuclear factor kappa-B (NF-κB), and transforming growth factor-beta (TGF-β) are central players in the pathogenesis of pulmonary fibrosis, and each of these factors influences distinct yet interconnected processes that collectively contribute to disease progression: Nrf2 upregulates antioxidants to mitigate oxidative stress, NF-κB modulates inflammatory responses, and TGF-β promotes fibroblast activation and extracellular matrix (ECM) deposition, leading to fibrosis. Targeting these pathways may offer therapeutic strategies, uncover new insights and provide potential therapeutic targets for PF. Absolutely, the interactions between Nrf2, NF-κB, and TGF-β pathways are complex and can significantly influence the progression of PF, which indicated that targeting a single pathway may show poor efficacy in managing the condition. Moreover, few therapies that effectively intervene in these pathways have been approved. This review focused on the molecular mechanisms of Nrf2, NF-κB, and TGF-β involving in PF and the material basis of the naturally medicinal and edible homologous herbs, which provides a solid foundation for understanding the disease's pathogenesis, and supports the development of therapeutic drugs or treatments for addressing the complex nature of PF.
Collapse
Affiliation(s)
- Yan Li
- Chongqing Academy of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 400065, PR China; Chongqing Key Laboratory of Chinese Medicine & Health Science, Chongqing 400065, PR China.
| | - Jia Zheng
- Chongqing University of Chinese Medicine, Chongqing 402760, PR China.
| | - Fei Liu
- Chongqing Academy of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 400065, PR China.
| | - Xianfeng Tan
- Chongqing Baijiahuan Health Technology Co., Ltd, Chongqing 400065, China.
| | - Huiping Jiang
- Chongqing Baijiahuan Health Technology Co., Ltd, Chongqing 400065, China.
| | - Yongde Wang
- Chongqing Academy of Chinese Materia Medica, Chongqing University of Chinese Medicine, Chongqing 400065, PR China; Chongqing Key Laboratory of Chinese Medicine & Health Science, Chongqing 400065, PR China.
| |
Collapse
|
3
|
Chen YZ, Zhao L, Wei W, Gu J, Liu ZH, Shan WY, Dong J, Li C, Qin LQ, Xu JY. The Effect of Metformin on Radiation-Induced Lung Fibrosis in Mice. Dose Response 2024; 22:15593258241308051. [PMID: 39664837 PMCID: PMC11632958 DOI: 10.1177/15593258241308051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction: Radiation-induced lung fibrosis (RILF) is a common complication of thoracic radiotherapy. Metformin has been suggested to have a radioprotective effect. Objective: This study explored the radioprotective effects of metformin on RILF and its mechanisms. Methods: C57BL/6J mice were randomly divided into control, ionizing radiation (IR), low-dose metformin (L-Met), and high-dose metformin (H-Met) groups. The IR, L-Met, and H-Met groups received 15 Gy chest irradiation. The L-Met and H-Met groups were administrated 100 or 200 mg/kg metformin from 3 days before irradiation and continued for 6 months. The mice were then sacrificed, and samples were collected for further analysis. Results: RILF was induced in the irradiated mice. Metformin improved lung pathology, inhibited collagen deposition, and reduced inflammatory factors such as high mobility group box 1 (HMGB1), interleukin-1 beta, interleukin-6, tumor necrosis factor alpha in lung tissue, lavage fluid, and serum. Western blot and quantitative real-time PCR analyses revealed that metformin downregulated HMGB1, toll-like receptor 4 (TLR4), and nuclear factor kappaB (NF-κB) expression. Additionally, metformin reversed the irradiation-induced reduction in the abundance of Lactobacillus and Lachnospiraceae at the genus level. Conclusion: Our findings indicated that metformin ameliorates RILF by downregulating the inflammatory-related HMGB1/TLR4/NF-κB pathway and improving intestinal flora disorder.
Collapse
Affiliation(s)
- Yu-Zhong Chen
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- Yancheng Municipal Center for Disease Control and Prevention, Yancheng, China
| | - Lin Zhao
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wei Wei
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jia Gu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhen-Hua Liu
- Department of Radiotherapy, The Yancheng Clinical College of Xuzhou Medical University, The First people’s Hospital of Yancheng, Yancheng, China
| | - Wen-Yue Shan
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jie Dong
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Li
- Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jia-Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Yuan J, Guo L, Ma J, Zhang H, Xiao M, Li N, Gong H, Yan M. HMGB1 as an extracellular pro-inflammatory cytokine: Implications for drug-induced organic damage. Cell Biol Toxicol 2024; 40:55. [PMID: 39008169 PMCID: PMC11249443 DOI: 10.1007/s10565-024-09893-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024]
Abstract
Drug-induced organic damage encompasses various intricate mechanisms, wherein HMGB1, a non-histone chromosome-binding protein, assumes a significant role as a pivotal hub gene. The regulatory functions of HMGB1 within the nucleus and extracellular milieu are interlinked. HMGB1 exerts a crucial regulatory influence on key biological processes including cell survival, inflammatory regulation, and immune response. HMGB1 can be released extracellularly from the cell during these processes, where it functions as a pro-inflammation cytokine. HMGB1 interacts with multiple cell membrane receptors, primarily Toll-like receptors (TLRs) and receptor for advanced glycation end products (RAGE), to stimulate immune cells and trigger inflammatory response. The excessive or uncontrolled HMGB1 release leads to heightened inflammatory responses and cellular demise, instigating inflammatory damage or exacerbating inflammation and cellular demise in different diseases. Therefore, a thorough review on the significance of HMGB1 in drug-induced organic damage is highly important for the advancement of pharmaceuticals, ensuring their effectiveness and safety in treating inflammation as well as immune-related diseases. In this review, we initially outline the characteristics and functions of HMGB1, emphasizing their relevance in disease pathology. Then, we comprehensively summarize the prospect of HMGB1 as a promising therapeutic target for treating drug-induced toxicity. Lastly, we discuss major challenges and propose potential avenues for advancing the development of HMGB1-based therapeutics.
Collapse
Affiliation(s)
- JianYe Yuan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pathology, The Eight Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Lin Guo
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - JiaTing Ma
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - HeJian Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - MingXuan Xiao
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - Ning Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - Miao Yan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, China.
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China.
| |
Collapse
|
5
|
Xiong W, Chen S, Xiang H, Zhao S, Xiao J, Li J, Liu Y, Shu Z, Ouyang J, Zhang J, Liu H, Wang X, Zou H, Chen Y, Chen A, Lu H. S1PR1 attenuates pulmonary fibrosis by inhibiting EndMT and improving endothelial barrier function. Pulm Pharmacol Ther 2023:102228. [PMID: 37295666 DOI: 10.1016/j.pupt.2023.102228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic fatal disease of unknown etiology. Its pathological manifestations include excessive proliferation and activation of fibroblasts and deposition of extracellular matrix. Endothelial cell-mesenchymal transformation (EndMT), a novel mechanism that generates fibroblast during IPF, is responsible for fibroblast-like phenotypic changes and activation of fibroblasts into hypersecretory cells. However, the exact mechanism behind EndMT-derived fibroblasts and activation is uncertain. Here, we investigated the role of sphingosine 1-phosphate receptor 1 (S1PR1) in EndMT-driven pulmonary fibrosis. METHODS We treated C57BL/6 mice with bleomycin (BLM) in vivo and pulmonary microvascular endothelial cells with TGF-β1 in vitro. Western blot,flow cytometry, and immunofluorescence were used to detect the expression of S1PR1 in endothelial cells. To evaluate the effect of S1PR1 on EndMT and endothelial barrier and its role in lung fibrosis and related signaling pathways, S1PR1 agonist and antagonist were used in vitro and in vivo. RESULTS Endothelial S1PR1 protein expression was downregulated in both in vitro and in vivo models of pulmonary fibrosis induced by TGF-β1 and BLM, respectively. Downregulation of S1PR1 resulted in EndMT, indicated by decreased expression of endothelial markers CD31 and VE-cadherin, increased expression of mesenchymal markers α-SMA and nuclear transcription factor Snail, and disruption of the endothelial barrier. Further mechanistic studies found that stimulation of S1PR1 inhibited TGF-β1-mediated activation of the Smad2/3 and RhoA/ROCK1 pathways. Moreover, stimulation of S1PR1 attenuated Smad2/3 and RhoA/ROCK1 pathway-mediated damage to endothelial barrier function. CONCLUSIONS Endothelial S1PR1 provides protection against pulmonary fibrosis by inhibiting EndMT and attenuating endothelial barrier damage. Accordingly, S1PR1 may be a potential therapeutic target in progressive IPF.
Collapse
Affiliation(s)
- Wenfang Xiong
- Health Management Center, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China; Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Shuhua Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, 410013, PR China
| | - Hong Xiang
- Center for Experimental Medicine, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Shaoli Zhao
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jie Xiao
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jialing Li
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Yulan Liu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Zhihao Shu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jie Ouyang
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Jing Zhang
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Huiqin Liu
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Xuewen Wang
- Department of Cardiology, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Hang Zou
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, 410013, PR China
| | - Ying Chen
- Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan, 410013, PR China
| | - Alex Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Hongwei Lu
- Health Management Center, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China; Center for Experimental Medicine, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China.
| |
Collapse
|
6
|
Dorababu A, Maraswami M. Recent Advances (2015-2020) in Drug Discovery for Attenuation of Pulmonary Fibrosis and COPD. Molecules 2023; 28:molecules28093674. [PMID: 37175084 PMCID: PMC10179756 DOI: 10.3390/molecules28093674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
A condition of scarring of lung tissue due to a wide range of causes (such as environmental pollution, cigarette smoking (CS), lung diseases, some medications, etc.) has been reported as pulmonary fibrosis (PF). This has become a serious problem all over the world due to the lack of efficient drugs for treatment or cure. To date, no drug has been designed that could inhibit fibrosis. However, few medications have been reported to reduce the rate of fibrosis. Meanwhile, ongoing research indicates pulmonary fibrosis can be treated in its initial stages when symptoms are mild. Here, an attempt is made to summarize the recent studies on the effects of various chemical drugs that attenuate PF and increase patients' quality of life. The review is classified based on the nature of the drug molecules, e.g., natural/biomolecule-based, synthetic-molecule-based PF inhibitors, etc. Here, the mechanisms through which the drug molecules attenuate PF are discussed. It is shown that inhibitory molecules can significantly decrease the TGF-β1, profibrotic factors, proteins responsible for inflammation, pro-fibrogenic cytokines, etc., thereby ameliorating the progress of PF. This review may be useful in designing better drugs that could reduce the fibrosis process drastically or even cure the disease to some extent.
Collapse
Affiliation(s)
- Atukuri Dorababu
- Department of Chemistry, SRMPP Government First Grade College, Huvinahadagali 583219, India
| | - Manikantha Maraswami
- Department of Chemistry, Abzena LLC., 360 George Patterson Blvd, Bristol, PA 19007, USA
| |
Collapse
|
7
|
Depleted uranium causes renal mitochondrial dysfunction through the ETHE1/Nrf2 pathway. Chem Biol Interact 2023; 372:110356. [PMID: 36681261 DOI: 10.1016/j.cbi.2023.110356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/08/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
The kidney is the main organ affected by acute depleted uranium (DU) toxicity. The mechanism of nephrotoxicity induced by DU is complex and needs to be further explored. This study aimed to elucidate the function of mitochondrial dysfunction in nephrotoxicity generated by DU and confirm the latent mechanism. We verified that DU (2.5-10 mg/kg) caused mitochondrial dysfunction in male rat kidneys and decreased ATP content and the mitochondrial membrane potential. In addition, melatonin (20 mg/kg), as an antioxidant, alleviated DU-induced oxidative stress and mitochondrial dysfunction in male rats, further reducing kidney damage caused by DU. These results indicate that mitochondrial dysfunction plays a vital role in DU nephrotoxicity. When ethylmalonic encephalopathy 1 (ETHE1) was knocked down, DU-induced oxidative stress and mitochondrial dysfunction were increased, and renal injury was aggravated. When exogenous ETHE1 protein was applied to renal cells, the opposite changes were observed. We also found that ETHE1 knockdown increased the expression of NF-E2-related factor 2 (Nrf2), a vital oxidative stress regulator, and its downstream molecules heme oxygenase-1 (HO-1) and NADPH quinone oxidoreductase 1 (NQO1). Nrf2 knockout also aggravated DU-induced oxidative stress, mitochondrial dysfunction, and kidney damage. In conclusion, DU causes oxidative stress and antioxidant defense imbalance in renal cells through the ETHE1/Nrf2 pathway, further causing mitochondrial dysfunction and ultimately leading to nephrotoxicity.
Collapse
|
8
|
Zhang X, Duan XJ, Li LR, Chen YP. lncRNA NEAT1 promotes hypoxia-induced inflammation and fibrosis of alveolar epithelial cells via targeting miR-29a/NFATc3 axis. Kaohsiung J Med Sci 2022; 38:739-748. [PMID: 35708150 DOI: 10.1002/kjm2.12535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 11/09/2022] Open
Abstract
The objective of the present study was to explore the function and mechanism of long noncoding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) in pulmonary fibrosis (PF) progression. HPAEpic cells and A549 cells were exposed to hypoxic conditions to establish an in vitro model. Cell apoptosis was detected by TUNEL assay, and inflammatory cytokine levels were detected by ELISA. Gene and protein expression levels were identified by qRT-PCR and Western blot assays, respectively. The interaction among NEAT1, miR-29a, and NFATc3 was identified by dual-luciferase reporter and RNA pull-down assays. In hypoxia-treated cells, hypoxia markers (HIF-1α and HIF-2α), cytokines (TNF-α, IL-1β, and IL-6) and fibrotic markers (α-SMA, collagen I and collagen III) were significantly enhanced. Consistently, the expression levels of NEAT1 and NFATc3 were increased, but miR-29a was decreased in hypoxia-stimulated cells. Knockdown of NEAT1 significantly decreased cell apoptosis and the releases of TNF-α, IL-1β, and IL-6 as well as reduced the levels of α-SMA, collagen I, and collagen III. Moreover, NEAT1 positively regulated NFATc3 expression by directly targeting miR-29a. Functional experiments showed that the anti-apoptotic, anti-inflammatory, and anti-fibrotic effects mediated by NETA1 silencing were impeded by miR-29a inhibition or NFATc3 overexpression in hypoxia-stimulated HPAEpic and A549 cells. Collectively, these data demonstrated that NEAT1 knockdown inhibited hypoxia-induced cell apoptosis, inflammation, and fibrosis by targeting the miR-29a/NFATc3 axis in PF, suggesting that NEAT1 might be a potential therapeutic target for relieving PF progression.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Respiratory Medicine, Hunan Children's Hospital, Changsha, Hunan Province, P. R. China
| | - Xiao-Jun Duan
- Department of Respiratory Medicine, Hunan Children's Hospital, Changsha, Hunan Province, P. R. China
| | - Lin-Rui Li
- Department of Respiratory Medicine, Hunan Children's Hospital, Changsha, Hunan Province, P. R. China
| | - Yan-Ping Chen
- Department of Respiratory Medicine, Hunan Children's Hospital, Changsha, Hunan Province, P. R. China
| |
Collapse
|
9
|
Comparing Outcomes of Two Antiviral Therapy Combinations among COVID-19 Patients. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1522426. [PMID: 35013710 PMCID: PMC8742149 DOI: 10.1155/2022/1522426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/08/2021] [Indexed: 01/10/2023]
Abstract
Several therapeutic regimens for COVID-19 have been studied, such as combination antiviral therapies. We aimed to compare outcome of two types of combination therapies atazanavir/ritonavir (ATV/r) or lopinavir/ritonavir (LPV/r) plus hydroxychloroquine among COVID-19 patients. 108 patients with moderate and severe forms of COVID-19 were divided into two groups (each group 54 patients). One group received ATV/r plus hydroxychloroquine, and the other group received hydroxychloroquine plus LPV/r. Then, both groups were evaluated and compared for clinical symptoms, recovery rates, and complications of treatment regimens. Our findings showed a significant increase in bilirubin in ATV/r-receiving group compared to LPV/r receivers. There was also a significant increase in arrhythmias in the LPV/r group compared to the ATV/r group during treatment. Other findings including length of hospital stay, outcome, and treatment complications were not statistically significant. There is no significant difference between protease inhibitor drugs including ATV/r and LPV/r in the treatment of COVID-19 regarding clinical outcomes. However, some side effects such as hyperbilirubinemia and arrhythmia were significantly different by application of atazanavir or lopinavir.
Collapse
|
10
|
Wang X, Ren R, Xu Z, Huang H, Jiang W, Ma J. Tirbanibulin Attenuates Pulmonary Fibrosis by Modulating Src/STAT3 Signaling. Front Pharmacol 2021; 12:693906. [PMID: 34349652 PMCID: PMC8326405 DOI: 10.3389/fphar.2021.693906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022] Open
Abstract
Tirbanibulin (KX-01) is the first clinical Src inhibitor of the novel peptidomimetic class that targets the peptide substrate site of Src providing more specificity toward the Src kinase. This study assessed the impact of KX-01 on cobalt chloride (CoCl2)-treated L929 cells and bleomycin (BLM)-induced pulmonary fibrosis in rats to evaluate the efficacy of this compound in vitro and in vivo, respectively. In CoCl2-treated L929 cells, KX-01 significantly reduced the expression of smooth muscle actin (α-SMA), collagen I, collagen III, hypoxia inducing factor (HIF-1α), signal transducers and transcriptional activators (p-STAT3), and p-Src. In BLM-induced pulmonary fibrosis rats, KX-01 reduced pathological scores, collagen deposition, α-SMA, collagen I, collagen III, p-Src, HIF-1α, and p-STAT3. Overall, these findings revealed that KX-01 can alleviate experimental pulmonary fibrosis via suppressing the p-SRC/p-STAT3 signaling pathways.
Collapse
Affiliation(s)
- Xin Wang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Rui Ren
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zehui Xu
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Haidi Huang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Wanglin Jiang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Jinbo Ma
- Medicine & Pharmacy Research Center, Binzhou Medical University, Yantai, China
| |
Collapse
|
11
|
Wang S, Li J, He Y, Ran Y, Lu B, Gao J, Shu C, Li J, Zhao Y, Zhang X, Hao Y. Protective effect of melatonin entrapped PLGA nanoparticles on radiation-induced lung injury through the miR-21/TGF-β1/Smad3 pathway. Int J Pharm 2021; 602:120584. [PMID: 33887395 DOI: 10.1016/j.ijpharm.2021.120584] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/18/2021] [Accepted: 04/04/2021] [Indexed: 01/04/2023]
Abstract
Radiation-induced lung injury (RILI) is a complication commonly found in victims suffering from nuclear accidents and patients treated with chest tumor radiotherapy, and drugs are limited for effective prevention and treatment. Melatonin (MET) has an anti-radiation effect, but its metabolic period in the body is short. In order to prolong the metabolism period of MET, we prepared MET entrapped poly (lactic-co-glycolic acid) nanoparticles (MET/PLGANPS) for the treatment of RILI. As a result, the release rate of MET/PLGANPS in vitro was lower than MET, with stable physical properties, and it caused no changes in histopathology and biochemical indicators. After 2 weeks and 16 weeks of irradiation with the dose of 15 Gy, MET and MET/PLGANPS could reduce the expression of caspase-3 proteins, inflammatory factors, TGF-β1 and Smad3 to alleviate radiation-induced lung injury. MET/PLGANPS showed better therapeutic effect on RILI than MET. In addition, we also found that high expression of miR-21 could increase the expression levels of TGF-β1, and inhibit the protective effect of MET/PLGANPS. In conclusion, MET/PLGANPS may alleviate RILI by inhibiting the miR-21/TGF-β1/Smad3 pathway, which would provide a new target for the treatment of radiation-induced lung injury.
Collapse
Affiliation(s)
- Shuang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Juan Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Yingjuan He
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Yonghong Ran
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Binghui Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Jining Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Chang Shu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Jie Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Yazhen Zhao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China
| | - Xin Zhang
- Chongqing Normal University, No.37, Middle University Road, Shapingba District, Chongqing 401331, China
| | - Yuhui Hao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, No.30 Gaotanyan Street, Shapingba District, Chongqing 400038, China.
| |
Collapse
|
12
|
Yun E, Kook Y, Yoo KH, Kim KI, Lee MS, Kim J, Lee A. Endothelial to Mesenchymal Transition in Pulmonary Vascular Diseases. Biomedicines 2020; 8:biomedicines8120639. [PMID: 33371458 PMCID: PMC7767472 DOI: 10.3390/biomedicines8120639] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Lung diseases, such as pulmonary hypertension and pulmonary fibrosis, are life-threatening diseases and have common features of vascular remodeling. During progression, extracellular matrix protein deposition and dysregulation of proteolytic enzymes occurs, which results in vascular stiffness and dysfunction. Although vasodilators or anti-fibrotic therapy have been mainly used as therapy owing to these characteristics, their effectiveness does not meet expectations. Therefore, a better understanding of the etiology and new therapeutic approaches are needed. Endothelial cells (ECs) line the inner walls of blood vessels and maintain vascular homeostasis by protecting vascular cells from pathological stimuli. Chronic stimulation of ECs by various factors, including pro-inflammatory cytokines and hypoxia, leads to ECs undergoing an imbalance of endothelial homeostasis, which results in endothelial dysfunction and is closely associated with vascular diseases. Emerging studies suggest that endothelial to mesenchymal transition (EndMT) contributes to endothelial dysfunction and plays a key role in the pathogenesis of vascular diseases. EndMT is a process by which ECs lose their markers and show mesenchymal-like morphological changes, and gain mesenchymal cell markers. Despite the efforts to elucidate these molecular mechanisms, the role of EndMT in the pathogenesis of lung disease still requires further investigation. Here, we review the importance of EndMT in the pathogenesis of pulmonary vascular diseases and discuss various signaling pathways and mediators involved in the EndMT process. Furthermore, we will provide insight into the therapeutic potential of targeting EndMT.
Collapse
Affiliation(s)
- Eunsik Yun
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
| | - Yunjin Kook
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
| | - Kyung Hyun Yoo
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Keun Il Kim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Myeong-Sok Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
- Correspondence: (J.K.); (A.L.); Tel.: +82-2-710-9553 (J.K. & A.L.); Fax: +82-2-2077-7322 (J.K. & A.L.)
| | - Aram Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Correspondence: (J.K.); (A.L.); Tel.: +82-2-710-9553 (J.K. & A.L.); Fax: +82-2-2077-7322 (J.K. & A.L.)
| |
Collapse
|
13
|
Xian S, Chen A, Wu X, Lu C, Wu Y, Huang F, Zeng Z. Activation of activin/Smad2 and 3 signaling pathway and the potential involvement of endothelial‑mesenchymal transition in the valvular damage due to rheumatic heart disease. Mol Med Rep 2020; 23:10. [PMID: 33179113 PMCID: PMC7673319 DOI: 10.3892/mmr.2020.11648] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Rheumatic heart disease (RHD) is an autoimmune disease caused by rheumatic fever following group A hemolytic streptococcal infection and primarily affects the mitral valve. RHD is currently a major global health problem. However, the exact pathological mechanisms associated with RHD-induced cardiac valve damage remain to be elucidated. The endothelial-mesenchymal transition (EndMT) serves a key role in a number of diseases with an important role in cardiac fibrosis and the activin/Smad2 and 3 signaling pathway is involved in regulating the EndMT. Nevertheless, there are no studies to date, to the best of the authors' knowledge, investigating the association between RHD and EndMT. Thus, the aim of the current study was to investigate the potential role of EndMT in cardiac valve damage and assess whether activin/Smad2 and 3 signaling was activated during RHD-induced valvular injury in a rat model of RHD induced by inactivated Group A streptococci and complete Freund's adjuvant. Inflammation and fibrosis were assessed by hematoxylin and eosin and Sirius red staining. Serum cytokine and rheumatoid factor levels were measured using ELISA kits. Expression levels of activin/Smad2 and 3 signaling pathway-related factors [activin A, Smad2, Smad3, phosphorylated (p-)Smad2 and p-Smad3], EndMT-related factors [lymphoid enhancer factor-1 (LEF-1), Snail1, TWIST, zinc finger E-box-binding homeobox (ZEB)1, ZEB2, α smooth muscle actin (α-SMA) and type I collagen α 1 (COL1A1)], apoptosis-related markers (BAX and cleaved caspase-3) and valvular inflammation markers (NF-κB and p-NF-κB) were detected using reverse transcription-quantitative PCR and western blot analyses. Compared with the control group, the degree of valvular inflammation and fibrosis, serum levels of IL-6, IL-17, TNF-α and expression of apoptosis-related markers (BAX and cleaved caspase-3) and valvular inflammation marker (p-NF-κB), activin/Smad2 and 3 signaling pathway-related factors (activin A, p-Smad2 and p-Smad3), EndMT-related factors (LEF-1, Snail1, TWIST, ZEB 1, ZEB2, α-SMA and COL1A1) were significantly increased in the RHD group. These results suggested that the activin/Smad2 and 3 signaling pathway was activated during the development of valvular damage caused by RHD and that the EndMT is involved in RHD-induced cardiac valve damage.
Collapse
Affiliation(s)
- Shenglin Xian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ang Chen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaodan Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chuanghong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yunjiao Wu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhiyu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
14
|
Atazanavir, Alone or in Combination with Ritonavir, Inhibits SARS-CoV-2 Replication and Proinflammatory Cytokine Production. Antimicrob Agents Chemother 2020; 64:AAC.00825-20. [PMID: 32759267 PMCID: PMC7508582 DOI: 10.1128/aac.00825-20] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/02/2020] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is already responsible for far more deaths than previous pathogenic coronaviruses (CoVs) from 2002 and 2012. The identification of clinically approved drugs to be repurposed to combat 2019 CoV disease (COVID-19) would allow the rapid implementation of potentially life-saving procedures. The major protease (Mpro) of SARS-CoV-2 is considered a promising target, based on previous results from related CoVs with lopinavir (LPV), an HIV protease inhibitor. However, limited evidence exists for other clinically approved antiretroviral protease inhibitors. Extensive use of atazanavir (ATV) as antiretroviral and previous evidence suggesting its bioavailability within the respiratory tract prompted us to study this molecule against SARS-CoV-2. Our results show that ATV docks in the active site of SARS-CoV-2 Mpro with greater strength than LPV, blocking Mpro activity. We confirmed that ATV inhibits SARS-CoV-2 replication, alone or in combination with ritonavir (RTV) in Vero cells and a human pulmonary epithelial cell line. ATV/RTV also impaired virus-induced enhancement of interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α) levels. Together, our data strongly suggest that ATV and ATV/RTV should be considered among the candidate repurposed drugs undergoing clinical trials in the fight against COVID-19.
Collapse
|
15
|
Zhao J, Liu Y, Sun J, Zhu H, Chen Y, Dong T, Sang R, Gao X, Yang W, Deng Y. Magnetic targeting cobalt nanowire-based multifunctional therapeutic system for anticancer treatment and angiogenesis. Colloids Surf B Biointerfaces 2020; 194:111217. [PMID: 32622255 DOI: 10.1016/j.colsurfb.2020.111217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/03/2020] [Accepted: 06/23/2020] [Indexed: 12/09/2022]
Abstract
In order to improve the anticancer therapeutic efficacy and postoperative recovery efficacy, the novel anticancer therapeutic system should have the ability to promote angiogenesis after anticancer therapy besides the excellent anticancer therapeutic efficacy. We present herein a magnetic targeting multifunctional anticancer therapeutic system based on cobalt nanowires (CoNWs) for anticancer therapy and angiogenesis. Magnetic characterization shows that the CoNWs can be concentrated in desired locations under the external magnetic field, which is favorable for anticancer target therapy. Besides, drug loading/release characterization reveals that the CoNWs interact with doxorubicin (DOX) by electrostatic interaction, and accordingly form a composite which can release DOX with temperature increase under near-infrared light (NIR) treatment. And anticancer test reveals that the nanowires loaded with the DOX (CoNWs-DOX) can produce an effective chemo-photothermal synergistic therapeutic effect against murine breast cancer cell lines (4T1) and human osteosarcoma cell lines (MG63) under NIR treatment. Furthermore, angiogenesis assessment reveals that the released cobalt ion from the nanowires can significantly enhance the angiogenesis efficacy after cancer treatment. These results suggest that the constructed anticancer therapeutic system provides a promising multifunctional platform for cancer treatment and postoperative recovery.
Collapse
Affiliation(s)
- Jiankui Zhao
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Yunxiu Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Jiamin Sun
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Huang Zhu
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Yong Chen
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Taosheng Dong
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Rui Sang
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xiangyu Gao
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Weizhong Yang
- College of Materials Science and Engineering, Sichuan University, Chengdu 610065, China.
| | - Yi Deng
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
16
|
Li Z, Li X, Zhu Y, Chen Q, Li B, Zhang F. Protective effects of acetylcholine on hypoxia-induced endothelial-to-mesenchymal transition in human cardiac microvascular endothelial cells. Mol Cell Biochem 2020; 473:101-110. [PMID: 32602017 DOI: 10.1007/s11010-020-03811-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/18/2020] [Indexed: 11/26/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT) has been reported as a key factor in myocardial fibrosis. Acetylcholine (ACh), a neurotransmitter of the vagus nerve, has been confirmed to exert cardio-protective properties with unclear mechanisms. In this study, the specific markers of cell injury, EndMT, inflammation, and autophagy were measured. We found that treatment with ACh prevented hypoxia-induced cell viability reduction and apoptosis in human cardiac microvascular endothelial cells (HCMECs). Additionally, our results indicate that pre-treatment with ACh significantly suppresses hypoxia-induced EndMT and NF-κB activation in HCMECs. ACh also reduced hypoxia-inducible factor (HIF)-1ɑ protein levels under hypoxia. Knock down of HIF-1ɑ enhanced the inhibitory effect of ACh on NF-κB activation. The NF-κB-specific small molecule inhibitor BAY 11-7082, prostaglandin E2, and LY294002 prevented hypoxia-induced EndMT. Moreover, our data show that hypoxia triggers autophagy in HCMECs, and ACh significantly upregulates autophagy activity. Pre-treatment of HCMECs with 3-methyladenine or chloroquine partially reversed ACh-induced EndMT inhibition. These results suggest that ACh may confer protection against hypoxia-induced EndMT through the inhibition of NF-κB and the induction of autophagy.
Collapse
Affiliation(s)
- Zhiyang Li
- Grade 2016 Class 2, The First School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Xuelian Li
- Department of Cardiology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, 266011, China
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yeqian Zhu
- The Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 211166, China
| | - Qiushi Chen
- The Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 211166, China
| | - Bingong Li
- Department of Cardiology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, 266011, China
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Fengxiang Zhang
- The Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 211166, China.
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
17
|
Barillari G. The Impact of Matrix Metalloproteinase-9 on the Sequential Steps of the Metastatic Process. Int J Mol Sci 2020; 21:ijms21124526. [PMID: 32630531 PMCID: PMC7350258 DOI: 10.3390/ijms21124526] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
In industrialized countries, cancer is the second leading cause of death after cardiovascular disease. Most cancer patients die because of metastases, which consist of the self-transplantation of malignant cells in anatomical sites other than the one from where the tumor arose. Disseminated cancer cells retain the phenotypic features of the primary tumor, and display very poor differentiation indices and functional regulation. Upon arrival at the target organ, they replace preexisting, normal cells, thereby permanently compromising the patient's health; the metastasis can, in turn, metastasize. The spread of cancer cells implies the degradation of the extracellular matrix by a variety of enzymes, among which the matrix metalloproteinase (MMP)-9 is particularly effective. This article reviews the available published literature concerning the important role that MMP-9 has in the metastatic process. Additionally, information is provided on therapeutic approaches aimed at counteracting, or even preventing, the development of metastasis via the use of MMP-9 antagonists.
Collapse
Affiliation(s)
- Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 1 via Montpellier, 00133 Rome, Italy
| |
Collapse
|
18
|
Huang H, Wang X, Zhang X, Wang H, Jiang W. Roxadustat attenuates experimental pulmonary fibrosis in vitro and in vivo. Toxicol Lett 2020; 331:112-121. [PMID: 32534005 DOI: 10.1016/j.toxlet.2020.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 05/15/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022]
Abstract
Roxadustat is the first orally administered, small-molecule hypoxia-inducible factor (HIF) prolyl hydroxylase inhibitor that has been submitted for FDA regulatory approval to treat anemia secondary to chronic kidney diseases. Its usage has also been suggested for pulmonary fibrosis; however, the corresponding therapeutic effects remain to be investigated. The in vitro effects of roxadustat on cobalt chloride (CoCl2)-stimulated pulmonary fibrosis with L929 mouse fibroblasts as well as on an in vivo pulmonary fibrosismice model induced with bleomycin (BLM; intraperitoneal injection, 50 mg/kg twice a week for 4 continuous weeks) were investigated. It found that the proliferation of L929 cells was inhibited and the production of collagen I, collagen III, prolyl hydroxylase domain protein 2 (PHD2), HIF-1α, α-smooth muscle actin (α-SMA), connective tissue growth factor (CTGF), transforming growth factor-β1 (TGF-β1) and p-Smad3 were reduced relative to that in the CoCl2 or BLM group after roxadustat treatment. Roxadustat ameliorated pulmonary fibrosis by reducing the pathology score and collagen deposition as well as decreasing the expression of collagen I, collagen III, PHD2, HIF-1α, α-SMA, CTGF, TGF-β1 and p-Smad3/Smad3. Our cumulative results demonstrate that roxadustat administration can attenuate experimental pulmonary fibrosis via the inhibition of TGF-β1/Smad activation.
Collapse
Affiliation(s)
- Haidi Huang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Xin Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Xue Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China
| | - Hongbo Wang
- School of Pharmacy, Yantai University, Yantai, 264003, PR China
| | - Wanglin Jiang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, PR China.
| |
Collapse
|
19
|
Barillari G. The Anti-Angiogenic Effects of Anti-Human Immunodeficiency Virus Drugs. Front Oncol 2020; 10:806. [PMID: 32528888 PMCID: PMC7253758 DOI: 10.3389/fonc.2020.00806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 04/23/2020] [Indexed: 12/17/2022] Open
Abstract
The growth and metastasis of malignant tumors benefit from the formation of blood vessels within the tumor area. There, new vessels originate from angiogenesis (the sprouting of pre-existing neighboring vessels) and/or vasculogenesis (the mobilization of bone marrow-derived endothelial cell precursors which incorporate in tumor vasculature and then differentiate into mature endothelial cells). These events are induced by soluble molecules (the angiogenic factors) and modulated by endothelial cell interactions with the perivascular matrix. Given angiogenesis/vasculogenesis relevance to tumor progression, anti-angiogenic drugs are often employed to buttress surgery, chemotherapy or radiation therapy in the treatment of a wide variety of cancers. Most of the anti-angiogenic drugs have been developed to functionally impair the angiogenic vascular endothelial growth factor: however, this leaves other angiogenic factors unaffected, hence leading to drug resistance and escape. Other anti-angiogenic strategies have exploited classical inhibitors of enzymes remodeling the perivascular matrix. Disappointingly, these inhibitors have been found toxic and/or ineffective in clinical trials, even though they block angiogenesis in pre-clinical models. These findings are stimulating the identification of other anti-angiogenic compounds. In this regard, it is noteworthy that drugs utilized for a long time to counteract human immune deficiency virus (HIV) can directly and effectively hamper molecular pathways leading to blood vessel formation. In this review the mechanisms leading to angiogenesis and vasculogenesis, and their susceptibility to anti-HIV drugs will be discussed.
Collapse
Affiliation(s)
- Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
20
|
Zhang X, Huang H, Zhang G, Li D, Wang H, Jiang W. Raltegravir Attenuates Experimental Pulmonary Fibrosis In Vitro and In Vivo. Front Pharmacol 2019; 10:903. [PMID: 31481891 PMCID: PMC6710384 DOI: 10.3389/fphar.2019.00903] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022] Open
Abstract
Raltegravir, an inhibitor of human immunodeficiency virus-1 (HIV-1) integrase, has been used to treat HIV/acquired immunodeficiency syndrome; however, its therapeutic effects on pulmonary fibrosis have not been investigated. In this study, the in vitro effects of raltegravir (RAV) on transforming growth factor beta 1 (TGF-β1)-induced pulmonary fibrosis on L929 mouse fibroblasts were investigated. In addition, the effects of RAV on an in vivo pulmonary fibrosis model induced by intratracheal instillation of bleomycin were investigated. The proliferation of L929 cells was inhibited after RAV treatment. Meanwhile, the in vitro and in vivo protein expression of nucleotide-binding oligomerization domain-like receptor 3 (NLRP3), high-mobility group box 1 (HMGB1), toll-like receptor 4 (TLR4), prolyl hydroxylase domain protein 2, phosphorylated nuclear factor-κB (p-NF-κB), hypoxia-inducible factor-1α (HIF-1α), collagens I and III was reduced relative to TGF-β1 or the bleomycin group. Raltegravir ameliorated pulmonary fibrosis by reducing the pathology score, collagen deposition, and expression of α-smooth muscle actin, NLRP3, HMGB1, TLR4, inhibitor of kappa B, p-NF-κB, HIF-1α, collagen I, and collagen III. The results of this study demonstrate that RAV attenuated experimental attenuates pulmonary fibrosis by inhibiting NLRP3 activation.
Collapse
Affiliation(s)
- Xue Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Haidi Huang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Guanghua Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Defang Li
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Hongbo Wang
- School of Pharmacy, Yantai University, Yantai, China
| | - Wanglin Jiang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| |
Collapse
|