1
|
Cui Y, Yang K, Guo C, Xia Z, Jiang B, Xue Y, Song B, Hu W, Zhang M, Wei Y, Zhang C, Zhang S, Fang J. Carbon monoxide as a negative feedback mechanism on HIF-1α in the progression of metabolic-associated fatty liver disease. Nitric Oxide 2024; 153:1-12. [PMID: 39369813 DOI: 10.1016/j.niox.2024.10.001] [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: 07/24/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) encompasses various chronic liver conditions, yet lacks approved drugs. Hypoxia-inducible factor-1α (HIF-1α) is pivotal in MAFLD development. Our prior research highlighted the efficacy of the nano-designed carbon monoxide (CO) donor, targeting HIF-1α in a mouse hepatic steatosis model. Given heme oxygenase-1 (HO-1, a major downstream molecule of HIF-1α) as the primary source of intrinsic CO, we hypothesized that upregulation of HO-1/CO, responsive to HIF-1α, forms a negative feedback loop regulating MAFLD progression. In this study, we explored the potential negative feedback mechanism of CO on HIF-1α and its downstream effects on MAFLD advancement. HIF-1α emerges early in hepatic steatosis induced by a high-fat (HF) diet, triggering increased HO-1 and inflammation. SMA/CORM2 effectively suppresses HIF-1α and steatosis progression when administered within the initial week of HF diet initiation but loses impact later. In adipose tissues, concurrent metabolic dysfunction and inflammation with HIF-1α activation suggest adipose tissue expansion initiates HF-induced steatosis, triggering hypoxia and liver inflammation. Notably, in an in vitro study using mouse hepatocytes treated with fatty acids, downregulating HO-1 intensified HIF-1α induction at moderate fatty acid concentrations. However, this effect diminished at high concentrations. These results suggest the HIF-1α-HO-1-CO axis as a feedback loop under physiological and mild pathological conditions. Excessive HIF-1α upregulation in pathological conditions overwhelms the CO feedback loop. Additional CO application effectively suppresses HIF-1α and disease progression, indicating potential application for MAFLD control.
Collapse
Affiliation(s)
- Yingying Cui
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, 750000, China; Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Kai Yang
- Department of Medical Technology, Anhui Medical College, No.632, Furong Road, Hefei, Anhui Province, China
| | - Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Zhengmei Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Benchun Jiang
- Department of Gastricintestinal Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanni Xue
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Weirong Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Mingjie Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanyan Wei
- Department of Infectious Disease, the First Affiliated Hospital of Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Shichen Zhang
- Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China; Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto, 860-0082, Japan.
| |
Collapse
|
2
|
Zhuang J, Zhang H, Wu J, Hu D, Meng T, Xue J, Xu H, Wang G, Wang H, Zhang G. Redox-Responsive AIEgen Diselenide-Covalent Organic Framework Composites Targeting Hepatic Macrophages for Treatment of Drug-induced Liver Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402656. [PMID: 39140196 DOI: 10.1002/smll.202402656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/03/2024] [Indexed: 08/15/2024]
Abstract
The escalating misuse of antipyretic and analgesic drugs, alongside the rising incidents of acute drug-induced liver injury, underscores the need for a precisely targeted drug delivery system. Herein, two isoreticular covalent organic frameworks (Se-COF and Se-BCOF) are developed by Schiff-base condensation of emissive tetraphenylethylene and diselenide-bridged monomers. Leveraging the specific affinity of macrophages for mannose, the first precise targeting of these COFs to liver macrophages is achieved. The correlation is also explored between the therapeutic effects of COFs and the NLRP3/ASC/Caspase-1 signaling pathway. Utilizing this innovative delivery vehicle, the synergistic delivery of matrine and berberine are accomplished, compounds extracted from traditional Chinese medicine. This approach not only demonstrated the synergistic effects of the drugs but also mitigated their toxicity. Notably, berberine, through phosphorylation of JNK and up-regulation of nuclear Nrf-2 and its downstream gene Mn-SOD expression, simultaneously countered excessive ROS and suppressed the activation of the NLRP3/ASC/Caspase-1 signaling pathway in injured liver tissues. This multifaceted approach proved highly effective in safeguarding against acute drug-induced liver injury, ultimately restoring liver health to normalcy. These findings present a novel and promising strategy for the treatment of acute drug-induced liver injury.
Collapse
Affiliation(s)
- Jialu Zhuang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Hao Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Jin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Danyou Hu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Tao Meng
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jing Xue
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Hanyang Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Guiyang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
3
|
Wu Y, Chen X, Zeng Z, Chen B, Wang Z, Song Z, Xie H. Self-assembled carbon monoxide nanogenerators managing sepsis through scavenging multiple inflammatory mediators. Bioact Mater 2024; 39:595-611. [PMID: 38883313 PMCID: PMC11179263 DOI: 10.1016/j.bioactmat.2024.04.013] [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: 01/09/2024] [Revised: 03/29/2024] [Accepted: 04/14/2024] [Indexed: 06/18/2024] Open
Abstract
Sepsis, a life-threatening syndrome of organ damage resulting from dysregulated inflammatory response, is distinguished by overexpression of inflammatory cytokines, excessive generation of reactive oxygen/nitrogen species (RONS), heightened activation of pyroptosis, and suppression of autophagy. However, current clinical symptomatic supportive treatment has failed to reduce the high mortality. Herein, we developed self-assembled multifunctional carbon monoxide nanogenerators (Nano CO), as sepsis drug candidates, which can release CO in response to ROS, resulting in clearing bacteria and activating the heme oxygenase-1/CO system. This activation strengthened endogenous protection and scavenged multiple inflammatory mediators to alleviate the cytokine storm, including scavenging RONS and cfDNA, inhibiting macrophage activation, blocking pyroptosis and activating autophagy. Animal experiments show that Nano CO has a good therapeutic effect on mice with LPS-induced sepsis, which is manifested in hypothermia recovery, organ damage repair, and a 50% decrease in mortality rates. Taken together, these results illustrated the efficacy of multifunctional Nano CO to target clearance of multiple mediators in sepsis treatment and act against other refractory inflammation-related diseases.
Collapse
Affiliation(s)
- Yang Wu
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Ångmedicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xia Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bei Chen
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhenxing Wang
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Ångmedicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhiyong Song
- State Key Laboratory of Agriculture Microbiology, College of Science, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Hui Xie
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Ångmedicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
4
|
Xu S, Cheng B, Xie B, He N. Haeme Oxygenas-1-Induced Liver Regeneration Protects Graft Against Small-for-Size Syndrome in Rats. Transplant Proc 2024; 56:335-342. [PMID: 38360463 DOI: 10.1016/j.transproceed.2023.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/20/2023] [Accepted: 12/31/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Haeme oxygenase (HO-1) affords protection against ischemia/reperfusion (I/R) injury; however, its effects on liver regeneration remain poorly explored. Our previous studies have shown that HO-1 is probably involved in liver regeneration, but its role in small-for-size syndrome (SFSS) is still unclear. Therefore, this study aims to investigate the effects of HO-1 on small-for-size graft (SFSG) and the underlying mechanism. METHODS Knockout of HO-1 rats by TALEN technique. Immunohistochemistry was used to detect HO-1 nuclear translocation. Haeme oxygenase activity was measured by detecting the amount of carbon monoxide (CO) generated from cell lysates. Flow cytometry was used to detect cell apoptosis and cell cycle. Western blot were performed to measure the expression level of HO-1 protein. RESULTS We identified that HO-1 was involved in SFSG regeneration; HO-1-knockout rats demonstrated significantly decreased liver proliferation and recovery. Interestingly, our results showed HO-1-induced SFSG regeneration was more likely to be the primary protector against SFSS than IRI. Furthermore, we verified the nuclear translocation of HO-1 and its protective effect on hypoxia/reoxygenation (H/R) damage in clone9 cells. Our results indicated that the HO-1 protein itself rather than heme breakdown metabolites might play a key role in liver regeneration. CONCLUSIONS The HO-1 protein itself rather than its metabolites possess a protective effect on small-for-size graft (SFSG) against SFSS via nuclear translocation.
Collapse
Affiliation(s)
- Shiguo Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Bing Cheng
- Department of Department of Geriatric Medicine, Shulan (Hangzhou) Hospital, Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, PR China
| | - Bo Xie
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ning He
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
5
|
Fan M, Zhou Z, Zhu W, Li M, Tu Y, Yu Z, Li J, Zhang M, Liang K. Reinforced dentin remineralization via a novel dual-affinity peptide. Dent Mater 2024; 40:254-266. [PMID: 37989605 DOI: 10.1016/j.dental.2023.11.009] [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: 07/25/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 11/23/2023]
Abstract
OBJECTIVES In light of the constantly flowing saliva, anti-caries remineralization agents are inclined to be taken away. Owing to their limited residence time, the remineralization effect is not as desirable as expected. Hence, our study aimed to synthesize a novel peptide (DGP) with high affinity to both collagen fibrils and hydroxyapatite, and investigated its dentin remineralization efficacy in vitro and anti-caries capability in vivo. METHODS DGP was synthesized through Fmoc solid-phase reaction. The binding ability and interaction mechanism of DGP to demineralized dentin were investigated. Dentin specimens were demineralized, then treated with DGP and deionized water respectively. The specimens were incubated in artificial saliva and in-vitro remineralization effectiveness was analyzed after 14 days. The rat caries model was established to further scrutinize the in-vivo efficacy of caries prevention. RESULTS DGP possesses an enhanced adhesion force of 12.29 ± 1.12 nN to demineralized dentin. The favorable adsorption capacity is ascribed to the stable hydrogen bonds between S2P-101 and ASP-100 of DGP and GLY33 and PRO-16 of collagen fibers. Abundant mineral deposits and remarkable tubule occlusion were observed in the DGP group. DGP-treated dentin obtained notable microhardness recovery and higher mineral content after a 14-day remineralization regimen. DGP also demonstrated potent caries prevention in vivo, with substantially fewer carious lesions and significantly lower Keyes scoring. SIGNIFICANCE DGP proves to possess a high affinity to demineralized dentin regardless of saliva flowing, thus enhancing remineralization potency significantly in vitro and in vivo, potential for dental caries prevention and combatting initial dentin caries clinically.
Collapse
Affiliation(s)
- Menglin Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zilin Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wanchun Zhu
- Chongqing Medical University Stomatology College, Chongqing 401147, China
| | - Meng Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuanyuan Tu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhaohan Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Min Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Kunneng Liang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Cao Z, Lu P, Li L, Geng Q, Lin L, Yan L, Zhang L, Shi C, Li L, Zhao N, He X, Tan Y, Lu C. Bioinformatics-led discovery of liver-specific genes and macrophage infiltration in acute liver injury. Front Immunol 2023; 14:1287136. [PMID: 38130716 PMCID: PMC10733525 DOI: 10.3389/fimmu.2023.1287136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Background Acute liver injury (ALI) is an important global health concern, primarily caused by widespread hepatocyte cell death, coupled with a complex immune response and a lack of effective remedies. This study explores the underlying mechanisms, immune infiltration patterns, and potential targets for intervention and treatment ALI. Methods The datasets of acetaminophen (APAP), carbon tetrachloride (CCl4), and lipopolysaccharide (LPS)-induced ALI were obtained from the GEO database. Differentially expressed genes (DEGs) were individually identified using the limma packages. Functional enrichment analysis was performed using KEGG, GO, and GSEA methods. The overlapping genes were extracted from the three datasets, and hub genes were identified using MCODE and CytoHubba algorithms. Additionally, PPI networks were constructed based on the String database. Immune cell infiltration analysis was conducted using ImmuCellAI, and the correlation between hub genes and immune cells was determined using the Spearman method. The relationship between hub genes, immune cells, and biochemical indicators of liver function (ALT, AST) was validated using APAP and triptolide (TP) -induced ALI mouse models. Results Functional enrichment analysis indicated that all three ALI models were enriched in pathways linked to fatty acid metabolism, drug metabolism, inflammatory response, and immune regulation. Immune analysis revealed a significant rise in macrophage infiltration. A total of 79 overlapping genes were obtained, and 10 hub genes were identified that were consistent with the results of the biological information analysis after screening and validation. Among them, Clec4n, Ms4a6d, and Lilrb4 exhibited strong associations with macrophage infiltration and ALI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Chen M, Chen T, Bai J, He S, Luo M, Zeng Y, Peng W, Zhao Y, Wang J, Zhu X, Zhi W, Weng J, Zhang K, Zhang X. A Nature-Inspired Versatile Bio-Adhesive. Adv Healthc Mater 2023; 12:e2301560. [PMID: 37548628 DOI: 10.1002/adhm.202301560] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/17/2023] [Indexed: 08/08/2023]
Abstract
The application of most hydrogel bio-adhesives is greatly limited due to their high swelling, low underwater adhesion, and single function. Herein, a spatial multi-level physical-chemical and bio-inspired in-situ bonding strategy is proposed, to develop a multifunctional hydrogel bio-glue using polyglutamic acid (PGA), tyramine hydrochloride (TYR), and tannic acid (TA) as precursors and 4-(4,6-dimethoxytriazine-2-yl) -4-methylmorpholine hydrochloride(DMTMM) as condensation agent, which is used for tissue adhesion, hemostasis and repair. By introducing TYR and TA into the PGA chain, it is demonstrated that not only can the strong adhesion of bio-glue to the surface of various fresh tissues and wet materials be realized through the synergistic effect of spatial multi-level physical and chemical bonding, but also this glue can be endowed with the functions of anti-oxidation and hemostasis. The excellent performance of such bio-glue in the repair of the wound, liver, and cartilage is achieved, showing a great potential in clinical application for such bio-glue. This study will open up a brand-new avenue for the development of multifunctional hydrogel biological adhesive.
Collapse
Affiliation(s)
- Mingxia Chen
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Taijun Chen
- Chengdu University of Traditional Chinese Medicine, School of Intelligent Medicine, Chengdu, 611137, China
| | - Jiafan Bai
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Siyuan He
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Minyue Luo
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Yili Zeng
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Wenzhen Peng
- Department of Biochemistry and Molecular Biology, College of Basic and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yuancong Zhao
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Jianxin Wang
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Wei Zhi
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Jie Weng
- Key Laboratory of Advance Technologies of Materials, Ministry of Education, College of Medicine, School of Materials Science and Engineering, Southwest Jiaotong University, 610031, Chengdu, China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
8
|
Cui Y, Guo C, Xia Z, Xue Y, Song B, Hu W, He X, Liang S, Wei Y, Zhang C, Wang H, Xu D, Zhang S, Fang J. Exploring the therapeutic potential of a nano micelle containing a carbon monoxide-releasing molecule for metabolic-associated fatty liver disease by modulating hypoxia-inducible factor-1α. Acta Biomater 2023; 169:500-516. [PMID: 37574157 DOI: 10.1016/j.actbio.2023.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/20/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Metabolic-associated fatty liver disease (MAFLD) encompasses a spectrum of chronic liver diseases, including steatohepatitis, cirrhosis, and liver cancer. Despite the increasing prevalence and severity of MAFLD, no approved pharmacological interventions are currently available. Hypoxia-inducible factor-1α (HIF-1α) has emerged as a crucial early mediator in the pathogenesis of MAFLD. Previously, we demonstrated the potent anti-inflammatory properties of the nano-designed carbon monoxide (CO) donor, styrene maleic acid copolymer (SMA) encapsulating CO-releasing molecule (SMA/CORM2), which effectively suppressed HIF-1α in various inflammatory disorders. Here, we investigated the therapeutic potential of SMA/CORM2 in a mouse model of MAFLD induced by a high-fat methionine- and choline-deficient (HF-MCD) diet. Following 4 weeks of HF-MCD diet consumption, we observed pronounced hepatic lipid accumulation accompanied by disrupted lipid metabolism, polarization of macrophages towards the pro-inflammatory M1 phenotype, activation of the NLRP3 inflammasome, and upregulation of the TGF-β fibrosis signaling pathway. Notably, the early and upstream event driving these pathological changes was the upregulation of HIF-1α. Treatment with SMA/CORM2 (10 mg/kg, three times per week) led to a significant increase in CO levels in both the circulation and liver, resulting in remarkable suppression of HIF-1α expression even before the onset of apparent pathological changes induced by the HF-MCD diet. Consequently, SMA/CORM2 administration exerted a significantly protective and therapeutic effect on MAFLD. In vitro studies using hepatocytes treated with high concentrations of fatty acids further supported these findings, as knockdown of HIF-1α using short hairpin RNA (shRNA) elicited similar effects to SMA/CORM2 treatment. Collectively, our results highlight the therapeutic potential of SMA/CORM2 in the management of MAFLD through suppression of HIF-1α. We anticipate that SMA/CORM2, with its ability to modulate HIF-1α expression, may hold promise for future applications in the treatment of MAFLD. STATEMENT OF SIGNIFICANCE: Carbon monoxide (CO) is a crucial gaseous signaling molecule that plays a vital role in maintaining homeostasis and is a potential target for treating many inflammatory diseases. Developing drug delivery systems that can deliver CO stably and target specific tissues is of great interest. Our team previously developed a nano micellar CO donor, SMA/CORM2, which exhibits superior bioavailability to native CORM2 and shows therapeutic potential in many inflammatory disease models. In this study, we showed that SMA/CORM2, through controlled CO release, significantly ameliorated steatohepatitis and liver fibrosis induced by an HF-MCD diet by suppressing an HIF-1α mediated inflammatory cascade. These findings provide new insight into the anti-inflammatory function of CO and a promising approach for controlling metabolic-associated fatty liver disease.
Collapse
Affiliation(s)
- Yingying Cui
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Peking University First Hispital Ningxia Women and Children's Hosptical (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan 750000, China
| | - Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Zhengmei Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Yanni Xue
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Weirong Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Xue He
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Shimin Liang
- Department of Gastroenterology, Anhui Provincial Key Laboratory of Digestive Disease, the First Affiliated Hospital of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yanyan Wei
- Department of Infectious Disease, the First Affiliated Hospital of Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Dexiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China
| | - Shichen Zhang
- School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei 230601, Anhui, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230022, Anhui, China; School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei 230601, Anhui, China; Faculty of Pharmaceutical Science, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan.
| |
Collapse
|
9
|
İpek E, Aşıcı GSE, Kurt BK, Epikmen ET, Özsoy ŞY, Tunca R. Carbon monoxide (CO) derived from the CO-releasing molecule CORM-2 reduces peritoneal adhesion formation in a rat model. Mol Biol Rep 2023; 50:8483-8495. [PMID: 37639152 DOI: 10.1007/s11033-023-08753-2] [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: 05/24/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Although low-dose carbon monoxide (CO) administration has been shown to have an anti-fibrotic effect in various fibrotic diseases, its effects on peritoneal adhesion (PA), one of the postoperative complications, are not elucidated. In this study, the effect of CO-releasing tricarbonyldichlororuthenium (II) dimer (CORM-2) administration on the formation of PA and the underlying factors of its potential effect were investigated. METHODS AND RESULTS After the induction of PA, rats were divided into four groups with 8 rats in each group. The rats received either (i) dimethyl sulfoxide:saline solution (1:10) as a vehicle, (ii) 2.5 mg/kg CORM-2, (iii) 5 mg/kg CORM-2, or (iv) inactive (i) CORM (iCORM) intragastrically every day for a duration of 7 days. PA was not induced in rats (n = 8) designated as sham controls. Gross, histological, immunohistochemical and quantitative real-time polymerase chain reaction analyses were performed to evaluate the effectiveness of CORM-2 administration. Gross analysis showed that CORM-2 administration reduced PA formation compared to rats treated with vehicle. Histological and immunohistochemical examinations showed that increased collagen deposition, myofibroblast accumulation, microvessel density, and M1 macrophage count in the peritoneal fibrosis area of vehicle-treated rats decreased following CORM-2 treatments. PCR analyses showed that CORM-2 treatments decreased hypoxia-induced Hif1a, profibrotic Tgfb1, ECM components Col1a1 and Col3a1, collagen degradation suppressor Timp1, fibrinolysis inhibitor Serpine1, and pro-inflammatory Tnf mRNA expressions, while increasing the M2 macrophage marker Arg1 mRNA expression. CONCLUSIONS These results suggested that CORM-2 administration reduces PA formation by affecting adhesiogenic processes such as pro-inflammatory response, fibrinolytic system, angiogenesis and fibrogenesis.
Collapse
Affiliation(s)
- Emrah İpek
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey.
| | - Gamze Sevri Ekren Aşıcı
- Department of Biochemistry, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Büşra Kibar Kurt
- Department of Surgery, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Erkmen Tuğrul Epikmen
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Şule Yurdagül Özsoy
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Recai Tunca
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| |
Collapse
|
10
|
Yang Y, Jia X, Qu M, Yang X, Fang Y, Ying X, Zhang M, Wei J, Pan Y. Exploring the potential of treating chronic liver disease targeting the PI3K/Akt pathway and polarization mechanism of macrophages. Heliyon 2023; 9:e17116. [PMID: 37484431 PMCID: PMC10361319 DOI: 10.1016/j.heliyon.2023.e17116] [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: 02/28/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 07/25/2023] Open
Abstract
Chronic liver disease is a significant public health issue that can lead to considerable morbidity and mortality, imposing an enormous burden on healthcare resources. Understanding the mechanisms underlying chronic liver disease pathogenesis and developing effective treatment strategies are urgently needed. In this regard, the activation of liver resident macrophages, namely Kupffer cells, plays a vital role in liver inflammation and fibrosis. Macrophages display remarkable plasticity and can polarize into different phenotypes according to diverse microenvironmental stimuli. The polarization of macrophages into M1 pro-inflammatory or M2 anti-inflammatory phenotypes is regulated by complex signaling pathways such as the PI3K/Akt pathway. This review focuses on investigating the potential of using plant chemicals targeting the PI3K/Akt pathway for treating chronic liver disease while elucidating the polarization mechanism of macrophages under different microenvironments. Studies have demonstrated that inhibiting M1-type macrophage polarization or promoting M2-type polarization can effectively combat chronic liver diseases such as alcoholic liver disease, non-alcoholic fatty liver disease, and liver fibrosis. The PI3K/Akt pathway acts as a pivotal modulator of macrophage survival, migration, proliferation, and their responses to metabolism and inflammatory signals. Activating the PI3K/Akt pathway induces anti-inflammatory cytokine expression, resulting in the promotion of M2-like phenotype to facilitate tissue repair and resolution of inflammation. Conversely, inhibiting PI3K/Akt signaling could enhance the M1-like phenotype, which exacerbates liver damage. Targeting the PI3K/Akt pathway has tremendous potential as a therapeutic strategy for regulating macrophage polarization and activity to treat chronic liver diseases with plant chemicals, providing new avenues for liver disease treatment.
Collapse
Affiliation(s)
- Yaqian Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiaotao Jia
- Department of Neurology, The Affifiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710003, PR China
| | - Mengyang Qu
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xinmao Yang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yan Fang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiaoping Ying
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Meiqian Zhang
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Jing Wei
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yanfang Pan
- Department of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| |
Collapse
|
11
|
Zheng J, Yang N, Wan Y, Cheng W, Zhang G, Yu S, Yang B, Liu X, Chen X, Ding X, Wu L, Yu X. Celastrol-loaded biomimetic nanodrug ameliorates APAP-induced liver injury through modulating macrophage polarization. J Mol Med (Berl) 2023:10.1007/s00109-023-02321-8. [PMID: 37129620 DOI: 10.1007/s00109-023-02321-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023]
Abstract
Drug-induced liver injury (DILI) is a major concern in clinical treatment as well as postmarketing surveillance, showing an urgent requirement for the development of protective medications. Celastrol (Cel), a highly active natural product extracted from the roots of Tripterygium wilfordii, has a potential liver protective activity due to its antioxidant and anti-inflammatory effects. However, the further application of Cel to DILI remains a challenge because of its short half-life, low solubility, and toxic side effects. Herein, we developed a Cel-loaded biomimetic nanodrug based on erythrocyte membrane vesicles (EMV) for protecting the liver from acetaminophen (APAP)-induced liver injury. The Cel-loaded EMV (C-EMV) with lower cytotoxicity had a well-sustained release effect and exhibited excellent ability for liver accumulation under physiological and pathological conditions. By suppressing the inflammatory response of pro-inflammatory macrophage M1 polarization while stimulating anti-inflammatory macrophage M2 polarization, C-EMV could significantly alleviate the primary pathological manifestations related to liver injury, including aberrant elevation of biochemical indicators, histopathological alterations, neutrophil infiltration as well as hepatocyte DNA fragmentation. The macrophage depletion experiment further demonstrated that the protective effect of C-EMV on APAP-induced liver injury appeared to be dependent on hepatic macrophages. Therefore, C-EMV as a biomimetic nanodrug exhibits great potential for attenuating the progress of DILI, providing a new approach to protecting the liver from DILI as well as other liver inflammatory diseases through a targeted nanodelivery system. KEY MESSAGES: EMV biomimetic nanocarrier has good monodispersity and sustained-release property. EMV biomimetic nanocarrier displays excellent liver-targeting capability under physiological and pathological conditions. C-EMV biomimetic nanodrug with lower cytotoxicity regulates macrophage polarization in vitro and in vivo. C-EMV biomimetic nanodrug can significantly alleviate APAP-induced liver injury. The protective effect of C-EMV on APAP-induced liver injury is dependent on hepatic macrophages.
Collapse
Affiliation(s)
- Jing Zheng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
- The People's Hospital of China Three Gorges University, Yichang, China
| | - Ni Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Yingying Wan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Wenjing Cheng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Gan Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Shi Yu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Baoye Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Xinyu Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
| | - Xingyan Chen
- Tong Ji Hospital, Tongji Medical College of HUST, Wuhan, China
| | - Xueliang Ding
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
- Medical College, China Three Gorges University, Yichang, China
- Department of Clinical Laboratory, Affiliated Renhe Hospital of China Three Gorges University, Yichang, China
| | - Ling Wu
- The People's Hospital of China Three Gorges University, Yichang, China.
| | - Xiang Yu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China.
- Institute of Infection and Inflammation, China Three Gorges University, Yichang, China.
- Medical College, China Three Gorges University, Yichang, China.
| |
Collapse
|
12
|
Xue Y, Zhang D, Wei Y, Guo C, Song B, Cui Y, Zhang C, Xu D, Zhang S, Fang J. Polymeric nano-micelle of carbon monoxide donor SMA/CORM2 ameliorates acetaminophen-induced liver injury via suppressing HMGB1/TLR4 signaling pathway. Eur J Pharm Sci 2023; 184:106413. [PMID: 36863618 DOI: 10.1016/j.ejps.2023.106413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/10/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023]
Abstract
Acetaminophen (APAP) overdose-induced hepatotoxicity is the most common cause of acute liver failure. Excessive generation of reactive oxygen species (ROS) and inflammatory responses are the major causes of necrosis and/or necroptosis of the liver cells. Currently, the treatment options for APAP-induced liver injury are very limited, N-acetylcysteine (NAC) is the only approved drug to treat APAP overdose patients. It is of great necessity to develop new therapeutic strategies. In a previous study, we focused on the anti-oxidative, anti-inflammatory signal molecule carbon monoxide (CO), and developed a nano-micelle encapsulating CO donor, i.e., SMA/CORM2. Administration of SMA/CORM2 to the mice exposed to APAP significantly ameliorated the liver injury and inflammatory process, in which modulating macrophage reprogramming plays a critical role. Along this line, in this study, we investigated the potential effect of SMA/CORM2 on toll-like receptor 4 (TLR4) and high mobility group protein B1 (HMGB1) signaling pathways that are known to be closely involved in many inflammatory responses and necroptosis. In a mouse APAP-induced liver injury model, similar to the previous study, SMA/CORM2 at 10 mg/kg remarkably improved the condition of the liver after injury as evidenced by histological examination and liver function. During the process of liver injury triggered by APAP, TLR4 expression gradually increased over time, and it was significantly upregulated as early as 4 h after APAP exposure, whereas, an increase of HMGB1 was a late-stage event. Notably, SMA/CORM2 treatment suppressed significantly both TLR4 and HMGB1, consequently inhibiting the progression of inflammation and liver injury. Compared to CORM2 without SMA modification (native CORM2) of 1 mg/kg that is equivalent to 10 mg/kg of SMA/CORM2 (the amount of CORM2 in SMA/CORM2 is 10% [w/w]), SMA/CORM2 exhibited a much better therapeutic effect, indicating its superior therapeutic efficacy to native CORM2. These findings revealed that SMA/CORM2 protects against APAP-induced liver injury via mechanisms involving the suppression of TLR4 and HMGB1 signaling pathways. Taking together the results in this study and previous studies, SMA/CORM2 exhibits great therapeutic potential for APAP overdose-induced liver injury, we thus anticipate the clinical application of SMA/CORM2 for the treatment of APAP overdose, as well as other inflammatory diseases.
Collapse
Affiliation(s)
- Yanni Xue
- Department of Maternal, Child and Adolescent Health, School of Public Health, and MOE Key Laboratory of Population Health Across Life Cycle/ Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Daoxu Zhang
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 MeiLong Road, Shanghai 200237, China
| | - Yanyan Wei
- Department of Infectious Disease, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Chunyu Guo
- Department of Toxicology, School of Public Health, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Yingying Cui
- Department of Toxicology, School of Public Health, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Dexiang Xu
- Department of Toxicology, School of Public Health, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Shichen Zhang
- School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei 230601, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei 230032, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, and Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei 230032, China; Faculty of Pharmaceutical Science, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan.
| |
Collapse
|
13
|
Gao S, Yang K, Nohara T, Ikeda T, Zhou JR, Yokomizo K, Fang J. Garlicnin B1, an Active Cyclic Sulfide from Garlic, Exhibits Potent Anti-Inflammatory and Anti-Tumor Activities. Antioxidants (Basel) 2023; 12:antiox12040869. [PMID: 37107245 PMCID: PMC10135383 DOI: 10.3390/antiox12040869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
This study aimed to investigate the pharmacological activities of garlicnin B1, a cyclic sulfide compound found abundantly in garlic and structurally similar to onionin A1, which has been shown to possess strong anti-tumor effects. In vitro studies demonstrated that garlicnin B1 significantly reduced intracellular reactive oxygen species triggered by hydrogen peroxide in colon cancer cells. In a mouse colitis model induced by dextran sulfate sodium, garlicnin B1 at a low dose (5 mg/kg) remarkably ameliorated the symptoms and pathological progression. Additionally, garlicnin B1 exhibited considerable tumoricidal activity with an IC50 value of ~20 μM, as observed in cytotoxicity assays. In vivo experiments using the mouse sarcoma S180 transplanted model and the azoxymethane (AOM) or DSS-induced colon cancer model showed that garlicnin B1 effectively suppressed tumor growth in a dose-dependent manner, with marked inhibition at 80 mg/kg. These results suggest that garlicnin B1 has diverse functions that could be achieved by carefully manipulating the dosing regimen. We anticipate that garlicnin B1 has the potential to be used beneficially in the future for the treatment of cancer and inflammatory diseases, although further studies are warranted to elucidate its mechanisms of action.
Collapse
Affiliation(s)
- Shanghui Gao
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Kai Yang
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
- Department of Medical Technology, Anhui Medical College, No. 632, Furong Road, Hefei 230601, China
| | - Toshihiro Nohara
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Tsuyoshi Ikeda
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Jian-Rong Zhou
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Kazumi Yokomizo
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Jun Fang
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| |
Collapse
|
14
|
Zhu L, Luo M, Zhang Y, Fang F, Li M, An F, Zhao D, Zhang J. Free radical as a double-edged sword in disease: Deriving strategic opportunities for nanotherapeutics. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
15
|
Protective effect of hepatocyte-enriched lncRNA-Mir122hg by promoting hepatocyte proliferation in acute liver injury. Exp Mol Med 2022; 54:2022-2035. [PMID: 36424455 PMCID: PMC9722683 DOI: 10.1038/s12276-022-00881-2] [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: 01/19/2022] [Revised: 06/01/2022] [Accepted: 07/10/2022] [Indexed: 11/25/2022] Open
Abstract
Some long noncoding RNAs (lncRNAs), which harbor microRNAs in their gene sequence and are also known as microRNA host gene derived lncRNAs (lnc-MIRHGs), play a dominant role alongside miRNAs, or both perform biological functions synergistically or independently. However, only a small number of lnc-MIRHGs have been identified. Here, multiple liver injury datasets were analyzed to screen and identify the target lncRNA Mir122hg. Mir122hg was mainly enriched in liver tissues with human-mouse homology. In both CCl4-induced acute liver injury and Dgal/LPS-induced fulminant liver failure in mice, Mir122hg was sharply downregulated at the early stage, while a subsequent significant increase was only found in the CCl4 group with liver recovery. Overexpression and silencing assays confirmed that Mir122hg played a protective role in acute injury by promoting hepatocyte proliferation in vivo and in vitro. Consistent with the results of gene enrichment analysis, Mir122hg binding to C/EBPα affected its transcriptional repression, promoted gene transcription of downstream chemokines, Cxcl2, Cxcl3, and Cxcl5, and exerted pro-proliferative effects on hepatocytes through activation of the AKT/GSK-3β/p27 signaling pathway by CXC/CXCR2 complexes. This study identifies a novel lncRNA with protective effects in acute liver injury and demonstrates that the binding of Mir122hg-C/EBPα promotes hepatocyte proliferation via upregulation of CXC chemokine and activation of AKT signaling.
Collapse
|
16
|
Li Q, Chen F, Wang F. The immunological mechanisms and therapeutic potential in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity. Cell Biosci 2022; 12:187. [PMID: 36414987 PMCID: PMC9682794 DOI: 10.1186/s13578-022-00921-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
Acute liver failure caused by drug overdose is a significant clinical problem in developed countries. Acetaminophen (APAP), a widely used analgesic and antipyretic drug, but its overdose can cause acute liver failure. In addition to APAP-induced direct hepatotoxicity, the intracellular signaling mechanisms of APAP-induced liver injury (AILI) including metabolic activation, mitochondrial oxidant stress and proinflammatory response further affect progression and severity of AILI. Liver inflammation is a result of multiple interactions of cell death molecules, immune cell-derived cytokines and chemokines, as well as damaged cell-released signals which orchestrate hepatic immune cell infiltration. The immunoregulatory interplay of these inflammatory mediators and switching of immune responses during AILI lead to different fate of liver pathology. Thus, better understanding the complex interplay of immune cell subsets in experimental models and defining their functional involvement in disease progression are essential to identify novel therapeutic targets for the treatment of AILI. Here, this present review aims to systematically elaborate on the underlying immunological mechanisms of AILI, its relevance to immune cells and their effector molecules, and briefly discuss great therapeutic potential based on inflammatory mediators.
Collapse
Affiliation(s)
- Qianhui Li
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| | - Feng Chen
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| | - Fei Wang
- grid.511083.e0000 0004 7671 2506Division of Gastroenterology, Seventh Affiliated Hospital of Sun Yat-sen University, No.628, Zhenyuan Road, Shenzhen, 518107 China
| |
Collapse
|
17
|
Zhang J, Tong D, Song H, Ruan R, Sun Y, Lin Y, Wang J, Hou L, Dai J, Ding J, Yang H. Osteoimmunity-Regulating Biomimetically Hierarchical Scaffold for Augmented Bone Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202044. [PMID: 35785450 DOI: 10.1002/adma.202202044] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/14/2022] [Indexed: 05/22/2023]
Abstract
Engineering a proper immune response following biomaterial implantation is essential to bone tissue regeneration. Herein, a biomimetically hierarchical scaffold composed of deferoxamine@poly(ε-caprolactone) nanoparticles (DFO@PCL NPs), manganese carbonyl (MnCO) nanosheets, gelatin methacryloyl hydrogel, and a polylactide/hydroxyapatite (HA) matrix is fabricated to augment bone repair by facilitating the balance of the immune system and bone metabolism. First, a 3D printed stiff scaffold with a well-organized gradient structure mimics the cortical and cancellous bone tissues; meanwhile, an inside infusion of a soft hydrogel further endows the scaffold with characteristics of the extracellular matrix. A Fenton-like reaction between MnCO and endogenous hydrogen peroxide generated at the implant-tissue site triggers continuous release of carbon monoxide and Mn2+ , thus significantly lessening inflammatory response by upregulating the M2 phenotype of macrophages, which also secretes vascular endothelial growth factor to induce vascular formation. Through activating the hypoxia-inducible factor-1α pathway, Mn2+ and DFO@PCL NP further promote angiogenesis. Moreover, DFO inhibits osteoclast differentiation and synergistically collaborates with the osteoinductive activity of HA. Based on amounts of data in vitro and in vivo, strong immunomodulatory, intensive angiogenic, weak osteoclastogenic, and superior osteogenic abilities of such an osteoimmunity-regulating scaffold present a profound effect on improving bone regeneration, which puts forward a worthy base and positive enlightenment for large-scale bone defect repair.
Collapse
Affiliation(s)
- Jin Zhang
- College of Chemical Engineering, Qingyuan Innovation Laboratory, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Dongmei Tong
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Honghai Song
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, P. R. China
| | - Renjie Ruan
- College of Chemical Engineering, Qingyuan Innovation Laboratory, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Yifu Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, P. R. China
| | - Yandai Lin
- College of Chemical Engineering, Qingyuan Innovation Laboratory, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Jun Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Linxi Hou
- College of Chemical Engineering, Qingyuan Innovation Laboratory, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
| | - Jiayong Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, P. R. China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, P. R. China
| |
Collapse
|
18
|
Pérez S, Rius-Pérez S. Macrophage Polarization and Reprogramming in Acute Inflammation: A Redox Perspective. Antioxidants (Basel) 2022; 11:antiox11071394. [PMID: 35883885 PMCID: PMC9311967 DOI: 10.3390/antiox11071394] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage polarization refers to the process by which macrophages can produce two distinct functional phenotypes: M1 or M2. The balance between both strongly affects the progression of inflammatory disorders. Here, we review how redox signals regulate macrophage polarization and reprogramming during acute inflammation. In M1, macrophages augment NADPH oxidase isoform 2 (NOX2), inducible nitric oxide synthase (iNOS), synaptotagmin-binding cytoplasmic RNA interacting protein (SYNCRIP), and tumor necrosis factor receptor-associated factor 6 increase oxygen and nitrogen reactive species, which triggers inflammatory response, phagocytosis, and cytotoxicity. In M2, macrophages down-regulate NOX2, iNOS, SYNCRIP, and/or up-regulate arginase and superoxide dismutase type 1, counteract oxidative and nitrosative stress, and favor anti-inflammatory and tissue repair responses. M1 and M2 macrophages exhibit different metabolic profiles, which are tightly regulated by redox mechanisms. Oxidative and nitrosative stress sustain the M1 phenotype by activating glycolysis and lipid biosynthesis, but by inhibiting tricarboxylic acid cycle and oxidative phosphorylation. This metabolic profile is reversed in M2 macrophages because of changes in the redox state. Therefore, new therapies based on redox mechanisms have emerged to treat acute inflammation with positive results, which highlights the relevance of redox signaling as a master regulator of macrophage reprogramming.
Collapse
|
19
|
Wang J, Zhang L, Shi Q, Yang B, He Q, Wang J, Weng Q. Targeting innate immune responses to attenuate acetaminophen-induced hepatotoxicity. Biochem Pharmacol 2022; 202:115142. [PMID: 35700755 DOI: 10.1016/j.bcp.2022.115142] [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: 04/25/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/02/2022]
Abstract
Acetaminophen (APAP) hepatotoxicity is an important cause of acute liver failure, resulting in massive deaths in many developed countries. Currently, the metabolic process of APAP in the body has been well studied. However, the underlying mechanism of APAP-induced liver injury remains elusive. Increasing clinical and experimental evidences indicate that the innate immune responses are involved in the pathogenesis of APAP-induced acute liver injury (AILI), in which immune cells have dual roles of inducing inflammation to exacerbate hepatotoxicity and removing dead cells and debris to help liver regeneration. In this review, we summarize the latest findings of innate immune cells involved in AILI, particularly emphasizing the activation of innate immune cells and their different roles during the injury and repair phases. Moreover, current available treatments are discussed according to the different roles of innate immune cells in the development of AILI. This review aims to update the knowledge about innate immune responses in the pathogenesis of AILI, and provide potential therapeutic interventions for AILI.
Collapse
Affiliation(s)
- Jincheng Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lulu Zhang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qi Shi
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
20
|
Zhai X, Dai T, Chi Z, Zhao Z, Wu G, Yang S, Dong D. Naringin alleviates acetaminophen-induced acute liver injury by activating Nrf2 via CHAC2 upregulation. ENVIRONMENTAL TOXICOLOGY 2022; 37:1332-1342. [PMID: 35179299 DOI: 10.1002/tox.23487] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/04/2022] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Severe acetaminophen (APAP)-induced hepatic damage is the second most common cause for hepatic transplantation. Clinically, hepatic damage caused by APAP is treated using N-acetyl-L-cysteine, which can induce numerous side effects. Naringin, a bioflavonoid abundant in grapefruit and other citrus fruits, displays marked antiinflammatory and antioxidant activities. Herein, we aimed to investigate the potential mechanism underlying naringin-mediated protection against APAP-induced acute hepatotoxicity. We observed that naringin afforded protection against APAP-induced acute liver failure in mice. Importantly, pretreatment with naringin before APAP administration further increased antioxidant enzyme expression, inhibited the production of proinflammatory cytokines, and activated apoptotic pathways. Furthermore, we observed that the protective effect was associated with the upregulation of cation transport regulator-like protein 2 (CHAC2) and nuclear factor erythroid derived-2-related factor 2 (Nrf2). Notably, CHAC2 knockdown inhibited Nrf2 activation and naringin-mediated antioxidant, antiinflammatory, and antiapoptotic effects in APAP-induced liver injury. Likewise, si-Nrf2 blocked the protective effect of naringin against APAP-induced liver injury. Collectively, our results indicate that naringin may be a potent CHAC2 activator, alleviating APAP-induced hepatitis via CHAC2-mediated activation of the Nrf2 pathway. These data provide new insights into mechanisms through which CHAC2 regulates APAP-induced liver injury by targeting Nrf2, which should be considered a novel therapeutic target.
Collapse
Affiliation(s)
- Xiaohan Zhai
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tiantian Dai
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Zhongchao Chi
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Zirui Zhao
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Gaolei Wu
- Department of Pharmacy, Dalian Municipal Women and Children's Medical Center, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Zafonte RD, Wang L, Arbelaez CA, Dennison R, Teng YD. Medical Gas Therapy for Tissue, Organ, and CNS Protection: A Systematic Review of Effects, Mechanisms, and Challenges. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104136. [PMID: 35243825 PMCID: PMC9069381 DOI: 10.1002/advs.202104136] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/10/2022] [Indexed: 05/13/2023]
Abstract
Gaseous molecules have been increasingly explored for therapeutic development. Here, following an analytical background introduction, a systematic review of medical gas research is presented, focusing on tissue protections, mechanisms, data tangibility, and translational challenges. The pharmacological efficacies of carbon monoxide (CO) and xenon (Xe) are further examined with emphasis on intracellular messengers associated with cytoprotection and functional improvement for the CNS, heart, retina, liver, kidneys, lungs, etc. Overall, the outcome supports the hypothesis that readily deliverable "biological gas" (CO, H2 , H2 S, NO, O2 , O3 , and N2 O) or "noble gas" (He, Ar, and Xe) treatment may preserve cells against common pathologies by regulating oxidative, inflammatory, apoptotic, survival, and/or repair processes. Specifically, CO, in safe dosages, elicits neurorestoration via igniting sGC/cGMP/MAPK signaling and crosstalk between HO-CO, HIF-1α/VEGF, and NOS pathways. Xe rescues neurons through NMDA antagonism and PI3K/Akt/HIF-1α/ERK activation. Primary findings also reveal that the need to utilize cutting-edge molecular and genetic tactics to validate mechanistic targets and optimize outcome consistency remains urgent; the number of neurotherapeutic investigations is limited, without published results from large in vivo models. Lastly, the broad-spectrum, concurrent multimodal homeostatic actions of medical gases may represent a novel pharmaceutical approach to treating critical organ failure and neurotrauma.
Collapse
Affiliation(s)
- Ross D. Zafonte
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Neurotrauma Recovery Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
- Spaulding Research InstituteSpaulding Rehabilitation Hospital NetworkBostonMA02129USA
| | - Lei Wang
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| | - Christian A. Arbelaez
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| | - Rachel Dennison
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| | - Yang D. Teng
- Department of Physical Medicine and RehabilitationHarvard Medical SchoolBostonMA02115USA
- Neurotrauma Recovery Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
- Spaulding Research InstituteSpaulding Rehabilitation Hospital NetworkBostonMA02129USA
- Laboratory of SCI, Stem Cell and Recovery Neurobiology Research, Department of Physical Medicine and RehabilitationSpaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical SchoolBostonMA02129USA
| |
Collapse
|
22
|
Xia Z, Zhang C, Guo C, Song B, Hu W, Cui Y, Xue Y, Xia M, Xu D, Zhang S, Fang J. Nanoformulation of a carbon monoxide releasing molecule protects against cyclosporin A-induced nephrotoxicity and renal fibrosis via the suppression of the NLRP3 inflammasome mediated TGF-β/Smad pathway. Acta Biomater 2022; 144:42-53. [PMID: 35304324 DOI: 10.1016/j.actbio.2022.03.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 12/14/2022]
Abstract
Cyclosporin A (CsA) induced nephrotoxicity i.e., renal fibrosis is a critical clinical problem in renal transplant patients, in which chronic inflammatory response is the major cause. Previously, we developed a nano-drug delivery system for carbon monoxide (CO), a multi-functional gaseous molecule with a potent anti-inflammatory effect, i.e., SMA/CORM2, which showed therapeutic potential in several inflammatory disease models. Accordingly, in this study, we explored the potential and usefulness of SMA/CORM2 on CsA induced renal fibrosis. When mice were exposed to CsA for 4 weeks, severe injuries in the kidney as revealed by decreased kidney function and histological examination, and activation of NLRP3 inflammasome, as well as renal fibrosis along with the upregulation of transforming growth factor β (TGFβ)/Smad signaling molecule were observed, whereas SMA/CORM2 (1 mg/kg) treatment remarkably ameliorated the inflammatory injury and fibrosis in the kidney. CO is the major effector molecule of SMA/CORM2 which significantly suppressed the activation of NLRP3 inflammasome, and induced the downregulation of TGFβ/Smad signaling. Inhibition of NLRP3 inflammasome by its inhibitor MCC950 also similarly decreased TGFβ/Smad expression and subsequently improved kidney injury and renal fibrosis, suggesting SMA/CORM2 induced suppression of TGFβ/Smad signaling and renal signaling via an NLRP3 inflammasome-dependent pathway. Compared to native CORM2, SMA/CORM2 exhibited better therapeutic/preventive effects owing to its superior water-solubility and bioavailability. These findings strongly indicated the applicability of SMA/CORM2 as an enhanced permeability and retention (EPR) effect-based nanomedicine for CsA induced renal fibrosis as well as other inflammatory diseases. STATEMENT OF SIGNIFICANCE: Carbon monoxide (CO) is an important gaseous signaling molecule that plays a crucial role in the maintenance of homeostasis. Because of its versatile functions, it exhibits the potential as the target molecule for many diseases, including inflammatory diseases and cancer. The development of stable and disease-targeted delivery systems of CO is thus of interest and importance. Previously we developed a nano micellar CO donor SMA/CORM2 which shows superior bioavailability and therapeutic potential in many inflammatory disease models. We reported here, SMA/CORM2, through controlled release of CO, greatly ameliorated CsA-induced renal fibrosis via suppressing the NLRP3 inflammasome mediated TGF-β/Smad pathway. These findings suggest a new anti-inflammatory mechanism of CO, which also provides a new approach for controlling CsA-induced nephrotoxicity.
Collapse
|
23
|
Liu M, Huang Q, Zhu Y, Chen L, Li Y, Gong Z, Ai K. Harnessing reactive oxygen/nitrogen species and inflammation: Nanodrugs for liver injury. Mater Today Bio 2022; 13:100215. [PMID: 35198963 PMCID: PMC8850330 DOI: 10.1016/j.mtbio.2022.100215] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 12/11/2022] Open
Abstract
Overall, 12% of the global population (800 million) suffers from liver disease, which causes 2 million deaths every year. Liver injury involving characteristic reactive oxygen/nitrogen species (RONS) and inflammation plays a key role in progression of liver disease. As a key metabolic organ of the human body, the liver is susceptible to injury from various sources, including COVID-19 infection. Owing to unique structural features and functions of the liver, most current antioxidants and anti-inflammatory drugs are limited against liver injury. However, the characteristics of the liver could be utilized in the development of nanodrugs to achieve specific enrichment in the liver and consequently targeted treatment. Nanodrugs have shown significant potential in eliminating RONS and regulating inflammation, presenting an attractive therapeutic tool for liver disease through controlling liver injury. Therefore, the main aim of the current review is to provide a comprehensive summary of the latest developments contributing to our understanding of the mechanisms underlying nanodrugs in the treatment of liver injury via harnessing RONS and inflammation. Meanwhile, the prospects of nanodrugs for liver injury therapy are systematically discussed, which provides a sound platform for novel therapeutic insights and inspiration for design of nanodrugs to treat liver disease.
Collapse
Affiliation(s)
- Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yan Zhu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Li Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yumei Li
- Department of Assisted Reproduction, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
24
|
Wang C, Ma C, Gong L, Guo Y, Fu K, Zhang Y, Zhou H, Li Y. Macrophage Polarization and Its Role in Liver Disease. Front Immunol 2022; 12:803037. [PMID: 34970275 PMCID: PMC8712501 DOI: 10.3389/fimmu.2021.803037] [Citation(s) in RCA: 253] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Macrophages are important immune cells in innate immunity, and have remarkable heterogeneity and polarization. Under pathological conditions, in addition to the resident macrophages, other macrophages are also recruited to the diseased tissues, and polarize to various phenotypes (mainly M1 and M2) under the stimulation of various factors in the microenvironment, thus playing different roles and functions. Liver diseases are hepatic pathological changes caused by a variety of pathogenic factors (viruses, alcohol, drugs, etc.), including acute liver injury, viral hepatitis, alcoholic liver disease, metabolic-associated fatty liver disease, liver fibrosis, and hepatocellular carcinoma. Recent studies have shown that macrophage polarization plays an important role in the initiation and development of liver diseases. However, because both macrophage polarization and the pathogenesis of liver diseases are complex, the role and mechanism of macrophage polarization in liver diseases need to be further clarified. Therefore, the origin of hepatic macrophages, and the phenotypes and mechanisms of macrophage polarization are reviewed first in this paper. It is found that macrophage polarization involves several molecular mechanisms, mainly including TLR4/NF-κB, JAK/STATs, TGF-β/Smads, PPARγ, Notch, and miRNA signaling pathways. In addition, this paper also expounds the role and mechanism of macrophage polarization in various liver diseases, which aims to provide references for further research of macrophage polarization in liver diseases, contributing to the therapeutic strategy of ameliorating liver diseases by modulating macrophage polarization.
Collapse
Affiliation(s)
- Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuqin Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Honglin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
25
|
Expression Dynamics of Heme Oxygenase-1 in Tumor Cells and the Host Contributes to the Progression of Tumors. J Pers Med 2021; 11:jpm11121340. [PMID: 34945812 PMCID: PMC8704574 DOI: 10.3390/jpm11121340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 01/05/2023] Open
Abstract
Heme oxygenase (HO-1) plays an important role in cellular protection against various stresses. The induction of HO-1 is an effective strategy for reactive oxygen species-related diseases, inflammatory diseases, as well as suppressing carcinogenesis. On the other hand, the high expression of HO-1 is now well known in many tumors. In this study, we investigated the dynamics of HO-1 expression in the host and the tumor. In the mouse sarcoma S180 solid tumor model and the rat hepatoma AH136B ascitic tumor model, HO-1 expression in the tumor, as indicated by the end product of HO-1 activation, i.e., carbon monoxide, gradually increased along with tumor growth. Over-expression of HO-1 expression in mouse colon cancer C26 tumor cells significantly promoted tumor growth as well as lung metastasis, whereas opposite results were found when the HO-1 expression was reduced in the cells. On the other hand, upregulating HO-1 levels in the host by using an HO-1 inducer protected the progression of the xenograft tumor in mice, whereas lowering HO-1 expression in the host with an HO-1 inhibitor showed accelerated tumor growth and lung metastasis after subcutaneous tumor xenograft inoculation. These findings strongly suggest that the balance of HO-1 levels in the host and the tumor cells is essential for the occurrence, progression, and prognosis of cancer. Maintenance of appropriately high HO-1 levels in the host is favorable for cancer prevention, whereas suppression of HO-1 in the tumor cells may thus become a therapeutic strategy for cancer.
Collapse
|
26
|
Watabe Y, Taguchi K, Sakai H, Enoki Y, Maruyama T, Otagiri M, Kohno M, Matsumoto K. Bioinspired carbon monoxide delivery using artificial blood attenuates the progression of obliterative bronchiolitis via suppression of macrophage activation by IL-17A. Eur J Pharm Biopharm 2021; 170:43-51. [PMID: 34864198 DOI: 10.1016/j.ejpb.2021.11.011] [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: 07/19/2021] [Revised: 10/28/2021] [Accepted: 11/28/2021] [Indexed: 12/18/2022]
Abstract
Carbon monoxide (CO) is expected to attenuate the progression of obliterative bronchiolitis (OB), which is a serious complication after lung transplantation. However, issues in terms of feasible exogenous CO supply, such as continuousness and safety, remain unsolved. Here, we applied nano red blood cells, namely hemoglobin vesicles (Hb-V), as a CO cargo based on the biomimetic concept and investigated the therapeutic potential of CO-loaded Hb-V on OB in orthotopic tracheal transplant model mice. The CO-loaded Hb-V was comprised of negatively charged liposomes encapsulating carbonylhemoglobin with a size of ca. 220 nm. The results of histological evaluation showed that allograft luminal occlusion and fibrosis were significantly ameliorated by treatment with CO-loaded Hb-V compared to treatment with saline, cyclosporine, and Hb-V. The therapeutic effects of CO-loaded Hb-V on OB were due to the suppression of M1 macrophage activation in tracheal allografts, resulting from decreased IL-17A production. Furthermore, the expression of TNF-α and TGF-β in tracheal allografts was decreased by CO-loaded Hb-V treatment but not saline and Hb-V treatment, indicating that CO liberated from CO-loaded Hb-V inhibits epithelial-mesenchymal transition. These findings suggest that CO-loaded Hb-V exerts strong therapeutic efficacy against OB via the regulation of macrophage activation by IL-17A and TGF-β-driven epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Yuki Watabe
- Faculty of Pharmacy, Keio University, Tokyo, Japan
| | | | - Hiromi Sakai
- Department of Chemistry, Nara Medical University, Kashihara, Japan
| | - Yuki Enoki
- Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Toru Maruyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan; DDS Research Institute, Sojo University, Kumamoto, Japan
| | - Mitsutomo Kohno
- Department of General Thoracic Surgery, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | | |
Collapse
|
27
|
Guo C, Zhang C, Xia Z, Song B, Hu W, Cui Y, Xue Y, Xia M, Xu D, Zhang S, Fang J. Nano-designed CO donor ameliorates bleomycin-induced pulmonary fibrosis via macrophage manipulation. J Control Release 2021; 341:566-577. [PMID: 34864115 DOI: 10.1016/j.jconrel.2021.11.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/29/2021] [Indexed: 02/08/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible interstitial pulmonary disease due to chronic inflammatory responses. The prognosis of IPF is very poor, however, the therapeutic options are very limited. Previously we developed a polymeric micellar drug delivery system of carbon monoxide (CO) that is a pivotal anti-inflammatory gaseous molecule, i.e., SMA/CORM2, which exhibited therapeutic potentials against dextran sulfate sodium (DSS)-induced mouse colitis and acetaminophen (APAP) induced liver injury. Along this line, here we investigate the applicability of SMA/CORM2 on IPF using a bleomycin (BLM)-induced pulmonary fibrosis model. Severe inflammation and the consequent pulmonary fibrosis were triggered by BLM, whereas SMA/CORM2 treatment remarkably suppressed the inflammation progression and ameliorated the formation of fibrosis. CO is the effector molecule of SMA/CORM2, which exerted the therapeutic/protective effect mostly through suppressing the reprogramming of anti-inflammatory macrophages as revealed by the decreased expressions of CD206 and arginase-1 that were remarkably upregulated by BLM exposure. The suppression of macrophage polarization accompanied the downregulated hypoxia-inducible factor-1α (HIF-1α) and its target molecule heme oxygenase-1 (HO-1), suggesting a HIF-1α/HO-1 pathway for modulating macrophage reprogramming. As the downstream event of anti-inflammatory macrophage polarization, the alveolar epithelial to mesenchymal transition that is the major source of myofibroblast, the hallmark of IPF, was significantly suppressed by SMA/CORM2 via a TGF-β/Smad2/3 pathway. Compared to native CORM2 of equivalent dose, SMA/CROM2 exhibited a much better protective effect indicating its superior bioavailability as an enhanced permeability and retention (EPR) effect-based nanomedicine. We thus anticipate the application of SMA/CORM2 as a therapeutic candidate for IPF as well as other inflammatory diseases and disorders.
Collapse
Affiliation(s)
- Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Zhengmei Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Weirong Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Yingying Cui
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Yanni Xue
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230022, China; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, No. 81 Meishan Road, Hefei 230032, China
| | - Mizhen Xia
- School of Life Science, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China
| | - Dexiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China
| | - Shichen Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei 230022, China; MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, No. 81 Meishan Road, Hefei 230032, China; School of Public Health and Health Management, Anhui Medical College, No. 632 Furong Road, Hefei 230601, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, School of Public Health, Anhui Medical University, Hefei 230022, China; Faculty of Pharmaceutical Science, Sojo University, Ikeda 4-22-1, Kumamoto 860-0082, Japan.
| |
Collapse
|
28
|
Yan J, Ruan P, Ge Y, Gao J, Tan H, Xiao C, Gao Q, Zhang Z, Gao Y. Mechanisms and Molecular Targets of Compound Danshen Dropping Pill for Heart Disease Caused by High Altitude Based on Network Pharmacology and Molecular Docking. ACS OMEGA 2021; 6:26942-26951. [PMID: 34693115 PMCID: PMC8529605 DOI: 10.1021/acsomega.1c03282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/21/2021] [Indexed: 05/30/2023]
Abstract
Compound Danshen dropping pill (CDDP), a famous Chinese medicine formula, has been widely used to treat high-altitude heart disease in China. However, its molecular mechanisms, potential targets, and bioactive ingredients remain elusive. In this study, network pharmacology, molecular docking, and validation experiments were combined to investigate the effective active ingredients and molecular mechanisms of CDDP in the treatment of high-altitude heart disease. Tan IIA may be the main active component of CDDP in the treatment of high-altitude heart disease via HIF-1/PI3K/Akt pathways.
Collapse
Affiliation(s)
- Jiayi Yan
- School
of Traditional Chinese Medicine, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
| | - Panpan Ruan
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
- School
of Life Science, Heibei University, Baoding 071000, China
| | - Yunxuan Ge
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
- College
of Life Science and Bioengineering, Beijing
University of Technology, Beijing 100124, China
| | - Jing Gao
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
- School
of Pharmacy, Henan University, Kaifeng 475004, China
| | - Hongling Tan
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
| | - Chengrong Xiao
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
| | - Quansheng Gao
- Institute
of Enviromental and Operational Medicine, Academy of Military Medical
Sciences, Academy of Military Sciences, Tianjin 300381, China
| | - Zhuo Zhang
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
| | - Yue Gao
- School
of Traditional Chinese Medicine, Guangdong
Pharmaceutical University, Guangzhou 510006, China
- Department
of Pharmaceutical Sciences, Beijing Institute
of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
29
|
Shaban MA, El-Husseiny AA, Shaban HM, Hamdi SAH, Elshafey MM. Hepatoprotective Effect of Alcoholic and N-hexane Extracts of Crayfish Procambarus Clarkii against CCl4-induced Damage in HepG2 Cells. JOURNAL OF AQUATIC FOOD PRODUCT TECHNOLOGY 2021. [DOI: 10.1080/10498850.2021.1988791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Mostafa Ahmed Shaban
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ahmed A. El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Hesham Mohamed Shaban
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | | | - Mostafa Mahmoud Elshafey
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
30
|
Zhai T, Zhong W, Gao Y, Zhou H, Zhou Z, Liu X, Yang S, Yang H. Tumor Microenvironment-Activated Nanoparticles Loaded with an Iron-Carbonyl Complex for Chemodynamic Immunotherapy of Lung Metastasis of Melanoma In Vivo. ACS APPLIED MATERIALS & INTERFACES 2021; 13:39100-39111. [PMID: 34382406 DOI: 10.1021/acsami.1c11485] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this work, a nanoplatform (FeCORM NPs) loaded with an iron-carbonyl complex was constructed. By exploiting chemodynamic therapy (CDT) and immunogenic cell death (ICD)-induced immunotherapy (IMT), the nanoparticles exhibited excellent efficacy against lung metastasis of melanoma in vivo. The iron-carbonyl compound of the nanomaterials could be initiated by both glutathione (GSH) and hydrogen peroxide (H2O2) to release CO and generate ferrous iron through ligand exchange and oxidative destruction pathways. The released CO caused mitochondria damage, whereas the generated ferrous iron led to oxidative stress via the Fenton reaction. On the other hand, the nanomaterials induced ICD-based IMT, which worked jointly with CDT to exhibit excellent effects against lung metastasis of melanoma through a mouse model. This work demonstrated how a nanoplatform, simple and stable but showing excellent efficacy against tumors, could be built using simple building blocks via a self-assembling approach. Importantly, the system took advantage of relatively high levels of GSH and H2O2 in tumors to initiate the therapeutic effects, which rendered the nanoplatform with a capability to differentiate normal cells from tumor cells. In principle, the system has great potential for future clinical applications, not only in the treatment of lung metastasis of melanoma but also in suppressing other types of tumors.
Collapse
Affiliation(s)
- Tianli Zhai
- College of Chemistry and Materials Science, International Joint Laboratory on Resource Chemistry of Ministry Education, Shanghai Normal University, Shanghai 200234, P. R. China
| | - Wei Zhong
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing 314001, P. R. China
| | - Yucong Gao
- College of Chemistry and Materials Science, International Joint Laboratory on Resource Chemistry of Ministry Education, Shanghai Normal University, Shanghai 200234, P. R. China
| | - Han Zhou
- College of Chemistry and Materials Science, International Joint Laboratory on Resource Chemistry of Ministry Education, Shanghai Normal University, Shanghai 200234, P. R. China
| | - Zhiguo Zhou
- College of Chemistry and Materials Science, International Joint Laboratory on Resource Chemistry of Ministry Education, Shanghai Normal University, Shanghai 200234, P. R. China
| | - Xiaoming Liu
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing 314001, P. R. China
| | - Shiping Yang
- College of Chemistry and Materials Science, International Joint Laboratory on Resource Chemistry of Ministry Education, Shanghai Normal University, Shanghai 200234, P. R. China
| | - Hong Yang
- College of Chemistry and Materials Science, International Joint Laboratory on Resource Chemistry of Ministry Education, Shanghai Normal University, Shanghai 200234, P. R. China
| |
Collapse
|
31
|
Carbon Monoxide Therapy Using Hybrid Carbon Monoxide-Releasing/Nrf2-Inducing Molecules through a Neuroprotective Lens. CHEMISTRY 2021. [DOI: 10.3390/chemistry3030057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Carbon monoxide (CO) has long been known for its toxicity. However, in recent decades, new applications for CO as a therapeutic compound have been proposed, and multiple forms of CO therapy have since been developed and studied. Previous research has found that CO has a role as a gasotransmitter and promotes anti-inflammatory and antioxidant effects, making it an avenue of interest for medicine. Such effects are possible because of the Nrf2/HO1 pathway, which has become a target for therapy development because its activation also leads to CO release. Currently, different forms of treatment involving CO include inhaled CO (iCO), carbon monoxide-releasing molecules (CORMs), and hybrid carbon monoxide-releasing molecules (HYCOs). In this article, we review the progression of CO studies to develop possible therapies, the possible mechanisms involved in the effects of CO, and the current forms of therapy using CO.
Collapse
|
32
|
LR12 Promotes Liver Repair by Improving the Resolution of Inflammation and Liver Regeneration in Mice with Thioacetamide- (TAA-) Induced Acute Liver Failure. Mediators Inflamm 2021; 2021:2327721. [PMID: 34135689 PMCID: PMC8179768 DOI: 10.1155/2021/2327721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022] Open
Abstract
Background Triggering receptor expressed on myeloid cells-1 (TREM-1) controls the mobilization of inflammatory cells in response to injury and consequently enhances liver damage. LR12 is a TREM-1 inhibitory peptide. However, the role of LR12 in acute liver failure (ALF) has remained elusive. This study was aimed at indicating whether LR12 could promote liver repair in mice with thioacetamide- (TAA-) induced ALF. Methods BALB/c mice were intraperitoneally injected with TAA, followed by intravenous injection of LR12. Damage and regeneration of the liver were assessed. LO2 cells and macrophages were used to assess the therapeutic effects of LR12. Results Mice treated with TAA for 24 h developed ALF, while liver inflammation was alleviated after LR12 treatment. Moreover, LR12 promoted hepatocyte regeneration in mice with TAA-induced ALF. In vitro, the supernatant from TAA+LR12-treated macrophages promoted the proliferation of LO2 cells. Cytokine protein microarray analysis suggested that LR12 promoted the secretion of C-C chemokine ligand 20 (CCL20) from macrophages. Besides, neutralization of CCL20 blocked the effects of LR12, thus inhibited the proliferation of LO2 cells in vitro, aggregated the liver inflammation, and restrained hepatocyte regeneration in ALF mice in vivo. Furthermore, we also found that LR12 activated the p38 mitogen-activated protein kinase (MAPK) pathway in hepatocytes through promoting the secretion of CCL20 from macrophages. Conclusions LR12 could improve the resolution of inflammation and liver regeneration in mice with TAA-induced ALF by promoting the secretion of CCL20 from macrophages and activating the p38 MAPK pathway. Therefore, LR12 could be an attractive therapeutic target for the treatment of ALF.
Collapse
|