1
|
Liu Z, Wei J, Sun H, Xu L. Plumbagin ameliorates LPS-induced acute lung injury by regulating PI3K/AKT/mTOR and Keap1-Nrf2/HO-1 signalling pathways. J Cell Mol Med 2024; 28:e18386. [PMID: 38990057 PMCID: PMC11238321 DOI: 10.1111/jcmm.18386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 07/12/2024] Open
Abstract
Acute lung injury (ALI) is a major pathophysiological problem characterized by severe inflammation, resulting in high morbidity and mortality. Plumbagin (PL), a major bioactive constituent extracted from the traditional Chinese herb Plumbago zeylanica, has been shown to possess anti-inflammatory and antioxidant pharmacological activities. However, its protective effect on ALI has not been extensively studied. The objective of this study was to investigate the protective effect of PL against ALI induced by LPS and to elucidate its possible mechanisms both in vivo and in vitro. PL treatment significantly inhibited pathological injury, MPO activity, and the wet/dry ratio in lung tissues, and decreased the levels of inflammatory cells and inflammatory cytokines TNF-α, IL-1β, IL-6 in BALF induced by LPS. In addition, PL inhibited the activation of the PI3K/AKT/mTOR signalling pathway, increased the activity of antioxidant enzymes CAT, SOD, GSH and activated the Keap1/Nrf2/HO-1 signalling pathway during ALI induced by LPS. To further assess the association between the inhibitory effects of PL on ALI and the PI3K/AKT/mTOR and Keap1/Nrf2/HO-1 signalling, we pretreated RAW264.7 cells with 740Y-P and ML385. The results showed that the activation of PI3K/AKT/mTOR signalling reversed the protective effect of PL on inflammatory response induced by LPS. Moreover, the inhibitory effects of PL on the production of inflammatory cytokines induced by LPS also inhibited by downregulating Keap1/Nrf2/HO-1 signalling. In conclusion, the results indicate that the PL ameliorate LPS-induced ALI by regulating the PI3K/AKT/mTOR and Keap1-Nrf2/HO-1 signalling, which may provide a novel therapeutic perspective for PL in inhibiting ALI.
Collapse
Affiliation(s)
- Zhengjia Liu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Jiahui Wei
- Department of RespiratoryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Hongbin Sun
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Lei Xu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
2
|
Li Y, Li J, Dong Y, Wang C, Cai Z. Bovine lactoferrin inhibits inflammatory response and apoptosis in lipopolysaccharide-induced acute lung injury by targeting the PPAR-γ pathway. Mol Biol Rep 2024; 51:492. [PMID: 38578368 DOI: 10.1007/s11033-024-09436-2] [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: 12/05/2023] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Lactoferrin (LF) is an iron-binding multifunctional cationic glycoprotein. Previous studies have demonstrated that LF may be a potential drug for treating acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). In this study, we explored the anti-inflammatory effect and mechanism of bovine lactoferrin (bLF) in ALI using the RNA sequencing (RNA-seq) technology and transcriptome analysis. METHODS AND RESULTS Based on the differentially expressed genes (DEGs) obtained from RNA-seq of the Lung from mouse model, the bioinformatics workflow was implemented using the BGISEQ-500 platform. The protein-protein interaction (PPI) network was obtained using STRING, and the hub gene was screened using Cytoscape. To verify the results of transcriptome analysis, the effects of bLF on Lipopolysaccharide (LPS)-induced BEAS-2B cells and its anti-reactive oxygen species (ROS), anti-inflammatory, and antiapoptotic effects were studied via Cell Counting Kit-8 (CCK-8) test, active oxygen detection test, ELISA, and western blot assay. Transcriptome analysis revealed that two hub gene modules of DEGs were screened via PPI analysis using the STRING and MCODE plug-ins of Cytoscape. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that these core modules are enriched in the PPAR (peroxisome proliferator-activated receptor) and AMPK (AMP-activated protein kinase) signaling pathways. Through cell experiments, our study shows that bLF can inhibit ROS, inflammatory reaction, and LPS-induced BEAS-2B cell apoptosis, which are significantly antagonized by the PPAR-γ inhibitor GW9662. CONCLUSION This study has suggested that the PPAR-γ pathway is the critical target of bLF in anti-inflammatory reactions and apoptosis of ALI, which provides a direction for further research.
Collapse
Affiliation(s)
- Yantao Li
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, 050000, China
| | - Junhu Li
- Emergency Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, 050000, China
| | - Yan Dong
- Emergency Department, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, 050000, China
| | - Can Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, 050000, China
| | - Zhigang Cai
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
- Hebei Key Laboratory of Respiratory Critical Care Medicine, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
- Hebei Institute of Respiratory Diseases, No. 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
3
|
Han L, Li Q, Du M, Mao X. Bovine milk osteopontin improved intestinal health of pregnant rats fed a high-fat diet through improving bile acid metabolism. J Dairy Sci 2024; 107:24-39. [PMID: 37690710 DOI: 10.3168/jds.2023-23802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/31/2023] [Indexed: 09/12/2023]
Abstract
The main purpose of the current study was to investigate the ameliorative effects of bovine milk osteopontin (bmOPN) on the gut dysfunction of pregnant rats fed a high-fat diet (HFD). Bovine milk osteopontin was supplemented at a dose of 6 mg/kg body weight. Bovine milk osteopontin supplementation during pregnancy reduced colonic inflammation of HFD dams, and it also increased the colonic expression of ZO-1 and claudin-4 of HFD dams. Bovine milk osteopontin significantly enriched the relative abundance of Bacteroidetes, whereas it decreased Proteobacteria, Helicobacteraceae, and Desulfovibrionaceae in feces of HFD dams. The levels of isobutyric acid and pentanoic acid in the HFD + bmOPN group were higher than that of the HFD group. Functional predication analysis of microbial genomes revealed that bmOPN supplementation to HFD pregnancies changed 4 Kyoto Encyclopedia of Genes and Genomes pathways including bile acid biosynthesis. Further, bmOPN enriched hepatic taurochenodeoxycholic acid and tauroursodeoxycholic acid plus taurohyodeoxycholic acid in the gut of HFD maternal rats. Our findings suggested that bmOPN improved the gut health of HFD pregnant rats partially through modulating bile acid biosynthesis.
Collapse
Affiliation(s)
- Lihua Han
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Qiqi Li
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99163
| | - Xueying Mao
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
4
|
Cho S, Ying F, Sweeney G. Sterile inflammation and the NLRP3 inflammasome in cardiometabolic disease. Biomed J 2023; 46:100624. [PMID: 37336361 PMCID: PMC10539878 DOI: 10.1016/j.bj.2023.100624] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023] Open
Abstract
Inflammation plays an important role in the pathophysiology of cardiometabolic diseases. Sterile inflammation, a non-infectious and damage-associated molecular pattern (DAMP)-induced innate response, is now well-established to be closely associated with development and progression of cardiometabolic diseases. The NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is well-established as a major player in sterile inflammatory responses. It is a multimeric cytosolic protein complex which regulates the activation of caspase-1 and subsequently promotes cleavage and release of interleukin (IL)-1 family cytokines, which have a deleterious impact on the development of cardiometabolic diseases. Therefore, targeting NLRP3 itself or the downstream consequences of NLRP3 activation represent excellent potential therapeutic targets in inflammatory cardiometabolic diseases. Here, we review our current understanding of the role which NLRP3 inflammasome regulation plays in cardiometabolic diseases such as obesity, diabetes, non-alcoholic steatohepatitis (NASH), atherosclerosis, ischemic heart disease and cardiomyopathy. Finally, we highlight the potential of targeting NLPR3 or related signaling molecules as a therapeutic approach.
Collapse
Affiliation(s)
- Sungji Cho
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Fan Ying
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Ianiro G, Rosa L, Bonaccorsi di Patti MC, Valenti P, Musci G, Cutone A. Lactoferrin: from the structure to the functional orchestration of iron homeostasis. Biometals 2023; 36:391-416. [PMID: 36214975 DOI: 10.1007/s10534-022-00453-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/25/2022] [Indexed: 11/02/2022]
Abstract
Iron is by far the most widespread and essential transition metal, possessing crucial biological functions for living systems. Despite chemical advantages, iron biology has forced organisms to face with some issues: ferric iron insolubility and ferrous-driven formation of toxic radicals. For these reasons, acquisition and transport of iron constitutes a formidable challenge for cells and organisms, which need to maintain adequate iron concentrations within a narrow range, allowing biological processes without triggering toxic effects. Higher organisms have evolved extracellular carrier proteins to acquire, transport and manage iron. In recent years, a renewed interest in iron biology has highlighted the role of iron-proteins dysregulation in the onset and/or exacerbation of different pathological conditions. However, to date, no resolutive therapy for iron disorders has been found. In this review, we outline the efficacy of Lactoferrin, a member of the transferrin family mainly secreted by exocrine glands and neutrophils, as a new emerging orchestrator of iron metabolism and homeostasis, able to counteract iron disorders associated to different pathologies, including iron deficiency and anemia of inflammation in blood, Parkinson and Alzheimer diseases in the brain and cystic fibrosis in the lung.
Collapse
Affiliation(s)
- Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | | | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Rome, Italy
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, Pesche, Italy.
| |
Collapse
|
6
|
Kaczyńska K, Jampolska M, Wojciechowski P, Sulejczak D, Andrzejewski K, Zając D. Potential of Lactoferrin in the Treatment of Lung Diseases. Pharmaceuticals (Basel) 2023; 16:192. [PMID: 37259341 PMCID: PMC9960651 DOI: 10.3390/ph16020192] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 11/07/2023] Open
Abstract
Lactoferrin (LF) is a multifunctional iron-binding glycoprotein that exhibits a variety of properties, such as immunomodulatory, anti-inflammatory, antimicrobial, and anticancer, that can be used to treat numerous diseases. Lung diseases continue to be the leading cause of death and disability worldwide. Many of the therapies currently used to treat these diseases have limited efficacy or are associated with side effects. Therefore, there is a constant pursuit for new drugs and therapies, and LF is frequently considered a therapeutic agent and/or adjunct to drug-based therapies for the treatment of lung diseases. This article focuses on a review of the existing and most up-to-date literature on the contribution of the beneficial effects of LF on the treatment of lung diseases, including asthma, viral infections, cystic fibrosis, or lung cancer, among others. Although in vitro and in vivo studies indicate significant potency of LF in the treatment of the listed diseases, only in the case of respiratory tract infections do human studies seem to confirm them by demonstrating the effectiveness of LF in reducing episodes of illness and shortening the recovery period. For lung cancer, COVID-19 and sepsis, the reports are conflicting, and for other diseases, there is a paucity of human studies conclusively confirming the beneficial effects of LF.
Collapse
Affiliation(s)
- Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Monika Jampolska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Piotr Wojciechowski
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Kryspin Andrzejewski
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Dominika Zając
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| |
Collapse
|
7
|
Wang F, Ma J, Wang J, Chen M, Xia H, Yao S, Zhang D. SIRT1 ameliorated septic associated-lung injury and macrophages apoptosis via inhibiting endoplasmic reticulum stress. Cell Signal 2022; 97:110398. [PMID: 35811055 DOI: 10.1016/j.cellsig.2022.110398] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND The inappropriate apoptosis of macrophages plays an important role in the pathogenesis of sepsis-induced acute lung injury, however, the detailed regulatory mechanisms remain largely unknown. As an endogenous apoptosis pathway, endoplasmic reticulum (ER) stress plays an important role in cell damage in patients with sepsis. Clarifying the ER stress response and its effect on macrophages during the development of sepsis is helpful to explore new strategies for the prevention and treatment of ALI in sepsis. METHODS The mouse model and the RAW264.7 inflammation model were stimulated with LPS to establish in vivo and in vitro. We explored the effects of different expression levels of silent information regulator factor 2-related enzyme 1 (SIRT1) on the ER stress response and apoptosis of macrophages in the sepsis-related injury model. RESULTS Our studies found that the increased expression of SIRT1 can significantly improve sepsis-related lung injury and relieve lung inflammation. SRT1720, a SIRT1 activator, can significantly inhibit the ER stress response of lung tissue and macrophages, inhibit the expression of pro-apoptotic proteins, promote the expression of anti-apoptotic proteins, and reduce macrophages of apoptosis. While the EX527, an inhibitor of SIRT1, had the opposite effect. CONCLUSION SIRT1 can significantly improve sepsis-associated lung injury and LPS-induced macrophage apoptosis. This protective effect is closely related to its inhibition of the ER stress response via the PERK/eIF2-α/ATF4/CHOP pathway.
Collapse
Affiliation(s)
- Fuquan Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiamin Ma
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jingxu Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ming Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haifa Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Dingyu Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Wuhan Jinyintan Hospital, Wuhan 430023, China.
| |
Collapse
|
8
|
Bao L, Dou G, Tian R, Lv Y, Ding F, Liu S, Zhao R, Zhao L, Zhou J, Weng L, Dong Y, Li B, Liu S, Chen X, Jin Y. Engineered neutrophil apoptotic bodies ameliorate myocardial infarction by promoting macrophage efferocytosis and inflammation resolution. Bioact Mater 2021; 9:183-197. [PMID: 34820565 PMCID: PMC8586716 DOI: 10.1016/j.bioactmat.2021.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 11/25/2022] Open
Abstract
Inflammatory response plays a critical role in myocardial infarction (MI) repair. The neutrophil apoptosis and subsequent macrophage ingestion can result in inflammation resolution and initiate regeneration, while the therapeutic strategy that simulates and enhances this natural process has not been established. Here, we constructed engineered neutrophil apoptotic bodies (eNABs) to simulate natural neutrophil apoptosis, which regulated inflammation response and enhanced MI repair. The eNABs were fabricated by combining natural neutrophil apoptotic body membrane which has excellent inflammation-tropism and immunoregulatory properties, and mesoporous silica nanoparticles loaded with hexyl 5-aminolevulinate hydrochloride (HAL). The eNABs actively targeted to macrophages and the encapsulated HAL simultaneously initiated the biosynthesis pathway of heme to produce anti-inflammatory bilirubin after intracellular release, thereby further enhancing the anti-inflammation effects. In in vivo studies, the eNABs efficiently modulated inflammation responses in the infarcted region to ameliorate cardiac function. This study demonstrates an effective biomimetic construction strategy to regulate macrophage functions for MI repair. Construction of engineered neutrophil apoptotic bodies to simulate natural neutrophil apoptosis. Engineered neutrophil apoptotic bodies with excellent inflammation-tropism and macrophage-specific targeting capacity. Engineered neutrophil apoptotic bodies enhance macrophage efferocytosis and reprogramming for inflammation resolution. Engineered neutrophil apoptotic bodies ameliorate myocardial infarction and promote cardiac tissue regeneration after MI.
Collapse
Affiliation(s)
- Lili Bao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Geng Dou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ran Tian
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, 710049, China
| | - Yajie Lv
- Department of Dermatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Feng Ding
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Siying Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ruifeng Zhao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Digital Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lu Zhao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jun Zhou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Lin Weng
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, 710049, China
| | - Yan Dong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Bei Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
- Corresponding author.
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, 710049, China
- Corresponding author.
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
- Corresponding author.
| |
Collapse
|
9
|
Barman TK, Metzger DW. Disease Tolerance during Viral-Bacterial Co-Infections. Viruses 2021; 13:v13122362. [PMID: 34960631 PMCID: PMC8706933 DOI: 10.3390/v13122362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/29/2021] [Accepted: 11/23/2021] [Indexed: 11/16/2022] Open
Abstract
Disease tolerance has emerged as an alternative way, in addition to host resistance, to survive viral-bacterial co-infections. Disease tolerance plays an important role not in reducing pathogen burden, but in maintaining tissue integrity and controlling organ damage. A common co-infection is the synergy observed between influenza virus and Streptococcus pneumoniae that results in superinfection and lethality. Several host cytokines and cells have shown promise in promoting tissue protection and damage control while others induce severe immunopathology leading to high levels of morbidity and mortality. The focus of this review is to describe the host cytokines and innate immune cells that mediate disease tolerance and lead to a return to host homeostasis and ultimately, survival during viral-bacterial co-infection.
Collapse
|
10
|
Zimecki M, Actor JK, Kruzel ML. The potential for Lactoferrin to reduce SARS-CoV-2 induced cytokine storm. Int Immunopharmacol 2021; 95:107571. [PMID: 33765614 PMCID: PMC7953442 DOI: 10.1016/j.intimp.2021.107571] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
The COVID-19 pandemic is a serious global health threat caused by severe acute respiratory syndrome of coronavirus 2 (SARS-CoV-2). Symptoms of COVID-19 are highly variable with common hyperactivity of immune responses known as a "cytokine storm". In fact, this massive release of inflammatory cytokines into in the pulmonary alveolar structure is a main cause of mortality during COVID-19 infection. Current management of COVID-19 is supportive and there is no common clinical protocol applied to suppress this pathological state. Lactoferrin (LF), an iron binding protein, is a first line defense protein that is present in neutrophils and excretory fluids of all mammals, and is well recognized for its role in maturation and regulation of immune system function. Also, due to its ability to sequester free iron, LF is known to protect against insult-induced oxidative stress and subsequent "cytokine storm" that results in dramatic necrosis within the affected tissue. Review of the literature strongly suggests utility of LF to silence the "cytokine storm", giving credence to both prophylactic and therapeutic approaches towards combating COVID-19 infection.
Collapse
Affiliation(s)
- Michał Zimecki
- The Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Jeffrey K Actor
- University of Texas, Health Science Center Houston, Texas, USA.
| | - Marian L Kruzel
- University of Texas, Health Science Center Houston, Texas, USA
| |
Collapse
|
11
|
Lee J, Lee J, Lee S, Ahmad T, Madhurakkat Perikamana SK, Kim EM, Lee SW, Shin H. Bioactive Membrane Immobilized with Lactoferrin for Modulation of Bone Regeneration and Inflammation. Tissue Eng Part A 2020; 26:1243-1258. [DOI: 10.1089/ten.tea.2020.0015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Jinki Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Taufiq Ahmad
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Sajeesh Kumar Madhurakkat Perikamana
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Eun Mi Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Sang Won Lee
- School of Biomedical Engineering, Korea University, Seoul, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
12
|
MHTP, a synthetic tetratetrahydroisoquinoline alkaloid, attenuates lipopolysaccharide-induced acute lung injury via p38MAPK/p65NF-κB signaling pathway-TLR4 dependent. Inflamm Res 2019; 68:1061-1070. [DOI: 10.1007/s00011-019-01291-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/24/2019] [Accepted: 10/02/2019] [Indexed: 01/07/2023] Open
|
13
|
Li W, Wu F, Chen L, Li Q, Ma J, Li M, Shi Y. Carbon Monoxide Attenuates Lipopolysaccharides (LPS)-Induced Acute Lung Injury in Neonatal Rats via Downregulation of Cx43 to Reduce Necroptosis. Med Sci Monit 2019; 25:6255-6263. [PMID: 31429423 PMCID: PMC6713028 DOI: 10.12659/msm.917751] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Acute lung injury (ALI) is one of major causes of death in newborns, making it urgent to improve therapy. Administration of low dose carbon monoxide (CO) plays a protective role in ALI but the mechanisms are not fully understood. This study was designed to test the therapeutic effect of monoxide-releasing molecule 3 (MORM3) in lipopolysaccharide (LPS) induced neonatal ALI and the possibly associated molecular mechanisms. Material/Methods For this study, 3- to 8-day old Newborn Sprague-Dawley rats were subjected to intraperitoneal injection of 3 mg/kg LPS to induce ALI. Then animals received intraperitoneal injection of carbon monoxide-releasing molecules 3 (CORM3) (8 mg/kg) or inactive CORM3 (iCORM3) for 7 consecutive days. Lung tissues were collected for histological examination and total cell counts and protein content in bronchoalveolar lavage fluid (BALF) were measured. Expression of Cx43 and necroptosis-related markers were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Results LPS exposure induced significant lung injury indicated by histological damage, increased lung wet/dry weight ratio (W/D) and increased total cell counts and protein concentration in BALF. These changes were significantly ameliorated by administration of CORM3 but not iCORM3. LPS also increased necroptosis-related markers RIP1, RIP3, and MLKL and their elevation was blocked by CORM3. CORM3 administration ameliorated LPS induced elevation of Cx43 expression and adenoviral overexpression of Cx43 abolished lung protective effect of CORM3. CORM3 administration attenuated LPS induced activation of extracellular-signal-regulated kinase (ERK) and its protection against necroptosis was abolished by ERK inhibitor U0126. Conclusions CORM3 attenuates LPS-Induced ALI in neonatal rats and its lung protective effect might be through downregulation of Cx43 to attenuate ERK signaling and ameliorate necroptosis, suggesting CORM3 as a potential therapeutic drug for ALI in neonates.
Collapse
Affiliation(s)
- Wanwei Li
- Department of Pediatrics, Daping Hospital, Army Medical University, Chongqing, China (mainland)
| | - Fang Wu
- Department of Neonatology, Chongqing Angel Women's and Children's Hospital, Chongqing, China (mainland)
| | - Long Chen
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China (mainland)
| | - Qian Li
- Department of Pediatrics, Daping Hospital, Army Medical University, Chongqing, China (mainland)
| | - Juan Ma
- Department of Pediatrics, Daping Hospital, Army Medical University, Chongqing, China (mainland)
| | - Mengchun Li
- Department of Pediatrics, Daping Hospital, Army Medical University, Chongqing, China (mainland)
| | - Yuan Shi
- Department of Pediatrics, Daping Hospital, Army Medical University, Chongqing, China (mainland).,Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China (mainland)
| |
Collapse
|
14
|
Yen CC, Chang WH, Tung MC, Chen HL, Liu HC, Liao CH, Lan YW, Chong KY, Yang SH, Chen CM. Lactoferrin Protects Hyperoxia-Induced Lung and Kidney Systemic Inflammation in an In Vivo Imaging Model of NF-κB/Luciferase Transgenic Mice. Mol Imaging Biol 2019; 22:526-538. [DOI: 10.1007/s11307-019-01390-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
Heikal MM, Shaaban AA, Elkashef WF, Ibrahim TM. Effect of febuxostat on biochemical parameters of hyperlipidemia induced by a high-fat diet in rabbits. Can J Physiol Pharmacol 2019; 97:611-622. [DOI: 10.1139/cjpp-2018-0731] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Febuxostat, a highly potent xanthine oxidase inhibitor with an antioxidant effect, inhibits elevated xanthine oxidase, leading to reduction of reactive oxygen species and oxidative stress, the main causes of vascular inflammation in hyperlipidemia. The aim of this study was to test the potential antioxidant and anti-inflammatory effects of febuxostat and (or) stopping a high-fat diet on the biochemical parameters in rabbits with hyperlipidemia induced by a high-fat diet. Male New Zealand rabbits were distributed into 3 groups: a normal control group fed standard chow for 12 weeks and 2 other groups fed a high-fat diet with 1% cholesterol for 8 weeks, and then shifted to standard chow for 4 weeks. During the last 4 weeks, one high-fat diet group received 0.5% carboxymethyl cellulose, whereas the other group was treated with febuxostat (2 mg/kg per day p.o.). Febuxostat significantly lowered low-density lipoprotein cholesterol (“bad” cholesterol) compared to the untreated group (high-fat diet group). Febuxostat also displayed a potent anti-inflammatory and antioxidant activity by decreasing serum levels of lipid peroxidation index, proinflammatory cytokines, and enhancing antioxidant enzyme activity. Stopping the hyperlipidemic diet in the high-fat diet group did not show improvement. These findings indicate the antioxidant and anti-inflammatory effects of febuxostat that may be common mechanisms of the anti-hyperlipidemic effect of this drug. Stopping a hyperlipidemic diet without treatment is not sufficient once injury has occurred.
Collapse
Affiliation(s)
- Mohammed M. Heikal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed A. Shaaban
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Faculty of Pharmacy, Aqaba University of Technology, Jordan
| | - Wagdi F. Elkashef
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Tarek M. Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
Wei YL, Xu JY, Zhang R, Zhang Z, Zhao L, Qin LQ. Effects of lactoferrin on X-ray-induced intestinal injury in Balb/C mice. Appl Radiat Isot 2019; 146:72-77. [DOI: 10.1016/j.apradiso.2019.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 01/03/2019] [Accepted: 01/24/2019] [Indexed: 01/09/2023]
|
17
|
Abstract
Neutrophilic bronchiolitis is the primary lesion in asthma-affected horses. Neutrophils are key actors in host defense, migrating toward sites of inflammation and infection, where they act as early responder cells toward external insults. However, neutrophils can also mediate tissue damage in various non-infectious inflammatory processes. Within the airways, these cells likely contribute to bronchoconstriction, mucus hypersecretion, and pulmonary remodeling by releasing pro-inflammatory mediators, including the cytokines interleukin (IL)-8 and IL-17, neutrophil elastase, reactive oxygen species (ROS), and neutrophil extracellular traps (NETs). The mechanisms that regulate neutrophil functions in the tissues are complex and incompletely understood. Therefore, the inflammatory activity of neutrophils must be regulated with exquisite precision and timing, a task achieved through a complex network of mechanisms that regulates neutrophil survival. The discovery and development of compounds that can help regulate ROS, NET formation, cytokine release, and clearance would be highly beneficial in the design of therapies for this disease in horses. In this review, neutrophil functions during inflammation will be discussed followed by a discussion of their contribution to airway tissue injury in equine asthma.
Collapse
|
18
|
Zeng S, Qiao H, Lv XW, Fan D, Liu T, Xie D. High-dose dexamethasone induced LPS-stimulated rat alveolar macrophages apoptosis. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3097-3104. [PMID: 29123381 PMCID: PMC5661847 DOI: 10.2147/dddt.s147014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prolonged administration of an excessive dose of corticosteroids proved to be harmful for patients with acute lung injury (ALI). A previous study has found that repeated administration of an excessive dose of methylprednisolone reduced alveolar macrophages (AMs) in bronchoalveolar lavage fluid (BALF) with an unknown mechanism. This study aimed to investigate the effect of excessive use of dexamethasone (Dex) on BALF AMs in vitro. Transmission electron microscopy and DNA fragmentation analysis demonstrated that 10-4 and 10-5 M Dex induced lipopolysaccharide-stimulated rat AMs apoptosis with downregulation of tumor necrosis factor-α, interleukin (IL)-12 and upregulation of IL-10, transforming growth factor-β. These results indicated that apoptosis might be a novel contribution involved in the detrimental effect of excessive dose of Dex clinically used to treat ALI.
Collapse
Affiliation(s)
- Si Zeng
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu
| | - Hui Qiao
- Department of Anesthesiology, The Eye and ENT Hospital of Fudan University, Shanghai, China
| | - Xue-Wen Lv
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu
| | - Dan Fan
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu
| | - Tong Liu
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu
| | - Dongli Xie
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu
| |
Collapse
|
19
|
Brimelow RE, West NP, Williams LT, Cripps AW, Cox AJ. A role for whey-derived lactoferrin and immunoglobulins in the attenuation of obesity-related inflammation and disease. Crit Rev Food Sci Nutr 2017; 57:1593-1602. [PMID: 26068582 DOI: 10.1080/10408398.2014.995264] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Obesity is a strong predictive factor in the development of chronic disease and has now superseded undernutrition as a major public health issue. Chronic inflammation is one mechanism thought to link excess body weight with disease. Increasingly, the gut and its extensive population of commensal microflora are recognized as playing an important role in the development of obesity-related chronic inflammation. Obesity and a high fat diet are associated with altered commensal microbial communities and increased intestinal permeability which contributes to systemic inflammation as a result of the translocation of lipopolysaccharide into the circulation and metabolic endotoxemia. Various milk proteins are showing promise in the prevention and treatment of obesity and chronic low-grade inflammation via reductions in visceral fat, neutralization of bacteria at the mucosa and reduced intestinal permeability. In this review, we focus on evidence supporting the potential antiobesogenic and anti-inflammatory effects of bovine whey-derived lactoferrin and immunoglobulins.
Collapse
Affiliation(s)
- Rachel E Brimelow
- a School of Medical Science, Griffith University , Southport , Queensland , Australia
| | - Nicholas P West
- a School of Medical Science, Griffith University , Southport , Queensland , Australia.,b Menzies Health Institute Queensland , Southport , Queensland , Australia
| | - Lauren T Williams
- b Menzies Health Institute Queensland , Southport , Queensland , Australia.,c School of Allied Health Sciences, Griffith University , Southport , Queensland Australia
| | - Allan W Cripps
- b Menzies Health Institute Queensland , Southport , Queensland , Australia.,d School of Medicine, Griffith University , Southport , Queensland , Australia
| | - Amanda J Cox
- a School of Medical Science, Griffith University , Southport , Queensland , Australia.,b Menzies Health Institute Queensland , Southport , Queensland , Australia
| |
Collapse
|
20
|
Drago-Serrano ME, Campos-Rodríguez R, Carrero JC, de la Garza M. Lactoferrin: Balancing Ups and Downs of Inflammation Due to Microbial Infections. Int J Mol Sci 2017; 18:E501. [PMID: 28257033 PMCID: PMC5372517 DOI: 10.3390/ijms18030501] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/13/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Lactoferrin (Lf) is a glycoprotein of the primary innate immune-defense system of mammals present in milk and other mucosal secretions. This protein of the transferrin family has broad antimicrobial properties by depriving pathogens from iron, or disrupting their plasma membranes through its highly cationic charge. Noteworthy, Lf also exhibits immunomodulatory activities performing up- and down-regulation of innate and adaptive immune cells, contributing to the homeostasis in mucosal surfaces exposed to myriad of microbial agents, such as the gastrointestinal and respiratory tracts. Although the inflammatory process is essential for the control of invasive infectious agents, the development of an exacerbated or chronic inflammation results in tissue damage with life-threatening consequences. In this review, we highlight recent findings in in vitro and in vivo models of the gut, lung, oral cavity, mammary gland, and liver infections that provide experimental evidence supporting the therapeutic role of human and bovine Lf in promoting some parameters of inflammation and protecting against the deleterious effects of bacterial, viral, fungal and protozoan-associated inflammation. Thus, this new knowledge of Lf immunomodulation paves the way to more effective design of treatments that include native or synthetic Lf derivatives, which may be useful to reduce immune-mediated tissue damage in infectious diseases.
Collapse
Affiliation(s)
- Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco (UAM-X), CdMx 04960, Mexico.
| | - Rafael Campos-Rodríguez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional (ESM-IPN), CdMx 11340, Mexico.
| | - Julio César Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (IIB-UNAM), CdMx 70228, Mexico.
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), CdMx 07360, Mexico.
| |
Collapse
|
21
|
Wang X, Wang X, Hao Y, Teng D, Wang J. Research and development on lactoferrin and its derivatives in China from 2011–2015. Biochem Cell Biol 2017; 95:162-170. [DOI: 10.1139/bcb-2016-0073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lactoferrin (Lf), a multifunctional glycoprotein, is an important antimicrobial and immune regulatory protein present in neutrophils and most exocrine secretions of mammals. Lactoferricin (Lfcin) is located in the N-terminal region of this protein. In this review, the current state of research into Lf and Lfcin in China is described. Searching with HistCite software in Web Sci located 118 papers published by Chinese researchers from 2011–2015, making China one of the top 3 producers of Lf research and development in the world. The biological functions of Lf and Lfcin are discussed, including antibacterial, antiviral, antifungal, anticarcinogenic, and anti-inflammatory activities; targeted drug delivery, induction of neurocyte, osteoblast, and tenocyte growth, and possible mechanisms of action. The preparation and heterologous expression of Lf in animals, bacteria, and yeast are discussed in detail. Five Lf-related food additive factories and 9 Lf-related health food production companies are certified by the China Food and Drug Administration (CFDA). The latest progress in the generation of transgenic livestock in China, the safety of the use of transgenic animals, and future prospects for the uses of Lf and Lfcin are also covered.
Collapse
Affiliation(s)
- Xiao Wang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture, Beijing 100081, P.R. China
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| | - Xiumin Wang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture, Beijing 100081, P.R. China
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| | - Ya Hao
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture, Beijing 100081, P.R. China
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| | - Da Teng
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture, Beijing 100081, P.R. China
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| | - Jianhua Wang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture, Beijing 100081, P.R. China
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| |
Collapse
|
22
|
Kanwar JR, Kanwar RK, Stathopoulos S, Haggarty NW, MacGibbon AKH, Palmano KP, Roy K, Rowan A, Krissansen GW. Comparative activities of milk components in reversing chronic colitis. J Dairy Sci 2016; 99:2488-2501. [PMID: 26805965 DOI: 10.3168/jds.2015-10122] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/16/2015] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel disease (IBD) is a poorly understood chronic immune disorder for which there is no medical cure. Milk and colostrum are rich sources of bioactives with immunomodulatory properties. Here we compared the therapeutic effects of oral delivery of bovine milk-derived iron-saturated lactoferrin (Fe-bLF), angiogenin, osteopontin (OPN), colostrum whey protein, Modulen IBD (Nestle Healthsciences, Rhodes, Australia), and cis-9,trans-11 conjugated linoleic acid (CLA)-enriched milk fat in a mouse model of dextran sulfate-induced colitis. The CLA-enriched milk fat significantly increased mouse body weights after 24d of treatment, reduced epithelium damage, and downregulated the expression of proinflammatory cytokines and nitrous oxide. Modulen IBD most effectively decreased the clinical score at d 12, and Modulen IBD and OPN most effectively lowered the inflammatory score. Myeloperoxidase activity that denotes neutrophil infiltration was significantly lower in mice fed Modulen IBD, OPN, angiogenin, and Fe-bLF. A significant decrease in the numbers of T cells, natural killer cells, dendritic cells, and a significant decrease in cytokine expression were observed in mice fed the treatment diets compared with dextran sulfate administered mice. The Fe-bLF, CLA-enriched milk fat, and Modulen IBD inhibited intestinal angiogenesis. In summary, each of the milk components attenuated IBD in mice, but with differing effectiveness against specific disease parameters.
Collapse
Affiliation(s)
- J R Kanwar
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), Centre for Molecular and Medical Research (C-MMR), School of Medicine (SoM), Faculty of Health, Deakin University, Waurn Ponds, Victoria 3217, Australia.
| | - R K Kanwar
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), Centre for Molecular and Medical Research (C-MMR), School of Medicine (SoM), Faculty of Health, Deakin University, Waurn Ponds, Victoria 3217, Australia
| | | | - N W Haggarty
- Fonterra Research Centre, Palmerston North, New Zealand
| | | | - K P Palmano
- Fonterra Research Centre, Palmerston North, New Zealand
| | - K Roy
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (NLIMBR), Centre for Molecular and Medical Research (C-MMR), School of Medicine (SoM), Faculty of Health, Deakin University, Waurn Ponds, Victoria 3217, Australia
| | - A Rowan
- Fonterra Research Centre, Palmerston North, New Zealand
| | - G W Krissansen
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
23
|
Li W, Fu K, Lv X, Wang Y, Wang J, Li H, Tian W, Cao R. Lactoferrin suppresses lipopolysaccharide-induced endometritis in mice via down-regulation of the NF-κB pathway. Int Immunopharmacol 2015; 28:695-9. [DOI: 10.1016/j.intimp.2015.07.040] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/14/2022]
|
24
|
Gualdi L, Mertz S, Gomez AM, Ramilo O, Wittke A, Mejias A. Lack of effect of bovine lactoferrin in respiratory syncytial virus replication and clinical disease severity in the mouse model. Antiviral Res 2013; 99:188-95. [PMID: 23735300 DOI: 10.1016/j.antiviral.2013.05.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Revised: 05/10/2013] [Accepted: 05/24/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Lactoferrin (LF) is a glycoprotein present in human milk with known antimicrobial effects. In vitro, LF has demonstrated antiviral activity against respiratory syncytial virus (RSV). We sought to assess the effect of bovine (b)LF in RSV replication, lung inflammation and function, cytokine profiles and clinical disease in an in vivo murine model. METHODS Female BALB/c mice were inoculated with 10(7)PFU RSV A2 or 10% EMEM. bLF or placebo (DPBS) were administered once or twice daily by oral gavage or intraperitoneal (IP) injection at doses ranging from 2 to 10mg/animal/day, from 48h before until 96h post-RSV inoculation. Bronchoalveolar lavage (BAL), whole lung and serum samples were harvested on day 5 post-inoculation to asses RSV loads, lung inflammation and cytokine concentrations. Weight loss, airway obstruction and disease severity were assessed daily in all groups. RESULTS On day 5 post-inoculation BAL RSV loads, lung inflammation and serum innate, Th1, Th2 and Th17 cytokine concentrations showed no differences between RSV infected mice treated with bLF and RSV infected but untreated mice independent of bLF dosing and administration route (p>0.05). In addition, all bLF groups showed similar weight loss, degree of airway obstruction, and disease severity scores on days 1-5 post-inoculation which was comparable to infected untreated mice (p>0.05) but higher than uninfected controls. CONCLUSIONS Administration of oral or IP bLF at different doses did not demonstrate antiviral activity or significant effects on disease severity in the RSV mouse model. Whether these observations could be extrapolated to infants at risk for RSV infection needs to be further explored.
Collapse
Affiliation(s)
- Lucien Gualdi
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, United States
| | | | | | | | | | | |
Collapse
|
25
|
Ortega-Gómez A, Perretti M, Soehnlein O. Resolution of inflammation: an integrated view. EMBO Mol Med 2013; 5:661-74. [PMID: 23592557 PMCID: PMC3662311 DOI: 10.1002/emmm.201202382] [Citation(s) in RCA: 512] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/22/2013] [Accepted: 03/01/2013] [Indexed: 12/16/2022] Open
Abstract
Resolution of inflammation is a coordinated and active process aimed at restoration of tissue integrity and function. This review integrates the key molecular and cellular mechanisms of resolution. We describe how abrogation of chemokine signalling blocks continued neutrophil tissue infiltration and how apoptotic neutrophils attract monocytes and macrophages to induce their clearance. Uptake of apoptotic neutrophils by macrophages reprograms macrophages towards a resolving phenotype, a key event to restore tissue homeostasis. Finally, we highlight the therapeutic potential that derives from understanding the mechanisms of resolution.
Collapse
Affiliation(s)
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of MedicineLondon, UK
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, LMUMunich, Germany
- Department of Pathology, AMCAmsterdam, The Netherlands
| |
Collapse
|