1
|
Lee S, Kim B, Lee MJ, Kim D, Park S, Kim J, Arai Y, Lee SH. Curcumin-PLGA NPs coated with targeting biomimetic personalized stem cell membranes for osteoarthritis therapy. J Control Release 2025; 381:113625. [PMID: 40081740 DOI: 10.1016/j.jconrel.2025.113625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Traditional drug delivery systems for OA treatments face limitations due to rapid clearance within the joint and low biocompatibility. Moreover, the inflammation associated with OA exacerbates tissue damage and delays the regenerative capacity of therapeutics. To overcome these limitations, an OA-specific drug delivery system designated dCOL2-CM-Cur-PNPs is developed herein to target OA cartilage for anti-inflammatory and cartilage regeneration purposes. This system is constructed using cell membranes obtained from induced pluripotent stem cell -derived mesenchymal stem cells (iMSC-CMs), poly(D,l-lactide-co-glycolide) (PLGA) nanoparticles loaded with the well-known anti-inflammatory and cartilage-regenerating agent curcumin (Cur-PNPs), and damaged type II collagen (dCOL2)-targeting phospholipids. Coating the Cur-PNPs with iMSC-CMs enhances the sustained release of curcumin and improves its cellular uptake by OA-induced chondrocytes. The dCOL2-CM-Cur-PNPs restores the chondrogenic properties of the OA-induced chondrocytes, inhibit the pro-inflammatory function of M1 macrophages, and promote the anti-inflammatory function of M2 macrophages. The dCOL2-targeting phospholipids integrated on the surface of the iMSC-CMs facilitate specific binding to OA cartilage, as validated by in-vitro and in-vivo experiments. Additionally, the dCOL2-CM-Cur-PNPs alleviate OA progression in a DMM rat model. This drug delivery system based on iMSC-CMs modified with dCOL2-targeting phospholipids demonstrates significant potential as a next-generation platform for promoting cartilage regeneration through OA-specific therapy.
Collapse
Affiliation(s)
- Sunjun Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Bowon Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Min-Ju Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Deogil Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Sunghyun Park
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Jinsik Kim
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620 Seoul, South Korea.
| |
Collapse
|
2
|
Vichare R, Kulahci Y, McCallin R, Zor F, Selek FN, Liu L, Crelli C, Troidle A, Herneisey M, Nichols JM, Shepherd AJ, Gorantla VS, Janjic JM. Theranostic nanoemulsions suppress macrophage-mediated acute inflammation in rats. J Nanobiotechnology 2025; 23:80. [PMID: 39905487 PMCID: PMC11792706 DOI: 10.1186/s12951-025-03164-w] [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: 08/03/2024] [Accepted: 01/25/2025] [Indexed: 02/06/2025] Open
Abstract
In inflammatory diseases or following an injury, dysregulated inflammation is a common driver of pain and tissue damage. Macrophages are immune cells that contribute to the initiation, maintenance, and resolution of inflammation due to their phenotypic plasticity in response to signals from inflammatory microenvironments. Macrophages infiltrate and polarize toward a pro-inflammatory phenotype (M1-like), thereby increasing the severity of inflammation. Therefore, we aimed to suppress the pro-inflammatory activity of M1-like macrophages and decrease their infiltration at the site of inflammatory insult to resolve tissue inflammation. To achieve this, we developed a theranostic curcumin-loaded nanoemulsion platform that delivers a low dose of curcumin, a known anti-inflammatory phytochemical, to macrophages and allows in vivo tracking of macrophages by near-infrared fluorescence (NIRF) imaging technique. In vitro, we showed that curcumin-loaded nanoemulsion suppressed polarization of macrophages towards M1-like phenotype, consequently decreasing the release of pro-inflammatory cytokines and mediators like IL-6, IL-[Formula: see text] TNF-[Formula: see text], and nitric oxide (NO). Furthermore, curcumin-loaded nanoemulsion increased the level of IL-10, an anti-inflammatory cytokine, and protected macrophages against ferroptosis compared to drug-free nanoemulsion. In a rodent model of Complete Freund's adjuvant (CFA)-induced inflammation, we demonstrated that infiltrating macrophages sequestered curcumin-loaded nanoemulsion droplets and acted as cellular drug depots at the site of inflammation. This consequently decreased macrophage infiltration at the CFA-induced inflammation site in both sexes compared to drug-free nanoemulsion, as demonstrated by NIRF imaging, H&E staining, and immunofluorescence. Taken together, our results indicated that the anti-inflammatory efficacy of curcumin was significantly improved when directly delivered to pro-inflammatory macrophages via theranostic nanoemulsion. This work opens an avenue for exploring theranostic nanoemulsions as a platform for delivering natural anti-inflammatory products for immune modulation.
Collapse
Affiliation(s)
- Riddhi Vichare
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Yalcin Kulahci
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Rebecca McCallin
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Fatih Zor
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Fatma Nurefsan Selek
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lu Liu
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Caitlin Crelli
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Anneliese Troidle
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Michele Herneisey
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - James M Nichols
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Shepherd
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vijay S Gorantla
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jelena M Janjic
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Cervia D, Zecchini S, Pincigher L, Roux-Biejat P, Zalambani C, Catalani E, Arcari A, Del Quondam S, Brunetti K, Ottria R, Casati S, Vanetti C, Barbalace MC, Prata C, Malaguti M, Casati SR, Lociuro L, Giovarelli M, Mocciaro E, Falcone S, Fenizia C, Moscheni C, Hrelia S, De Palma C, Clementi E, Perrotta C. Oral administration of plumbagin is beneficial in in vivo models of Duchenne muscular dystrophy through control of redox signaling. Free Radic Biol Med 2024; 225:193-207. [PMID: 39326684 DOI: 10.1016/j.freeradbiomed.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/12/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease. Recently approved molecular/gene treatments do not solve the downstream inflammation-linked pathophysiological issues such that supportive therapies are required to improve therapeutic efficacy and patients' quality of life. Over the years, a plethora of bioactive natural compounds have been used for human healthcare. Among them, plumbagin, a plant-derived analog of vitamin K3, has shown interesting potential to counteract chronic inflammation with potential therapeutic significance. In this work we evaluated the effects of plumbagin on DMD by delivering it as an oral supplement within food to dystrophic mutant of the fruit fly Drosophila melanogaster and mdx mice. In both DMD models, plumbagin show no relevant adverse effect. In terms of efficacy plumbagin improved the climbing ability of the dystrophic flies and their muscle morphology also reducing oxidative stress in muscles. In mdx mice, plumbagin enhanced the running performance on the treadmill and the muscle strength along with muscle morphology. The molecular mechanism underpinning these actions was found to be the activation of nuclear factor erythroid 2-related factor 2 pathway, the re-establishment of redox homeostasis and the reduction of inflammation thus generating a more favorable environment for skeletal muscles regeneration after damage. Our data provide evidence that food supplementation with plumbagin modulates the main, evolutionary conserved, mechanistic pathophysiological hallmarks of dystrophy, thus improving muscle function in vivo; the use of plumbagin as a therapeutic in humans should thus be explored further.
Collapse
MESH Headings
- Naphthoquinones/administration & dosage
- Naphthoquinones/pharmacology
- Animals
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Mice
- Administration, Oral
- Mice, Inbred mdx
- Oxidation-Reduction/drug effects
- Signal Transduction/drug effects
- Disease Models, Animal
- Drosophila melanogaster
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Oxidative Stress/drug effects
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/genetics
- Humans
- Male
Collapse
Affiliation(s)
- Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università Degli Studi Della Tuscia, Viterbo, 01100, Italy
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy
| | - Luca Pincigher
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-Università di Bologna, Bologna, 40126, Italy
| | - Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy
| | - Chiara Zalambani
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-Università di Bologna, Bologna, 40126, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università Degli Studi Della Tuscia, Viterbo, 01100, Italy
| | - Alessandro Arcari
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy
| | - Simona Del Quondam
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università Degli Studi Della Tuscia, Viterbo, 01100, Italy
| | - Kashi Brunetti
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università Degli Studi Della Tuscia, Viterbo, 01100, Italy
| | - Roberta Ottria
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy
| | - Sara Casati
- Department of Biomedical, Surgical, and Dental Science (DISBIOC), Università Degli Studi di Milano, Milano, 20133, Italy
| | - Claudia Vanetti
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy; Department of Pathophysiology and Transplantation (DEPT), Università Degli Studi di Milano, Milano, 20122, Italy
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, 47921, Italy
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-Università di Bologna, Bologna, 40126, Italy
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, 47921, Italy
| | - Silvia Rosanna Casati
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università Degli Studi di Milano, 20054, Segrate, Italy
| | - Laura Lociuro
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, 47921, Italy
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy
| | - Emanuele Mocciaro
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy; Gene Expression and Muscular Dystrophy Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milano, 20132, Italy
| | - Sestina Falcone
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, F-75013, France
| | - Claudio Fenizia
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy; Department of Pathophysiology and Transplantation (DEPT), Università Degli Studi di Milano, Milano, 20122, Italy
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, 47921, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università Degli Studi di Milano, 20054, Segrate, Italy
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy; IRCCS Eugenio Medea, Bosisio Parini, 23842, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences (DIBIC), Università Degli Studi di Milano, Milano, 20157, Italy.
| |
Collapse
|
4
|
Tian L, Chen J, Yang M, Chen L, Qiu J, Jiang Y, Tan X, Qian Q, Liang X, Dou X. Xiezhuo Tiaozhi formula inhibits macrophage pyroptosis in the non-alcoholic fatty liver disease by targeting the SIRT1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155776. [PMID: 38851104 DOI: 10.1016/j.phymed.2024.155776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a challenging disease to interfere with and represents a potential long-term risk factor for hepatic fibrosis and liver cancer. The Xiezhuo Tiaozhi (XZTZ) formula, a water extract from crude herbs, has been widely used as an anti-NAFLD agent through clinical observation. However, the underlying pharmacological mechanisms of the XZTZ formula and its impact on the potential pathways against NAFLD have not been elucidated. PURPOSE Our study aims to investigate the pharmacological effects and underlying regulatory mechanisms of the XZTZ formula to treat NAFLD. METHODS The possible active components and pharmacological mechanisms of the XZTZ formula against NAFLD were identified using ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS) and molecular docking. To further explore the potential mechanisms, forty-eight 6-week-old male C57BL/6 J mice were given individual attention with high-fat and high-sugar diet (HFHSD) or relevant control (Ctrl) diets for 16 weeks to successfully construct a NAFLD mouse model. Subsequently, the levels of serum biochemicals, pathological changes in the liver, and pyroptosis levels were assessed in mice to investigate the therapeutic effects of the XZTZ formula. Further, LPS-induced RAW264.7 cells and Immortalized Mouse Kupffer cells (ImKC) were used to verify the potential mechanisms of the XZTZ formula against NAFLD in vitro. RESULTS We identified 7 chemical compounds and 2 potential therapeutic targets as plausible therapeutic points for the treatment of NAFLD using the XZTZ formula. Subsequent histopathological analysis revealed marked hepatic steatosis and lipid accumulation in the HFHSD mice liver, while conditions were effectively ameliorated by administration of the XZTZ formula. Additionally, our work demonstrated that the XZTZ formula could attenuate M1 polarization, promote M2 polarization, and suppress pyroptosis via the SIRT1 pathway in tissue samples. Moreover, validation performed through LPS-induced RAW264.7 and ImKC cells by showing that silencing SIRT1 weaken the effects of the XZTZ formula on relative pyroptosis affirmed that its role was associated with the SIRT1 pathway in macrophage. CONCLUSION These findings suggest that the XZTZ formula alleviated hepatic steatosis and lipid accumulation in NAFLD mice. These ameliorations are associated with mechanisms involving the attenuation of M1 polarization, promotion of M2 polarization, and anti-pyroptosis effects through the SIRT1 pathway.
Collapse
Affiliation(s)
- Lulu Tian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Meiqi Yang
- Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, Liaoning, China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuwei Jiang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaolong Tan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qianyu Qian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiao Liang
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Di Paola A, Marrapodi MM, Pota E, Colucci Cante R, Rana D, Giliberti G, Di Feo G, Ahmed S, Roberti D, Nigro R, Rossi F, Argenziano M. Role of Nutraceuticals in Counteracting Inflammation in In Vitro Macrophages Obtained from Childhood Cancer Survivors. Cancers (Basel) 2024; 16:714. [PMID: 38398105 PMCID: PMC10886672 DOI: 10.3390/cancers16040714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
The advancement of anti-cancer therapies has markedly improved the survival rate of children with cancer, making them long-term childhood cancer survivors (CCS). Nevertheless, these treatments cause a low-grade inflammatory state, determining inflamm-aging and, thus, favoring the early onset of chronic diseases normally associated with old age. Identification of novel and safer therapeutic strategies is needed to counteract and prevent inflamm-aging. Macrophages are cells involved in immune and inflammatory responses, with a pivotal role in iron metabolism, which is related to inflammation. We obtained macrophages from CCS patients and evaluated their phenotype markers, inflammatory states, and iron metabolism by Western blotting, ELISA, and iron assays. We observed a strong increase in classically activated phenotype markers (M1) and iron metabolism alteration in CCS, with an increase in intracellular iron concentration and inflammatory markers. These results suggest that the prevalence of M1 macrophages and alteration of iron metabolism could be involved in the worsening of inflammation in CCS. Therefore, we propose macrophages and iron metabolism as novel therapeutic targets to counteract inflamm-aging. To avoid toxic regimens, we tested some nutraceuticals (resveratrol, curcumin, and oil-enriched lycopene), which are already known to exert anti-inflammatory properties. After their administration, we observed a macrophage switch towards the anti-inflammatory phenotype M2, as well as reductions in pro-inflammatory cytokines and the intracellular iron concentration. Therefore, we suggest-for the first time-that nutraceuticals reduce inflammation in CCS macrophages through a novel anti-inflammatory mechanism of action, modulating iron metabolism.
Collapse
Affiliation(s)
- Alessandra Di Paola
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.D.P.); (M.M.M.); (E.P.); (G.D.F.); (D.R.); (M.A.)
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.D.P.); (M.M.M.); (E.P.); (G.D.F.); (D.R.); (M.A.)
| | - Elvira Pota
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.D.P.); (M.M.M.); (E.P.); (G.D.F.); (D.R.); (M.A.)
| | - Rosa Colucci Cante
- Department of Industrial Engineering, University of Niccolò Cusano, 00166 Rome, Italy;
| | - Deeksha Rana
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (D.R.); (G.G.); (S.A.)
| | - Giulia Giliberti
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (D.R.); (G.G.); (S.A.)
| | - Giuseppe Di Feo
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.D.P.); (M.M.M.); (E.P.); (G.D.F.); (D.R.); (M.A.)
| | - Shakeel Ahmed
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (D.R.); (G.G.); (S.A.)
| | - Domenico Roberti
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.D.P.); (M.M.M.); (E.P.); (G.D.F.); (D.R.); (M.A.)
| | - Roberto Nigro
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, 80125 Naples, Italy;
| | - Francesca Rossi
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.D.P.); (M.M.M.); (E.P.); (G.D.F.); (D.R.); (M.A.)
| | - Maura Argenziano
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.D.P.); (M.M.M.); (E.P.); (G.D.F.); (D.R.); (M.A.)
| |
Collapse
|
6
|
Apeku E, Tantuoyir MM, Zheng R, Tanye N. Exploring the polarization of M1 and M2 macrophages in the context of skin diseases. Mol Biol Rep 2024; 51:269. [PMID: 38302766 DOI: 10.1007/s11033-023-09014-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/30/2023] [Indexed: 02/03/2024]
Abstract
Macrophages are critical components of the immune system and play vital roles in pathogen defense, immune regulation, and tissue repair. These cells exhibit different polarization states depending on environmental signals, and the M1/M2 paradigm is a useful tool for comprehending these states. This review article comprehensively presents the underlying mechanisms of M1 and M2 macrophage polarization and examines their polarization in various skin diseases. Additionally, this paper discusses therapeutic strategies that target M1 and M2 macrophage polarization in skin diseases. A more profound understanding of macrophage polarization in skin diseases could provide valuable insights for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Ernestina Apeku
- Department of Dermatology, The 1st Hospital of Shanxi Medical University; Graduate Department of Shanxi Medical University, Taiyuan, Shanxi, China
| | | | - Rui Zheng
- Department of Dermatology, The 1st Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Nestor Tanye
- School of Automation Science and Engineering, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
7
|
Yang C, Zhu Q, Chen Y, Ji K, Li S, Wu Q, Pan Q, Li J. Review of the Protective Mechanism of Curcumin on Cardiovascular Disease. Drug Des Devel Ther 2024; 18:165-192. [PMID: 38312990 PMCID: PMC10838105 DOI: 10.2147/dddt.s445555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death worldwide and has been the focus of research in the medical community. Curcumin is a polyphenolic compound extracted from the root of turmeric. Curcumin has been shown to have a variety of pharmacological properties over the past decades. Curcumin can significantly protect cardiomyocyte injury after ischemia and hypoxia, inhibit myocardial hypertrophy and fibrosis, improve ventricular remodeling, reduce drug-induced myocardial injury, improve diabetic cardiomyopathy(DCM), alleviate vascular endothelial dysfunction, inhibit foam cell formation, and reduce vascular smooth muscle cells(VSMCs) proliferation. Clinical studies have shown that curcumin has a protective effect on blood vessels. Toxicological studies have shown that curcumin is safe. But high doses of curcumin also have some side effects, such as liver damage and defects in embryonic heart development. This article reviews the mechanism of curcumin intervention on CVDs in recent years, in order to provide reference for the development of new drugs in the future.
Collapse
Affiliation(s)
- Chunkun Yang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qinwei Zhu
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Yanbo Chen
- Department of Arrhythmia, Weifang People's Hospital, Weifang, Shandong, People's Republic of China
| | - Kui Ji
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Shuanghong Li
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Qian Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qingquan Pan
- Department of Emergency, Weifang Hospital of Traditional Chinese Medicine, Weifang, Shandong, People's Republic of China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
8
|
Wang X, Wang Y, Tang T, Zhao G, Dong W, Li Q, Liang X. Curcumin-Loaded RH60/F127 Mixed Micelles: Characterization, Biopharmaceutical Characters and Anti-Inflammatory Modulation of Airway Inflammation. Pharmaceutics 2023; 15:2710. [PMID: 38140051 PMCID: PMC10747166 DOI: 10.3390/pharmaceutics15122710] [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: 09/28/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Curcumin's ability to impact chronic inflammatory conditions, such as metabolic syndrome and arthritis, has been widely researched; however, its poor bioavailability limits its clinical application. The present study is focused on the development of curcumin-loaded polymeric nanomicelles as a drug delivery system with anti-inflammatory effects. Curcumin was loaded in PEG-60 hydrogenated castor oil and puronic F127 mixed nanomicelles (Cur-RH60/F127-MMs). Cur-RH60/F127-MMs was prepared using the thin film dispersion method. The morphology and releasing characteristics of nanomicelles were evaluated. The uptake and permeability of Cur-RH60/F127-MMs were investigated using RAW264.7 and Caco-2 cells, and their bioavailability and in vivo/vitro anti-inflammatory activity were also evaluated. The results showed that Cur-RH60/F127-MMs have regular sphericity, possess an average diameter smaller than 20 nm, and high encapsulation efficiency for curcumin (89.43%). Cur-RH60/F127-MMs significantly increased the cumulative release of curcumin in vitro and uptake by cells (p < 0.01). The oral bioavailability of Cur-RH60/F127-MMs was much higher than that of curcumin-active pharmaceutical ingredients (Cur-API) (about 9.24-fold). The treatment of cell lines with Cur-RH60/F127-MMs exerted a significantly stronger anti-inflammatory effect compared to Cur-API. In addition, Cur-RH60/F127-MMs significantly reduced OVA-induced airway hyperresponsiveness and inflammation in an in vivo experimental asthma model. In conclusion, this study reveals the possibility of formulating a new drug delivery system for curcumin, in particular nanosized micellar aqueous dispersion, which could be considered a perspective platform for the application of curcumin in inflammatory diseases of the airways.
Collapse
Affiliation(s)
- Xinli Wang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.W.); (G.Z.); (W.D.); (Q.L.)
- Jiangxi Medical Device Testing Center, Nanchang 330029, China
| | - Yanyan Wang
- Clinical Medical School, Jiangxi University of Chinese Medicine, Nanchang 330004, China;
| | - Tao Tang
- Department of Pharmacy, Ji’an Central People’s Hospital, Ji’an 343000, China;
| | - Guowei Zhao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.W.); (G.Z.); (W.D.); (Q.L.)
| | - Wei Dong
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.W.); (G.Z.); (W.D.); (Q.L.)
| | - Qiuxiang Li
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.W.); (G.Z.); (W.D.); (Q.L.)
| | - Xinli Liang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.W.); (G.Z.); (W.D.); (Q.L.)
| |
Collapse
|
9
|
Galindo CL, Khan S, Zhang X, Yeh YS, Liu Z, Razani B. Lipid-laden foam cells in the pathology of atherosclerosis: shedding light on new therapeutic targets. Expert Opin Ther Targets 2023; 27:1231-1245. [PMID: 38009300 PMCID: PMC10843715 DOI: 10.1080/14728222.2023.2288272] [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: 06/28/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Abstract
INTRODUCTION Lipid-laden foam cells within atherosclerotic plaques are key players in all phases of lesion development including its progression, necrotic core formation, fibrous cap thinning, and eventually plaque rupture. Manipulating foam cell biology is thus an attractive therapeutic strategy at early, middle, and even late stages of atherosclerosis. Traditional therapies have focused on prevention, especially lowering plasma lipid levels. Despite these interventions, atherosclerosis remains a major cause of cardiovascular disease, responsible for the largest numbers of death worldwide. AREAS COVERED Foam cells within atherosclerotic plaques are comprised of macrophages, vascular smooth muscle cells, and other cell types which are exposed to high concentrations of lipoproteins accumulating within the subendothelial intimal layer. Macrophage-derived foam cells are particularly well studied and have provided important insights into lipid metabolism and atherogenesis. The contributions of foam cell-based processes are discussed with an emphasis on areas of therapeutic potential and directions for drug development. EXERT OPINION As key players in atherosclerosis, foam cells are attractive targets for developing more specific, targeted therapies aimed at resolving atherosclerotic plaques. Recent advances in our understanding of lipid handling within these cells provide insights into how they might be manipulated and clinically translated to better treat atherosclerosis.
Collapse
Affiliation(s)
- Cristi L. Galindo
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Saifur Khan
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Yu-Sheng Yeh
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Ziyang Liu
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
| |
Collapse
|
10
|
Wang X, Liao Z, Zhao G, Dong W, Huang X, Zhou X, Liang X. Curcumin nanocrystals self-stabilized Pickering emulsion freeze-dried powder: Development, characterization, and suppression of airway inflammation. Int J Biol Macromol 2023:125493. [PMID: 37348593 DOI: 10.1016/j.ijbiomac.2023.125493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/28/2023] [Accepted: 06/18/2023] [Indexed: 06/24/2023]
Abstract
Curcumin, a diketone compound extracted from turmeric's rhizome, is an effective anti-inflammatory drug with multiple pharmacological activities. However, its low oral bioavailability due to its low water solubility and permeability severely limits its clinical applications. Therefore, to enhance the oral bioavailability of curcumin, further enhance its anti-inflammatory effects, and improve its potential in the treatment of airway inflammation, a curcumin nanocrystalline self-stabilizing Pickering emulsion (Cur-NSSPE) was prepared through high-pressure homogenization. Next, Cur-NSSPE was dried using a freeze-drying method to produce Cur-NSSPE-FDP. The prepared Cur-NSSPE and Cur-NSSPE-FDP were physically characterized. The release behavior and transmembrane transport capability of Cur-NSSPE-FDP in vitro were evaluated. Pharmacokinetic study was performed to evaluate its oral bioavailability. The anti-inflammatory effects of Cur-NSSPE-FDP in vivo and in vitro were investigated using RAW 264.7 macrophage inflammation model induced by LPS and IFN-γ and asthma model in BALB/c mice induced by OVA. The average particle size of Cur-NSSPE was (163.66 ± 6.78) nm, and the average drug content was (2.78 ± 0.01) mg/mL. The transmission electron microscopy results showed that the droplets were spherical in shape with a relatively uniform size, and the curcumin nanocrystals formed a spherical core-shell structure wrapped at the interface of the droplets. The scanning electron microscopy showed that Cur-NSSPE-FDP was a neatly arranged, having loose and porous network structure. Furthermore, it can significantly improve the cumulative release of curcumin in vitro and improve oral bioavailability in rats, increase the uptake of RAW264.7 and Caco-2 cells, promote the transport of curcumin across Caco-2 cells, significantly inhibit the expression of inflammatory factors NO, IL-6, TNF-a, MDA, IgE and ICAM-1, and improve the expression of IL-10 and SOD. These results indicated that the curcumin nanocrystalline self-stabilizing Pickering emulsion-freeze dried powder improved the oral bioavailability of curcumin and enhanced its therapeutic effect in airway inflammation.
Collapse
Affiliation(s)
- Xinli Wang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Zhenggen Liao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Guowei Zhao
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Wei Dong
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Xiaoying Huang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Xiang Zhou
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, People's Republic of China.
| | - Xinli Liang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, People's Republic of China.
| |
Collapse
|
11
|
Jing J, Zhu C, Gong R, Qi X, Zhang Y, Zhang Z. Research progress on the active ingredients of traditional Chinese medicine in the intervention of atherosclerosis: A promising natural immunotherapeutic adjuvant. Biomed Pharmacother 2023; 159:114201. [PMID: 36610225 DOI: 10.1016/j.biopha.2022.114201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease caused by disorders of lipid metabolism. Abnormal deposition of low-density lipoproteins in the arterial wall stimulates the activation of immune cells, including the adhesion and infiltration of monocytes, the proliferation and differentiation of macrophages and lymphocytes, and the activation of their functions. The complex interplay between immune cells coordinates the balance between pro- and anti-inflammation and plays a key role in the progression of AS. Therefore, targeting immune cell activity may lead to the development of more selective drugs with fewer side effects to treat AS without compromising host defense mechanisms. At present, an increasing number of studies have found that the active ingredients of traditional Chinese medicine (TCM) can regulate the function of immune cells in multiple ways to against AS, showing great potential for the treatment of AS and promising clinical applications. In this paper, we review the mechanisms of immune cell action in AS lesions and the potential targets and/or pathways for immune cell regulation by the active ingredients of TCM to promote the understanding of the immune system interactions of AS and provide a relevant basis for the use of active ingredients of TCM as natural adjuvants for AS immunotherapy.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Chaojun Zhu
- Surgical Department of Traditional Chinese Medicine, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Rui Gong
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Xue Qi
- Department of General Surgery, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250001, China.
| | - Yue Zhang
- Peripheral Vascular Disease Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Zhaohui Zhang
- Surgical Department of Traditional Chinese Medicine, Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
12
|
Sesquiterpenoids from the Florets of Carthamus tinctorius (Safflower) and Their Anti-Atherosclerotic Activity. Nutrients 2022; 14:nu14245348. [PMID: 36558507 PMCID: PMC9783904 DOI: 10.3390/nu14245348] [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: 10/28/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
(1) Background: The florets of Carthamus tinctorius L. are traditionally used as a blood-activating drug and can be used for the treatment of atherosclerosis, but no compounds with anti-atherosclerotic activity have been reported. (2) Methods: This study investigated the chemical compounds from the florets of C. tinctorius. Comprehensive spectroscopic techniques revealed their structures, and ECD calculations established their absolute configurations. Nile Red staining, Oil Red O staining, and cholesterol assessment were performed on these compounds and their aglycones for the inhibitory activity against the formation of foam cells induced by oxidized low-density lipoprotein (ox-LDL) in RAW264.7 macrophages. In addition, RAW264.7 macrophages were tested for their anti-inflammatory activity by measuring the inhibition of NO production caused by LPS. (3) Results: Five new sesquiterpenoids (1-5) isolated from the florets of C. tinctorius were identified as (-)-(1R,4S,9S,11R)-caryophyll-8(13)-en-14-ol-5-one (1), (+)-(1R,4R,9S,11R)-caryophyll-8(13)-en-14-ol-5-one (2), (-)-(3Z,1R,5S,8S,9S,11R)-5,8-epoxycaryophyll-3-en-14-O-β-D-glucopyranoside (3), (+)-(1S,7R,10S)-guai-4-en-3-one-11-O-β-D-fucopyranoside (4), and (-)-(2R,5R,10R)-vetispir-6-en-8-one-11-O-β-D-fucopyranoside (5). All compounds except for compound 3 reduced the lipid content in ox-LDL-treated RAW264.7 cells. Compounds 3 and 4 and their aglycones were found to reduce the level of total cholesterol (TC) and free cholesterol (FC) in ox-LDL-treated RAW264.7 cells. However, no compounds showed anti-inflammatory activity. (4) Conclusion: Sesquiterpenoids from C. tinctorius help to decrease the content of lipids, TC and FC in RAW264.7 cells, but they cannot inhibit NO production, which implies that their anti-atherogenic effects do not involve the inhibition of inflammation.
Collapse
|
13
|
Molaaghaee‐Rouzbahani S, Asri N, Jahani‐Sherafat S, Amani D, Masotti A, Baghaei K, Yadegar A, Mirjalali H, Rostami‐Nejad M. The modulation of macrophage subsets in celiac disease pathogenesis. Immun Inflamm Dis 2022; 10:e741. [PMID: 36444633 PMCID: PMC9667199 DOI: 10.1002/iid3.741] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/12/2022] [Accepted: 11/06/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND So far, limited studies have focused on the role of Macrophages (MQs) in the development or progression of celiac disease (CD). Researchers believe that increasing knowledge about the function of MQs in inflammatory disorders plays a critical role in finding a new treatment for these kinds of diseases. MAIN BODY CD is a permanent autoimmune intestinal disorder triggered by gluten exposure in predisposed individuals. This disorder happens due to the loss of intestinal epithelial barrier integrity characterized by dysregulated innate and adaptive immune responses. MQs are known as key players of the innate immune system that link innate and adaptive immunity. MQs of human intestinal lamina propria participate in maintaining tissue homeostasis, and also intestinal inflammation development. Previous studies suggested that gliadin triggers a proinflammatory phenotype (M1 MQ) in human primary MQs. Moreover, M2-related immunosuppressive mediators are also present in CD. In fact, CD patients present an impaired transition from pro-inflammatory to anti-inflammatory responses due to inappropriate responses to gliadin peptides. CONCLUSION The M1/M2 MQs polarization balancing regulators can be considered novel therapeutic targets for celiac disease.
Collapse
Affiliation(s)
| | - Nastaran Asri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Somayeh Jahani‐Sherafat
- Laser Application in Medical Sciences Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Davar Amani
- Department of Immunology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Andrea Masotti
- Bambino Gesù Children's Hospital‐IRCCSResearch LaboratoriesRomeItaly
| | - Kaveh Baghaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Rostami‐Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver DiseasesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
14
|
Monmai C, Kim JS, Baek SH. Transgenic Rice Seed Extracts Exert Immunomodulatory Effects by Modulating Immune-Related Biomarkers in RAW264.7 Macrophage Cells. Nutrients 2022; 14:nu14194143. [PMID: 36235795 PMCID: PMC9573073 DOI: 10.3390/nu14194143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/17/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Protopanaxadiol (PPD), a native active triterpenoid present in Panax ginseng, has been reported to exert immune-related effects. We previously created PPD-producing transgenic rice by introducing the P. ginseng protopanaxadiol synthase and dammarenediol-II synthase genes into Dongjin rice. In the present study, the seeds of the T4 generation of this transgenic rice were tested for their immunomodulatory effects in RAW264.7 macrophage cells. Treatment with transgenic rice seed extract in RAW264.7 cells (i) significantly enhanced nitric oxide (NO) production in a dose-dependent manner without any cytotoxicity (up to 100 µg/mL), (ii) upregulated the expression of immune-related genes and increased production of the inflammation mediator prostaglandin E2 (PGE2), and (iii) activated nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) by promoting the phosphorylation of NF-κB p65, p38 MAPK, and c-Jun N-terminal protein kinase (JNK). In lipopolysaccharide (LPS)-treated RAW264.7 cells used to mimic the inflammation condition, treatment with transgenic rice seed extract significantly reduced NO production, proinflammatory cytokine expression, and PGE2 production, all of which are LPS-induced inflammation biomarkers, by inhibiting the phosphorylation of NF-κB p65, p38 MAPK, and JNK. Collectively, these results indicate that PPD-producing transgenic rice has immunomodulatory effects.
Collapse
|
15
|
Huwait E, Ayoub M, Karim S. Investigation of the Molecular Mechanisms Underlying the Antiatherogenic Actions of Kaempferol in Human THP-1 Macrophages. Int J Mol Sci 2022; 23:ijms23137461. [PMID: 35806463 PMCID: PMC9267302 DOI: 10.3390/ijms23137461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is causing high mortality worldwide (World Health Organization-WHO, 2015). Atherosclerosis, the hardening and narrowing of arteries caused by the accumulation of fatty acids and lipids (cholesterol plaques), is a main reason of stroke, myocardial infarction, and angina. Present therapies for cardiovascular disease basically use statins such as β-Hydroxy β-methylglutaryl-CoA, with <70% efficacy and multiple side effects. An in vitro investigation was conducted to evaluate the impact of kaempferol, a natural medication, in an atherosclerotic cell model. We used cytotoxicity assays, Boyden chamber invasion assays, and quantitative PCR. Affymetrix microarrays were used to profile the entire transcriptome of kaempferol-treated cell lines, and Partek Genomic Suite was used to interpret the results. Kaempferol was not cytotoxic to THP-1 macrophages. In comparison to the control, kaempferol reduced monocyte migration mediated by monocyte chemotactic protein 1 (MCP-1) by 80%. The qPCR results showed a 73.7-fold reduction in MCP-1 and a 2.5-fold reduction in intercellular adhesion molecule 1 (ICAM-1) expression in kaempferol-treated cells. In interferon gamma (IFN-γ) without kaempferol and IFN-γ with kaempferol treated cells, we found 295 and 168 differentially expressed genes (DEGs), respectively. According to DEG pathway analysis, kaempferol exhibits anti-atherosclerosis and anti-inflammatory characteristics. Kaempferol is an effective and safe therapy for atherosclerosis.
Collapse
Affiliation(s)
- Etimad Huwait
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.H.); (M.A.)
- Cell Culture Unit and Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Maha Ayoub
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (E.H.); (M.A.)
- Cell Culture Unit and Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Correspondence: ; Tel.: +966-557581741
| |
Collapse
|
16
|
Song L, Zhang J, Ma D, Fan Y, Lai R, Tian W, Zhang Z, Ju J, Xu H. A Bibliometric and Knowledge-Map Analysis of Macrophage Polarization in Atherosclerosis From 2001 to 2021. Front Immunol 2022; 13:910444. [PMID: 35795675 PMCID: PMC9250973 DOI: 10.3389/fimmu.2022.910444] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/20/2022] [Indexed: 12/24/2022] Open
Abstract
In recent years, studies of macrophage polarization in atherosclerosis have become an intense area of research. However, there are few bibliometric analyses regarding this area. In this review, we used CiteSpace 5.8.R3 and VOSviewer 1.6.16 software to perform text mining and knowledge-map analysis. We explored the development process, knowledge structure, research hotspots, and potential trends using a bibliometric and knowledge-map analysis to provide researchers with a macroscopic view of this field. The studies concerning macrophage polarization in atherosclerosis were downloaded from the Web of Science Core Collection. A total of 781 studies were identified and published by 954 institutions from 51 countries/regions. The number of studies of macrophage polarization in atherosclerosis increased over time. Arteriosclerosis Thrombosis and Vascular Biology published the highest number of articles and was the top co-cited journal. De Winther was the most prolific researcher, and Moore had the most co-citations. The author co-occurrence map illustrated that there was active cooperation among researchers. The most productive countries were the United States and China. Amsterdam University, Harvard University, and Maastricht University were the top three productive institutions in the research field. Keyword Co-occurrence, Clusters, and Burst analysis showed that “inflammation,” “monocyte,” “NF kappa B,” “mechanism,” and “foam cell” appeared with the highest frequency in studies. “Oxidative stress,” “coronary heart disease,” and “prevention” were the strongest citation burst keywords from 2019 to 2021.
Collapse
Affiliation(s)
- Luxia Song
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Ma
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yixuan Fan
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Runmin Lai
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wende Tian
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zihao Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianqing Ju
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Xu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Hao Xu,
| |
Collapse
|
17
|
Chen D, Yu C, Ying Y, Luo Y, Ren L, Zhu C, Yang K, Wu B, Liu Q. Study of the Osteoimmunomodulatory Properties of Curcumin-Modified Copper-Bearing Titanium. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103205. [PMID: 35630685 PMCID: PMC9144993 DOI: 10.3390/molecules27103205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/07/2022] [Accepted: 05/15/2022] [Indexed: 11/30/2022]
Abstract
Peri-implantitis can lead to implant failure. In this study, curcumin (CUR) was modified onto the copper-bearing titanium alloy (Cu-Ti) with the assistance of polydopamine (PDA) in order to study the bone immune response and subsequent osteogenesis. FE-SEM, XPS and water contact angle were utilized to characterize the coating surface. Bone marrow mesenchymal stem cells (BMSCs) and macrophages were cultured separately and together onto the CUR modified Cu-Ti. Cell activity, expression of relative genes and proteins, cell migration ability, and fluorescence staining of cells were performed. CUR modification slightly increased the activation of M1-type and M2-type cells under physiological conditions. In the inflammation state, CUR inhibited the overexpression of M1 macrophages and induced M2-type differentiation. In addition, the modification itself could provoke the expression of osteoblastic-related genes of BMSCs, while promoting the osteogenic differentiation of BMSCs through the activation of macrophages in both physiological and inflammatory states. The BMSCs migration was increased, the expression of osteogenic-related genes and proteins was up-regulated, and alkaline phosphatase activity (ALP) was increased. Thus, the modification of CUR can promote the osteointegration effect of Cu-Ti by bone immunomodulation and may, in addition, improve the success rate of implants.
Collapse
Affiliation(s)
- Danhong Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China; (D.C.); (C.Y.); (Y.Y.)
- College of Stomatology, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China
| | - Chengcheng Yu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China; (D.C.); (C.Y.); (Y.Y.)
- College of Stomatology, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China
| | - Ying Ying
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China; (D.C.); (C.Y.); (Y.Y.)
- College of Stomatology, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China
| | - Yuanyi Luo
- Institute of Low-dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China; (Y.L.); (C.Z.)
| | - Ling Ren
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China; (L.R.); (K.Y.)
| | - Caizhen Zhu
- Institute of Low-dimensional Materials Genome Initiative, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China; (Y.L.); (C.Z.)
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China; (L.R.); (K.Y.)
| | - Buling Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China; (D.C.); (C.Y.); (Y.Y.)
- College of Stomatology, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China
- Correspondence: (Q.L.); (B.W.); Tel.: +86-20-62787153 (Q.L.); +86-20-62787678 (B.W.)
| | - Qi Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China; (D.C.); (C.Y.); (Y.Y.)
- College of Stomatology, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou 510515, China
- Correspondence: (Q.L.); (B.W.); Tel.: +86-20-62787153 (Q.L.); +86-20-62787678 (B.W.)
| |
Collapse
|
18
|
Sottero B, Testa G, Gamba P, Staurenghi E, Giannelli S, Leonarduzzi G. Macrophage polarization by potential nutraceutical compounds: A strategic approach to counteract inflammation in atherosclerosis. Free Radic Biol Med 2022; 181:251-269. [PMID: 35158030 DOI: 10.1016/j.freeradbiomed.2022.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022]
Abstract
Chronic inflammation represents a main event in the onset and progression of atherosclerosis and is closely associated with oxidative stress in a sort of vicious circle that amplifies and sustains all stages of the disease. Key players of atherosclerosis are monocytes/macrophages. According to their pro- or anti-inflammatory phenotype and biological functions, lesional macrophages can release various mediators and enzymes, which in turn contribute to plaque progression and destabilization or, alternatively, lead to its resolution. Among the factors connected to atherosclerotic disease, lipid species carried by low density lipoproteins and pro-oxidant stimuli strongly promote inflammatory events in the vasculature, also by modulating the macrophage phenotyping. Therapies specifically aimed to balance macrophage inflammatory state are increasingly considered as powerful tools to counteract plaque formation and destabilization. In this connection, several molecules of natural origin have been recognized to be active mediators of diverse metabolic and signaling pathways regulating lipid homeostasis, redox state, and inflammation; they are, thus, considered as promising candidates to modulate macrophage responsiveness to pro-atherogenic stimuli. The current knowledge of the capability of nutraceuticals to target macrophage polarization and to counteract atherosclerotic lesion progression, based mainly on in vitro investigation, is summarized in the present review.
Collapse
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy.
| |
Collapse
|
19
|
Hastreiter AA, Paredes LC, Saraiva Camara NO. Metabolic requirement for macrophages. MACROPHAGES IN THE HUMAN BODY 2022:49-66. [DOI: 10.1016/b978-0-12-821385-8.00010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Xu Z, Deng B, Wang X, Yu J, Xu Z, Liu P, Liu C, Cai Y, Wang F, Zong R, Chen Z, Xing H, Chen G. Nanofiber-mediated sequential photothermal antibacteria and macrophage polarization for healing MRSA-infected diabetic wounds. J Nanobiotechnology 2021; 19:404. [PMID: 34865643 PMCID: PMC8647563 DOI: 10.1186/s12951-021-01152-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/19/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Diabetic wound healing remains a challenge because of its susceptibility to drug-resistant bacterial infection and its persistent proinflammatory state. Switching from proinflammatory M1 macrophages (Mφs) to proregenerative M2 dominant Mφs in a timely manner accelerates wound healing by coordinating inflammatory, proliferative, and angiogenic processes. METHODS We propose a sequential photothermal antibacterial and subsequent M2 Mφ polarization strategy based on nanofibers (NFs) consisting of polydopamine (PDA) coating on curcumin (Cur) nanocrystals to treat Methicillin-resistant Staphylococcus aureus (MRSA)-infected diabetic wounds. RESULTS The PDA/Cur NFs showed excellent photothermal conversion and antibacterial effects due to the PDA shell under laser irradiation, consequently resulting in the release of the inner Cur with the ability to promote cell proliferation and reinforce the M2 Mφ phenotype in vitro. In vivo studies on MRSA-infected diabetic wounds showed that PDA/Cur NFs not only inhibited MRSA infection but also accelerated the wound regeneration process. Furthermore, the NFs displayed the ability to promote the M2 Mφ phenotype with enhanced collagen deposition, angiogenesis, and cell proliferation. CONCLUSION Overall, the NFs displayed great potential as promising therapeutics for healing infected diabetic wounds through a sequential photothermal antibacterial and M2 Mφ polarization strategy.
Collapse
Affiliation(s)
- Zhou Xu
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, China
| | - Xuewen Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Jie Yu
- Department of Traditional Chinese Medicine, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, China
| | - Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Penggang Liu
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Caihong Liu
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Yuan Cai
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Fei Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Rongling Zong
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Zhiling Chen
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Hua Xing
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Gang Chen
- Institute of Comparative Medicine, College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.
- School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, 221004, China.
| |
Collapse
|
21
|
Wang D, Jiang S, Zhang F, Ma S, Heng BC, Wang Y, Zhu J, Xu M, He Y, Wei Y, Zhang X, Xia B, Deng X. Cell Membrane Vesicles with Enriched CXCR4 Display Enhances Their Targeted Delivery as Drug Carriers to Inflammatory Sites. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101562. [PMID: 34687286 PMCID: PMC8655180 DOI: 10.1002/advs.202101562] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/02/2021] [Indexed: 06/13/2023]
Abstract
Cell membrane vesicles (CMVs) are composed of natural cell membranes which makes them effective drug delivery systems with low immunogenicity and prolonged circulation time. However, targeting delivery of CMVs in vivo for clinical applications is still a major challenge. In this study, CXCR4 recombinant lentivirus is transfected into MC-3T3 cells and membrane CXCR4-enriched MC-3T3 cells are obtained. CMVs with enriched membrane CXCR4 display (CXCR4-CMVs) are obtained from the transfected MC-3T3 cells. Curcumin, an effective natural anti-inflammatory compound, is encapsulated into CXCR4-CMVs through physical entrapment (CXCR4/Cur-CMVs), with the membrane integrity of CXCR4/Cur-CMVs being well-preserved. CXCR4/Cur-CMVs induce enhanced M2 macrophage polarization, exhibit anti-inflammatory effects, and significantly improve homing via the CXCR4/CXCL12 axis in vitro. Utilizing ulcerative colitis and apical periodontitis as inflammatory disease models, it is found that CXCR4/Cur-CMVs are obviously aggregated within inflammatory areas after intravenous administration, which results in significant amelioration of ulcerative colitis and apical periodontitis. Therefore, this research may provide a feasible and innovative approach for fabricating an inflammatory site-targeting delivery system, by engineering CMVs to increase membrane-presenting CXCR4 receptor.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing, 100081, P. R. China
| | - Shengjie Jiang
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Fengyi Zhang
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Siqin Ma
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Boon Chin Heng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yuanyuan Wang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing, 100081, P. R. China
| | - Junxia Zhu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing, 100081, P. R. China
| | - Mingming Xu
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Ying He
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yan Wei
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Bin Xia
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Peking University, Beijing, 100081, P. R. China
| | - Xuliang Deng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| |
Collapse
|
22
|
He K, Luo X, Wen M, Wang C, Qin C, Shao J, Gan L, Dong R, Jiang H. Effect of acute ammonia toxicity on inflammation, oxidative stress and apoptosis in head kidney macrophage of Pelteobagrus fulvidraco and the alleviation of curcumin. Comp Biochem Physiol C Toxicol Pharmacol 2021; 248:109098. [PMID: 34139380 DOI: 10.1016/j.cbpc.2021.109098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 12/18/2022]
Abstract
Ammonia is one of the most major pollutant and stress factors of aquaculture systems, and has seriously endangered fish health. However, few studies have been performed on mechanisms of the detrimental impact of ammonia stress and mitigation in fish. A study was carried out to investigate the response of genes involved in inflammation, antioxidation, polarization and apoptosis in head kidney macrophages to acute ammonia toxicity, and the alleviation effect of curcumin. The cells were divided into six groups, as follows: The control group composed of untreated macrophages (CON), the experimental groups, consisting of macrophages treated with 0.23 mg L-1 ammonia (AM), 45 μmol L-1 curcumin (CUR), 0.23 mg L-1 ammonia and 5 μmol L-1 curcumin (5A), 0.23 mg L-1 ammonia and 25 μmol L-1 curcumin (25A), 0.23 mg L-1 ammonia and 45 μmol L-1 curcumin (45A). The cells were pretreated with different concentrations of curcumin for 1 h and then incubated with ammonia for 24 h. The results showed that ammonia poisoning could increase ROS levels, up-regulate the expression of antioxidant enzymes (SOD and GPx), inflammatory cytokines (IL-1, IL-6 and TNF-α) and inflammatory mediators (NF-κB p65 and COX-2), decrease cell viability, down-regulate the expression of M2 marker (Arg-1) and anti-apoptosis (Bcl-2), but curcumin could alleviate the adverse effect of ammonia toxicity. Overall, these results have important implications for understanding of the mechanism of ammonia toxicity and the mitigating effect of curcumin in fish.
Collapse
Affiliation(s)
- Kewei He
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Xueping Luo
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Ming Wen
- College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory for Animal Diseases and Veterinary Public Health of Guizhou Province, Guiyang 550025, China.
| | - Changan Wang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China.
| | - Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of theYangtze River, Neijiang Normal University, Neijiang 641100, China.
| | - Jian Shao
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Lei Gan
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Ranran Dong
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Haibo Jiang
- Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China; Key Laboratory for Animal Diseases and Veterinary Public Health of Guizhou Province, Guiyang 550025, China.
| |
Collapse
|
23
|
Vahdat-Lasemi F, Aghaee-Bakhtiari SH, Tasbandi A, Jaafari MR, Sahebkar A. Targeting interleukin-β by plant-derived natural products: Implications for the treatment of atherosclerotic cardiovascular disease. Phytother Res 2021; 35:5596-5622. [PMID: 34390063 DOI: 10.1002/ptr.7194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/21/2021] [Accepted: 05/29/2021] [Indexed: 01/31/2023]
Abstract
Inflammation is the main contributing factor to atheroma formation in atherosclerosis. Interleukin-1 beta (IL-1β) is an inflammatory mediator found in endothelial cells and resident leukocytes. Canakinumab is a selective monoclonal antibody against IL-1β which attenuates inflammation and concurrently precipitates fatal infections and sepsis. Natural products derived from medicinal plants, herbal remedy and functional foods are widely used nowadays. Experimental and clinical trial evidence supports that some natural products such as curcumin, resveratrol, and quercetin have potential effects on IL-1β suppression. In this review, we tried to document findings that used medicinal plants and plant-based natural products for treating atherosclerosis and its related diseases through the suppression of IL-1β.
Collapse
Affiliation(s)
- Fatemeh Vahdat-Lasemi
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Aida Tasbandi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia
| |
Collapse
|
24
|
Chang Z, Wang Y, Liu C, Smith W, Kong L. Natural Products for Regulating Macrophages M2 Polarization. Curr Stem Cell Res Ther 2021; 15:559-569. [PMID: 31120001 DOI: 10.2174/1574888x14666190523093535] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/23/2019] [Accepted: 04/26/2019] [Indexed: 02/06/2023]
Abstract
Macrophages M2 polarization have been taken as an anti-inflammatory progression during inflammation. Natural plant-derived products, with potential therapeutic and preventive activities against inflammatory diseases, have received increasing attention in recent years because of their whole regulative effects and specific pharmacological activities. However, the molecular mechanisms about how different kinds of natural compounds regulate macrophages polarization still unclear. Therefore, in the current review, we summarized the detailed research progress on the active compounds derived from herbal plants with regulating effects on macrophages, especially M2 polarization. These natural occurring compounds including flavonoids, terpenoids, glycosides, lignans, coumarins, alkaloids, polyphenols and quinones. In addition, we extensively discussed the cellular mechanisms underlying the M2 polarization for each compound, which could provide potential therapeutic strategies aiming macrophages M2 polarization.
Collapse
Affiliation(s)
- Zhen Chang
- Department of Spine Surgery, Honghui-hospital, Xi'an Jiaotong University, School of Medicine, Xi'an, China
| | - Youhan Wang
- Department of Spine Surgery, Honghui-hospital, Xi'an Jiaotong University, School of Medicine, Xi'an, China.,Shaanxi University of Chinese Medicine, Xian Yang, China
| | - Chang Liu
- Department of Spine Surgery, Honghui-hospital, Xi'an Jiaotong University, School of Medicine, Xi'an, China.,Shaanxi University of Chinese Medicine, Xian Yang, China
| | - Wanli Smith
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lingbo Kong
- Department of Spine Surgery, Honghui-hospital, Xi'an Jiaotong University, School of Medicine, Xi'an, China
| |
Collapse
|
25
|
Singh L, Sharma S, Xu S, Tewari D, Fang J. Curcumin as a Natural Remedy for Atherosclerosis: A Pharmacological Review. Molecules 2021; 26:molecules26134036. [PMID: 34279384 PMCID: PMC8272048 DOI: 10.3390/molecules26134036] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 01/08/2023] Open
Abstract
Curcumin, a natural polyphenolic compound present in Curcuma longa L. rhizomes, shows potent antioxidant, anti-inflammatory, anti-cancer, and anti-atherosclerotic properties. Atherosclerosis is a comprehensive term for a series of degenerative and hyperplasic lesions such as thickening or sclerosis in large- and medium-sized arteries, causing decreased vascular-wall elasticity and lumen diameter. Atherosclerotic cerebro-cardiovascular disease has become a major concern for human health in recent years due to its clinical sequalae of strokes and heart attacks. Curcumin concoction treatment modulates several important signaling pathways related to cellular migration, proliferation, cholesterol homeostasis, inflammation, and gene transcription, among other relevant actions. Here, we provide an overview of curcumin in atherosclerosis prevention and disclose the underlying mechanisms of action of its anti-atherosclerotic effects.
Collapse
Affiliation(s)
- Laxman Singh
- Centre of Biodiversity Conservation & Management, G.B.Pant National Institute of Himalayan Environment, Almora 263643, Uttarakhand, India;
| | - Shikha Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Suowen Xu
- Department of Endocrinology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230037, China
- Correspondence: (S.X.); (D.T.); (J.F.)
| | - Devesh Tewari
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
- Correspondence: (S.X.); (D.T.); (J.F.)
| | - Jian Fang
- Department of Pharmacy, Huadu District People’s Hospital, Southern Medical University, Guangzhou 510800, China
- Correspondence: (S.X.); (D.T.); (J.F.)
| |
Collapse
|
26
|
Sepand MR, Aghsami M, Keshvadi MH, Bigdelou B, Behzad R, Zanganeh S, Shadboorestan A. The role of macrophage polarization and function in environmental toxicant-induced cancers. ENVIRONMENTAL RESEARCH 2021; 196:110933. [PMID: 33689818 DOI: 10.1016/j.envres.2021.110933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/10/2021] [Accepted: 02/20/2021] [Indexed: 06/12/2023]
Abstract
Macrophages are a critical member of the innate immune system and can intensify tumor invasiveness and assist the growth of neoplastic cells. Moreover, they have the capability to reinforce immunosuppression and angiogenesis. Various investigations suggest that health-related issues, including inflammatory disorders and neoplastic diseases may be caused by environmental toxicant exposure. However, it is still unclear what role these environmental toxicants play in causing carcinogenesis by disturbing the mechanisms of migration, polarization, differentiation, and immune-stimulatory functions of macrophages. Accordingly, in this article, we will explore the interaction between environmental chemicals and inflammatory macrophage processes at the molecular level and their association with tumor progression and carcinogenesis.
Collapse
Affiliation(s)
- Mohammad Reza Sepand
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, Dartmouth, MA, 02747, USA
| | - Mehdi Aghsami
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Keshvadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Banafsheh Bigdelou
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, Dartmouth, MA, 02747, USA
| | - Ramina Behzad
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, Dartmouth, MA, 02747, USA
| | - Steven Zanganeh
- Department of Bioengineering, University of Massachusetts Dartmouth, 285 Old Westport Road, Dartmouth, MA, 02747, USA.
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
27
|
Chen S, Liang H, Ji Y, Kou H, Zhang C, Shang G, Shang C, Song Z, Yang L, Liu L, Wang Y, Liu H. Curcumin Modulates the Crosstalk Between Macrophages and Bone Mesenchymal Stem Cells to Ameliorate Osteogenesis. Front Cell Dev Biol 2021; 9:634650. [PMID: 33634135 PMCID: PMC7900185 DOI: 10.3389/fcell.2021.634650] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 01/08/2023] Open
Abstract
Bone healing is thought to be influenced by the cross-talk between bone forming and immune cells. In particular, macrophages play a crucial role in the regulation of osteogenesis. Curcumin, the major bioactive polyphenolic ingredient of turmeric, has been shown to regulate inflammatory response and osteogenic activities. However, whether curcumin could regulate macrophage polarization and subsequently influence osteogenesis remain to be elucidated. In this study, the potential immunomodulatory capability of curcumin on inflammatory response and phenotype switch of macrophages and the subsequent impact on osteogenic differentiation of MSCs are investigated. We demonstrated that curcumin exhibited significant anti-inflammatory effect by polarizing the macrophages toward anti-inflammatory phenotype, with increased expression of IL-4, IL-10, and CD206, and decreased expression of IL-1β, TNF-α, CCR7, and iNOS. In addition, curcumin could improve the osteo-immune microenvironment via promoting osteogenesis-related regenerative cytokine BMP-2 and TGF-β production. Moreover, the co-cultured test of macrophages and BMSCs showed that curcumin-modulated macrophages conditioned medium could promote osteogenic differentiation of BMSCs with increased gene (ALP, Runx-2, OCN, and OPN) and protein (Runx-2 and OCN) expression levels, enhanced ALP activity, and obvious formation of mineralized nodules. Taken together, with the interaction between curcumin-conditioned macrophage and curcumin-stimulated BMSCs, curcumin could remarkably enhance the osteogenic differentiation of BMSCs in LPS-activated inflammatory macrophage-BMSCs coculture system.
Collapse
Affiliation(s)
- Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hang Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanhui Ji
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongwei Kou
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Shang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunfeng Shang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zongmian Song
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Yang
- Department of Paediatrics, The Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lei Liu
- Department of Paediatrics, The Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yongkui Wang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Qing J, Zhang Z, Novák P, Zhao G, Yin K. Mitochondrial metabolism in regulating macrophage polarization: an emerging regulator of metabolic inflammatory diseases. Acta Biochim Biophys Sin (Shanghai) 2020; 52:917-926. [PMID: 32785581 DOI: 10.1093/abbs/gmaa081] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Indexed: 12/24/2022] Open
Abstract
As a major type of immune cells with heterogeneity and plasticity, macrophages are classically divided into inflammatory (M1) and alternative/anti-inflammatory (M2) types and play a crucial role in the progress of the inflammatory diseases. Recent studies have shown that metabolism is an important determinant of macrophage phenotype. Mitochondria, one of the most important compartments involving cell metabolism, are closely associated with the regulation of cell functions. In most types of cell, mitochondrial oxidative phosphorylation (OXPHOS) is the primary mode of cellular energy production. However, mitochondrial OXPHOS is inhibited in activated M1 macrophages, rendering them unable to be converted into M2 phenotype. Thus, mitochondrial metabolism is a crucial regulator in macrophage functions. This review summarizes the roles of mitochondria in macrophage polarization and analyzes the molecular mechanisms underlying mitochondrial metabolism and function, which may provide new approaches for the treatment of metabolic inflammatory diseases.
Collapse
Affiliation(s)
- Jina Qing
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
- Research Lab of translational medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Zizhen Zhang
- School of Medicine, Hunan Polytechnic of Environment and Biology, Hengyang 421001, China
| | - Petr Novák
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Guojun Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan City People's Hospital, Qingyuan 511518, China
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
- Research Lab of translational medicine, Hengyang Medical College, University of South China, Hengyang 421001, China
| |
Collapse
|
29
|
Application of film-forming solution as a transdermal delivery system of piperine-rich herbal mixture extract for anti-inflammation. Heliyon 2020; 6:e04139. [PMID: 32551384 PMCID: PMC7292918 DOI: 10.1016/j.heliyon.2020.e04139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/05/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022] Open
Abstract
Piperine-rich herbal mixture (PHM) used in this study is a traditional Thai medicine that contains 21 oriental herbs. It is called "Sahastara remedy" and is officially included in the Thai National List of Essential Medicine since A.D. 2011. PHM has been used orally to relieve muscle and bone pains. It contains Piper nigrum fruits as a major constituent and also Piper retrofractum fruits, PHM thus has anti-inflammatory activities that mostly come from the bioactivities of piperine consisting of these pepper fruits. Unfortunately, PHM usually causes gastrointestinal side effects. Consequently, a topical product containing an alcoholic extract of PHM (PHM-E), i.e., film-forming solution (FFS) was developed to overcome this drawback. The aims of this study were to investigate the anti-inflammatory activity of PHM-E, to evaluate physicochemical properties and the anti-inflammatory activity of FFS containing PHM-E (PHM-E FFS). Anti-inflammatory activities of PHM-E were investigated in the RAW 264.7 cells. Physicochemical properties, in vitro toxicities and anti-inflammatory activities of PHM-E FFS including its dry film (PHM-E film) were determined. PHM-E showed anti-inflammatory activities with dose dependent manners via inhibition of nitric oxide and prostaglandin E2 production by the RAW 264.7 cells and promotion of the cell phenotype polarization from M1 to M2. PHM-E FFS had low viscosity and exhibited the Newtonian behavior. It provided elastic PHM-E film with low tensile strength. The release profile of piperine from PHM-E film followed a zero-kinetic model. PHM-E FFS demonstrated compatibility with the skin cells, minimal ocular irritant when accidentally splashing into the eye and moderate-to-high potency for inhibition of inflammatory symptoms in the rats. PHM-E FFS thus had potential for use in the further clinical study to investigate its efficacy and safety in patients.
Collapse
|
30
|
Phenolic Compounds Exerting Lipid-Regulatory, Anti-Inflammatory and Epigenetic Effects as Complementary Treatments in Cardiovascular Diseases. Biomolecules 2020; 10:biom10040641. [PMID: 32326376 PMCID: PMC7226566 DOI: 10.3390/biom10040641] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is the main process behind cardiovascular diseases (CVD), maladies which continue to be responsible for up to 70% of death worldwide. Despite the ongoing development of new and potent drugs, their incomplete efficacy, partial intolerance and numerous side effects make the search for new alternatives worthwhile. The focus of the scientific world turned to the potential of natural active compounds to prevent and treat CVD. Essential for effective prevention or treatment based on phytochemicals is to know their mechanisms of action according to their bioavailability and dosage. The present review is focused on the latest data about phenolic compounds and aims to collect and correlate the reliable existing knowledge concerning their molecular mechanisms of action to counteract important risk factors that contribute to the initiation and development of atherosclerosis: dyslipidemia, and oxidative and inflammatory-stress. The selection of phenolic compounds was made to prove their multiple benefic effects and endorse them as CVD remedies, complementary to allopathic drugs. The review also highlights some aspects that still need clear scientific explanations and draws up some new molecular approaches to validate phenolic compounds for CVD complementary therapy in the near future.
Collapse
|
31
|
Davoodvandi A, Sahebnasagh R, Mardanshah O, Asemi Z, Nejati M, Shahrzad MK, Mirzaei HR, Mirzaei H. Medicinal Plants As Natural Polarizers of Macrophages: Phytochemicals and Pharmacological Effects. Curr Pharm Des 2019; 25:3225-3238. [DOI: 10.2174/1381612825666190829154934] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022]
Abstract
Macrophages are one of the crucial mediators of the immune response in different physiological and
pathological conditions. These cells have critical functions in the inflammation mechanisms that are involved in
the inhibition or progression of a wide range of diseases including cancer, autoimmune diseases, etc. It has been
shown that macrophages are generally divided into two subtypes, M1 and M2, which are distinguished on the
basis of their different gene expression patterns and phenotype. M1 macrophages are known as pro-inflammatory
cells and are involved in inflammatory mechanisms, whereas M2 macrophages are known as anti-inflammatory
cells that are involved in the inhibition of the inflammatory pathways. M2 macrophages help in tissue healing via
producing anti-inflammatory cytokines. Increasing evidence indicated that the appearance of different macrophage
subtypes is associated with the fate of diseases (progression versus suppression). Hence, polarization of
macrophages can be introduced as an important venue in finding, designing and developing novel therapeutic
approaches. Albeit, there are different pharmacological agents that are used for the treatment of various disorders,
it has been shown that several natural compounds have the potential to regulate M1 to M2 macrophage polarization
and vice versa. Herein, for the first time, we summarized new insights into the pharmacological effects of
natural compounds on macrophage polarization.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Roxana Sahebnasagh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Mardanshah
- Department of Laboratory Sciences, Sirjan Faculty of Medical Sciences, Sirjan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Majid Nejati
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad K. Shahrzad
- Department of Internal Medicine and Endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid R. Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
32
|
Han C, Yang J, Song P, Wang X, Shi W. Effects of Salvia miltiorrhiza Polysaccharides on Lipopolysaccharide-Induced Inflammatory Factor Release in RAW264.7 Cells. J Interferon Cytokine Res 2019; 38:29-37. [PMID: 29328882 DOI: 10.1089/jir.2017.0087] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This study investigated the anti-inflammatory effects and possible underlying mechanisms of Salvia miltiorrhiza polysaccharides (SMP) in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. The cytotoxicity of SMP was detected by the MTT method. The morphological change of RAW264.7 was observed by Diff-Quik staining. Enzyme-linked immunosorbent assay was used to evaluate the production of cytokines in LPS-induced RAW264.7 cells. The nitric oxide (NO) kit assay detected the NO release from LPS-induced RAW264.7 cells. Real-time polymerase chain reaction was used to detect the transcriptions of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), inducible NO synthase (iNOS), and cyclooxygenase (COX)-2 in LPS-induced RAW264.7 cells. The protein expression of nuclear NF-κB was measured by Western blot. The results showed that the safe medication range of SMP was less than 3 mg/mL. Compared with the LPS model group, SMP (2, 1, and 0.5 mg/mL) improved the degree of cell deformation and reduced the amount of pseudopodia, and statistically reduced the secretions of cytokines in cells induced by LPS (P < 0.01) at different time points. SMP significantly inhibited the mRNA transcriptions of TNF-α, IL-6, iNOS, and COX-2 and the protein expressions of NF-κB, p-p65, and p-IκBa. In conclusion, this study preliminarily proved the protective effect of SMP on LPS-induced RAW264.7 macrophage. Its mechanism might be related to inhibition of NF-κB signal pathway and the gene expressions and secretion of cytokines.
Collapse
Affiliation(s)
- Chao Han
- College of Traditional Chinese Veterinary Medicine, Agricultural University of Hebei , Baoding, China
| | - Jinkai Yang
- College of Traditional Chinese Veterinary Medicine, Agricultural University of Hebei , Baoding, China
| | - Pengyan Song
- College of Traditional Chinese Veterinary Medicine, Agricultural University of Hebei , Baoding, China
| | - Xiao Wang
- College of Traditional Chinese Veterinary Medicine, Agricultural University of Hebei , Baoding, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Agricultural University of Hebei , Baoding, China
| |
Collapse
|
33
|
The role of traditional Chinese medicine in the treatment of atherosclerosis through the regulation of macrophage activity. Biomed Pharmacother 2019; 118:109375. [PMID: 31548175 DOI: 10.1016/j.biopha.2019.109375] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/16/2019] [Accepted: 08/22/2019] [Indexed: 12/27/2022] Open
Abstract
Atherosclerosis (AS) is the main cause of ischemic cardiovascular, cerebrovascular and peripheral vascular diseases. Macrophage activity has been proven to play a critical role during the AS pathological process, which involves the adhesion, aggregation of mononuclear-macrophages, cell differentiation of M1/M2 macrophages as part of complex mechanisms occurring during lipid metabolism, apoptosis, autophagy, inflammation and immune reaction. Therefore, the development of effective AS treatments is likely to target macrophage activity. Certain herbal extracts (such as Salvia miltiorrhiza) have exhibited enormous potential for AS treatment in the past. Here, we aim to provide a summary on the current understanding of the type of action and the underlying target/pathway in macrophage regulation of certain herbal extracts used in Traditional Chinese Medicine for treatment of AS.
Collapse
|
34
|
Mohammadi A, Blesso CN, Barreto GE, Banach M, Majeed M, Sahebkar A. Macrophage plasticity, polarization and function in response to curcumin, a diet-derived polyphenol, as an immunomodulatory agent. J Nutr Biochem 2019; 66:1-16. [PMID: 30660832 DOI: 10.1016/j.jnutbio.2018.12.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/04/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022]
Abstract
Monocytes and macrophages are important cells of the innate immune system that have diverse functions, including defense against invading pathogens, removal of dead cells by phagocytosis, antigen presentation in the context of MHC class I and class II molecules, and production of various pro-inflammatory cytokines and chemokines such as IL-1β, IL-6, TNF-α and MCP-1. In addition, pro-inflammatory (M1) and anti-inflammatory (M2) macrophages clearly play important roles in the progression of several inflammatory diseases. Therefore, therapies that target macrophage polarization and function by either blocking their trafficking to sites of inflammation, or skewing M1 to M2 phenotype polarization may hold clinical promise in several inflammatory diseases. Dietary-derived polyphenols have potent natural anti-oxidative properties. Within this group of polyphenols, curcumin has been shown to suppress macrophage inflammatory responses. Curcumin significantly reduces co-stimulatory molecules and also inhibits MAPK activation and the translocation of NF-κB p65. Curcumin can also polarize/repolarize macrophages toward the M2 phenotype. Curcumin-treated macrophages have been shown to be highly efficient at antigen capture and endocytosis via the mannose receptor. These novel findings provide new perspectives for the understanding of the immunopharmacological role of curcumin, as well as its therapeutic potential for impacting macrophage polarization and function in the context of inflammation-related disease. However, the precise effects of curcumin on the migration, differentiation, polarization and immunostimulatory functions of macrophages remain unknown. Therefore, in this review, we summarized whether curcumin can influence macrophage polarization, surface molecule expression, cytokine and chemokine production and their underlying pathways in the prevention of inflammatory diseases.
Collapse
Affiliation(s)
- Asadollah Mohammadi
- Cellular & Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, University of Western Australia, Perth, Australia.
| |
Collapse
|
35
|
Momtazi-Borojeni AA, Abdollahi E, Nikfar B, Chaichian S, Ekhlasi-Hundrieser M. Curcumin as a potential modulator of M1 and M2 macrophages: new insights in atherosclerosis therapy. Heart Fail Rev 2019; 24:399-409. [DOI: 10.1007/s10741-018-09764-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
36
|
Fuhrmann G, Chandrawati R, Parmar PA, Keane TJ, Maynard SA, Bertazzo S, Stevens MM. Engineering Extracellular Vesicles with the Tools of Enzyme Prodrug Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706616. [PMID: 29473230 PMCID: PMC5901706 DOI: 10.1002/adma.201706616] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/14/2017] [Indexed: 05/26/2023]
Abstract
Extracellular vesicles (EVs) have recently gained significant attention as important mediators of intercellular communication, potential drug carriers, and disease biomarkers. These natural cell-derived nanoparticles are postulated to be biocompatible, stable under physiological conditions, and to show reduced immunogenicity as compared to other synthetic nanoparticles. Although initial clinical trials are ongoing, the use of EVs for therapeutic applications may be limited due to undesired off-target activity and potential "dilution effects" upon systemic administration which may affect their ability to reach their target tissues. To fully exploit their therapeutic potential, EVs are embedded into implantable biomaterials designed to achieve local delivery of therapeutics taking advantage of enzyme prodrug therapy (EPT). In this first application of EVs for an EPT approach, EVs are used as smart carriers for stabilizing enzymes in a hydrogel for local controlled conversion of benign prodrugs to active antiinflammatory compounds. It is shown that the natural EVs' antiinflammatory potential is comparable or superior to synthetic carriers, in particular upon repeated long-term incubations and in different macrophage models of inflammation. Moreover, density-dependent color scanning electron microscopy imaging of EVs in a hydrogel is presented herein, an impactful tool for further understanding EVs in biological settings.
Collapse
Affiliation(s)
- Gregor Fuhrmann
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Biogenic Nanotherapeutics Group, Campus E8.1, 66123, Saarbrücken, Germany
| | - Rona Chandrawati
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Paresh A Parmar
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Timothy J Keane
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Stephanie A Maynard
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering, University College London, Malet Place Engineering Building, London, WC1E 6BT, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
37
|
Song Z, Revelo X, Shao W, Tian L, Zeng K, Lei H, Sun HS, Woo M, Winer D, Jin T. Dietary Curcumin Intervention Targets Mouse White Adipose Tissue Inflammation and Brown Adipose Tissue UCP1 Expression. Obesity (Silver Spring) 2018; 26:547-558. [PMID: 29405636 DOI: 10.1002/oby.22110] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 12/01/2017] [Accepted: 12/06/2017] [Indexed: 12/27/2022]
Abstract
OBJECTIVE This study aimed to determine whether dietary curcumin intervention targets both white adipose tissue (WAT) inflammation and brown adipose tissue (BAT)-mediated energy expenditure. METHODS C57BL/6J mice were fed with a low-fat diet, high-fat diet (HFD), or HFD plus curcumin. In addition to assessing the effect of curcumin intervention on metabolic profiles, this study assessed WAT macrophage infiltration and composition and inflammatory cytokine production. Metabolic cages were applied for determining energy expenditure. Raw264.7 (ATCC, Manassas, Virginia) and other cell models were utilized to test the in vitro effect of curcumin treatment. RESULTS Curcumin intervention reduced WAT macrophage infiltration and altered macrophage functional polarity, as the ratio of M2-like versus M1-like macrophages increased after curcumin intervention. Curcumin treatment reduced M1-like macrophage markers or proinflammation cytokine expression in both macrophages and adipocytes. Curcumin intervention also increased energy expenditure and body temperature in response to a cold challenge. Finally, the in vivo and in vitro investigations suggested that curcumin increased expression of uncoupling protein 1 (UCP1), possibly involving PPAR-dependent and -independent mechanisms. CONCLUSIONS Curcumin intervention targets both WAT inflammation and BAT UCP1 expression. These observations advanced our knowledge on the metabolic beneficial effects of the curry compound curcumin, bringing us a novel perspective on dietary polyphenol research.
Collapse
Affiliation(s)
- Zhuolun Song
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xavier Revelo
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Weijuan Shao
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lili Tian
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kejing Zeng
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Helena Lei
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Minna Woo
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Daniel Winer
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Bisdemethoxycurcumin and Its Cyclized Pyrazole Analogue Differentially Disrupt Lipopolysaccharide Signalling in Human Monocyte-Derived Macrophages. Mediators Inflamm 2018; 2018:2868702. [PMID: 29576743 PMCID: PMC5822910 DOI: 10.1155/2018/2868702] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/31/2017] [Accepted: 11/29/2017] [Indexed: 02/06/2023] Open
Abstract
Several studies suggest that curcumin and related compounds possess antioxidant and anti-inflammatory properties including modulation of lipopolysaccharide- (LPS-) mediated signalling in macrophage cell models. We here investigated the effects of curcumin and the two structurally unrelated analogues GG6 and GG9 in primary human blood-derived macrophages as well as the signalling pathways involved. Macrophages differentiated from peripheral blood monocytes for 7 days were activated with LPS or selective Toll-like receptor agonists for 24 h. The effects of test compounds on cytokine production and immunophenotypes evaluated as CD80+/CCR2+ and CD206+/CD163+ subsets were examined by ELISA and flow cytometry. Signalling pathways were probed by Western blot. Curcumin (2.5–10 μM) failed to suppress LPS-induced inflammatory responses. While GG6 reduced LPS-induced IκB-α degradation and showed a trend towards reduced interleukin-1β release, GG9 prevented the increase in proinflammatory CD80+ macrophage subset, downregulation of the anti-inflammatory CD206+/CD163+ subset, increase in p38 phosphorylation, and increase in cell-bound and secreted interleukin-1β stimulated by LPS, at least in part through signalling pathways not involving Toll-like receptor 4 and nuclear factor-κB. Thus, the curcumin analogue GG9 attenuated the LPS-induced inflammatory response in human blood-derived macrophages and may therefore represent an attractive chemical template for macrophage pharmacological targeting.
Collapse
|
39
|
Farajzadeh R, Zarghami N, Serati-Nouri H, Momeni-Javid Z, Farajzadeh T, Jalilzadeh-Tabrizi S, Sadeghi-Soureh S, Naseri N, Pilehvar-Soltanahmadi Y. Macrophage repolarization using CD44-targeting hyaluronic acid–polylactide nanoparticles containing curcumin. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:2013-2021. [DOI: 10.1080/21691401.2017.1408116] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Raana Farajzadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Hamed Serati-Nouri
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Momeni-Javid
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Taher Farajzadeh
- Department of Microbiology, Zanjan Basic Sciences and Medicine Branch, Islamic Azad University, Zanjan, Iran
| | - Sepideh Jalilzadeh-Tabrizi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Shima Sadeghi-Soureh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Neda Naseri
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences, Tehran, Iran
| | - Younes Pilehvar-Soltanahmadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
40
|
Effect of luteolin on inflammatory responses in RAW264.7 macrophages activated with LPS and IFN-γ. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.02.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
41
|
Li B, Hu Y, Zhao Y, Cheng M, Qin H, Cheng T, Wang Q, Peng X, Zhang X. Curcumin Attenuates Titanium Particle-Induced Inflammation by Regulating Macrophage Polarization In Vitro and In Vivo. Front Immunol 2017; 8:55. [PMID: 28197150 PMCID: PMC5281580 DOI: 10.3389/fimmu.2017.00055] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/12/2017] [Indexed: 01/31/2023] Open
Abstract
Periprosthetic inflammatory osteolysis and subsequent aseptic loosening are commonly observed in total joint arthroplasty. Other than revision surgery, few approved treatments are available for this complication. Wear particle-induced inflammation and macrophage polarization state play critical roles in periprosthetic osteolysis. We investigated the effects of curcumin, a polyphenol extracted from Curcuma longa, on titanium (Ti) particle-induced inflammation and macrophage polarization in vitro using the murine cell line RAW 264.7 and in vivo using a murine air pouch model. The expression of specific macrophage markers was qualitatively analyzed by immunofluorescence (inducible nitric oxide synthase and CD206) and quantitatively analyzed by flow cytometry (CCR7 and CD206), representing M1 and M2 macrophages, respectively. Our results show that curcumin induced a higher percentage of M2 macrophages together with a higher concentration of anti-inflammatory cytokine IL-10, and a lower percentage of M1 macrophages with a lower concentration of pro-inflammatory cytokines (TNF-α and IL-6). The genes encoding CD86 (M1) and CD163 (M2), two additional markers, were shifted by curcumin toward an M2 phenotype. C57BL/J6 mice were injected with air and Ti particles to establish an air pouch model. Curcumin reduced cell infiltration in the pouch membrane and decreased membrane thickness. The analysis of exudates obtained from pouches demonstrated that the effects of curcumin on macrophage polarization and cytokine production were similar to those observed in vitro. These results prove that curcumin suppresses Ti particle-induced inflammation by regulating macrophage polarization. Thus, curcumin could be developed as a new therapeutic candidate for the prevention and treatment of inflammatory osteolysis and aseptic loosening.
Collapse
Affiliation(s)
- Bin Li
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Yan Hu
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Yaochao Zhao
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Mengqi Cheng
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Hui Qin
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Tao Cheng
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Qiaojie Wang
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Xiaochun Peng
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Sixth People' Hospital, Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
42
|
Yang Y, Wu BQ, Wang YH, Shi YF, Luo JM, Ba JH, Liu H, Zhang TT. Regulatory effects of miR-155 and miR-146a on repolarization and inflammatory cytokine secretion in human alveolar macrophages in vitro. Immunopharmacol Immunotoxicol 2016; 38:502-509. [DOI: 10.1080/08923973.2016.1248845] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Yang Yang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Ben-Quan Wu
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Yan-Hong Wang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Yun-Feng Shi
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jin-Mei Luo
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jun-Hui Ba
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Hui Liu
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Tian-Tuo Zhang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
43
|
Hoppstädter J, Hachenthal N, Valbuena-Perez JV, Lampe S, Astanina K, Kunze MM, Bruscoli S, Riccardi C, Schmid T, Diesel B, Kiemer AK. Induction of Glucocorticoid-induced Leucine Zipper (GILZ) Contributes to Anti-inflammatory Effects of the Natural Product Curcumin in Macrophages. J Biol Chem 2016; 291:22949-22960. [PMID: 27629417 DOI: 10.1074/jbc.m116.733253] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Indexed: 12/12/2022] Open
Abstract
GILZ (glucocorticoid-induced leucine zipper) is inducible by glucocorticoids and plays a key role in their mode of action. GILZ attenuates inflammation mainly by inhibition of NF-κB and mitogen-activated protein kinase activation but does not seem to be involved in the severe side effects observed after glucocorticoid treatment. Therefore, GILZ might be a promising target for new therapeutic approaches. The present work focuses on the natural product curcumin, which has previously been reported to inhibit NF-κB. GILZ was inducible by curcumin in macrophage cell lines, primary human monocyte-derived macrophages, and murine bone marrow-derived macrophages. The up-regulation of GILZ was neither associated with glucocorticoid receptor activation nor with transcriptional induction or mRNA or protein stabilization but was a result of enhanced translation. Because the GILZ 3'-UTR contains AU-rich elements (AREs), we analyzed the role of the mRNA-binding protein HuR, which has been shown to promote the translation of ARE-containing mRNAs. Our results suggest that curcumin treatment induces HuR expression. An RNA immunoprecipitation assay confirmed that HuR can bind GILZ mRNA. In accordance, HuR overexpression led to increased GILZ protein levels but had no effect on GILZ mRNA expression. Our data employing siRNA in LPS-activated RAW264.7 macrophages show that curcumin facilitates its anti-inflammatory action by induction of GILZ in macrophages. Experiments with LPS-activated bone marrow-derived macrophages from wild-type and GILZ knock-out mice demonstrated that curcumin inhibits the activity of inflammatory regulators, such as NF-κB or ERK, and subsequent TNF-α production via GILZ. In summary, our data indicate that HuR-dependent GILZ induction contributes to the anti-inflammatory properties of curcumin.
Collapse
Affiliation(s)
- Jessica Hoppstädter
- From the Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany
| | - Nina Hachenthal
- From the Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany
| | | | - Sebastian Lampe
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany, and
| | - Ksenia Astanina
- From the Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany
| | - Michael M Kunze
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany, and
| | - Stefano Bruscoli
- Department of Medicine, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Carlo Riccardi
- Department of Medicine, Section of Pharmacology, University of Perugia, 06132 Perugia, Italy
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany, and
| | - Britta Diesel
- From the Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany
| | - Alexandra K Kiemer
- From the Department of Pharmacy, Pharmaceutical Biology, Saarland University, 66041 Saarbrücken, Germany,
| |
Collapse
|
44
|
Curcumin alleviates renal dysfunction and suppresses inflammation by shifting from M1 to M2 macrophage polarization in daunorubicin induced nephrotoxicity in rats. Cytokine 2016; 84:1-9. [DOI: 10.1016/j.cyto.2016.05.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 01/13/2023]
|
45
|
Plants and their active compounds: natural molecules to target angiogenesis. Angiogenesis 2016; 19:287-95. [PMID: 27154020 DOI: 10.1007/s10456-016-9512-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/01/2016] [Indexed: 01/20/2023]
Abstract
Angiogenesis, or new blood vessel formation, is an important process in the pathogenesis of several diseases and thus has been targeted for the prevention and treatment for many disorders. However, the anti-angiogenic agents that are currently in use are mainly synthetic compounds and humanized monoclonal antibodies, which are either expensive or toxic, thereby limiting their use in many patients. Therefore, it is necessary to identify less toxic, inexpensive, novel and effective anti-angiogenic molecules. Several studies have indicated that natural plant products can meet these criteria. In this review, we discuss the anti-angiogenic properties of natural compounds isolated from plants and the molecular mechanisms by which these molecules act. Finally, we summarize the advantages of using plant products as anti-angiogenic agents. Compared with currently available anti-angiogenic drugs, plant products may not only have similar therapeutic potential but are also inexpensive, less toxic, and easy to administer. However, novel and effective strategies are necessary to improve their bioavailability for clinical use.
Collapse
|
46
|
Gao S, Zhou J, Liu N, Wang L, Gao Q, Wu Y, Zhao Q, Liu P, Wang S, Liu Y, Guo N, Shen Y, Wu Y, Yuan Z. Curcumin induces M2 macrophage polarization by secretion IL-4 and/or IL-13. J Mol Cell Cardiol 2015; 85:131-9. [DOI: 10.1016/j.yjmcc.2015.04.025] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 04/03/2015] [Accepted: 04/28/2015] [Indexed: 10/23/2022]
|