1
|
Premarathna AD, Sooäär A, Ahmed TA, Rjabovs V, Hincke MT, Tuvikene R. Isolation, structural characterization and biological activities of polysaccharides from Chondrus crispus. Food Hydrocoll 2024; 154:110131. [DOI: 10.1016/j.foodhyd.2024.110131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Liu Q, Zhao Y, Dong S, Bai X, Chen B, Liu X, Shen J, Zhu D. Characteristics of Neutrophil Migration and Function in Acute Inflammation Induced by Zymosan and Carrageenan in the Mice Air Pouch Model. Inflammation 2024:10.1007/s10753-024-02064-9. [PMID: 38902540 DOI: 10.1007/s10753-024-02064-9] [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: 02/28/2024] [Revised: 04/29/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024]
Abstract
Deciphering the complex and redundant process of acute inflammation remains challenging. The failure of numerous clinical trials assessing anti-inflammation agents which had promising preclinical effects inevitably questions the validity of current animal models of inflammation. This study aimed to better understand the process of immune inflammatory response and to select more suitable models to evaluate the effect of potential anti-inflammatory drugs. Zymosan and λ-carrageenan are the most used representatives of particulate and soluble irritants that trigger acute inflammation in the air pouch inflammation model. When zymosan was used, the number of exudate cells first increased at 4 h-8 h, followed by a drop at 12 h-24 h. While, the changes in number of leukocytes in peripheral blood and proportion of neutrophils in bone marrow have the opposite trend. Meanwhile, neutrophils released neutrophil extracellular traps (NETs) to clean zymosan particles. In contrast, the cell migration response to carrageenan increased during 4 h to 24 h, no obvious NETs were observed, and the number of leukocytes in peripheral blood increased and the proportion of neutrophils in bone marrow decreased slightly. This study indicated that although both zymosan and carrageenan are sterile irritants, the characteristics of the inflammatory response induced by each other were different. In the acute phase of inflammation, zymosan-stimulated neutrophils were mobilized, recruited, and engulfed, and then died by NETs. Carrageenan stimulated the production of cytokines/chemokines by neutrophils or macrophages, but did not lead to an obvious death by releasing NETs.
Collapse
Affiliation(s)
- Qi Liu
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yubo Zhao
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Shuai Dong
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Xingyuan Bai
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Bin Chen
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China
| | - Xijuan Liu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing), Core Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jing Shen
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing), Core Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Dan Zhu
- School of Pharmacy, Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing, 100081, China.
| |
Collapse
|
3
|
Zarkesh I, Movahedi F, Sadeghi-Abandansari H, Pahlavan S, Soleimani M, Baharvand H. ROS scavenging activity of polydopamine nanoparticle-loaded supramolecular gelatin-based hydrogel promoted cardiomyocyte proliferation. Int J Biol Macromol 2024; 259:129228. [PMID: 38184051 DOI: 10.1016/j.ijbiomac.2024.129228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/01/2024] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Reactive oxygen species (ROS) play essential roles in cellular functions, but maintaining ROS balance is crucial for effective therapeutic interventions, especially during cell therapy. In this study, we synthesized an injectable gelatin-based hydrogel, in which polydopamine nanoparticles were entrapped using supramolecular interactions. The surfaces of the nanoparticles were modified using adamantane, enabling their interactions with β-cyclodextrin-conjugated with gelatin. We evaluated the cytotoxicity and antioxidant properties of the hydrogel on neonatal rat cardiomyocytes (NRCM), where it demonstrated the ability to increase the metabolic activity of NRCMs exposed to hydrogen peroxide (H2O2) after 5 days. Hydrogel-entrapped nanoparticle exhibited a high scavenging capability against hydroxyl radical, 1'-diphenyl-2-picrylhydrazyl radicals, and H2O2, surpassing the effectiveness of ascorbic acid solution. Notably, the presence of polydopamine nanoparticles within the hydrogel promoted the proliferation activity of NRCMs, even in the absence of excessive ROS due to H2O2 treatment. Additionally, when the hydrogel with nanoparticles was injected into an air pouch model, it reduced inflammation and infiltration of immune cells. Notably, the levels of anti-inflammatory factors, IL-10 and IL-4, were significantly increased, while the pro-inflammatory factor TNF-α was suppressed. Therefore, this novel ROS-scavenging hydrogel holds promise for both efficient cell delivery into inflamed tissue and promoting tissue repair.
Collapse
Affiliation(s)
- Ibrahim Zarkesh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Cell engineering, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Fatemeh Movahedi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Hamid Sadeghi-Abandansari
- Department of Cell engineering, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran; Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Babol, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Anatomy Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
4
|
da Costa Salomé D, de Freitas RHCN, Fraga CAM, Fernandes PD. Novel Regioisomeric Analogues of Naphthyl- N-Acylhydrazone Derivatives and Their Anti-Inflammatory Effects. Int J Mol Sci 2022; 23:13562. [PMID: 36362349 PMCID: PMC9657883 DOI: 10.3390/ijms232113562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND When homeostasis is disturbed it can result in a pathological event named inflammation. The main drugs used in the treatment consist of non-steroidal and steroidal anti-inflammatory drugs. However, the side effects remain an obstacle during the treatments. In this study, we aimed to evaluate three new regioisomers analogues of naphthyl-N-acylhydrazone derivatives. METHODS Acute models of inflammation in vivo (formalin-induced licking and carrageenan-induced inflammation) as well as in vitro were used to evaluate the effects of LASSBio-2039, LASSBio-2040, and LASSBio-2041. RESULTS All three substances (at 1, 10 or 30 µmol/kg) presented significant effects in the in vivo model reducing leukocyte migration, nitric oxide (NO) and interleukin-1β production. It was observed that only LASSBio-2039 significantly reduced cell migration in vitro. None of the LASSBios affected inducible nitric oxide synthase activity nor presented nitric oxide (NO) scavenger effect. No toxic effect was observed, either in vivo or in vitro. The new regioisomers analogues of naphthyl-N-acylhydrazone derivatives presented significant anti-inflammatory activity, suggesting LASSBio-2039 has a direct effect in leukocytes migratory capacity. CONCLUSIONS Taken together, the data indicate that these substances present promising effects for the development of a prototype for new drugs.
Collapse
Affiliation(s)
- Dayana da Costa Salomé
- Laboratório de Farmacologia da Dor e da Inflamação, Programa de Pesquisa em Descoberta de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Rosana Helena Coimbra Nogueira de Freitas
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Programa de Pesquisa em Descoberta de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Programa de Pesquisa em Descoberta de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Patricia Dias Fernandes
- Laboratório de Farmacologia da Dor e da Inflamação, Programa de Pesquisa em Descoberta de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| |
Collapse
|
5
|
Moniot A, Braux J, Siboni R, Guillaume C, Audonnet S, Allart-Simon I, Sapi J, Tirouvanziam R, Gérard S, Gangloff SC, Velard F. Inhibition of Recruitment and Activation of Neutrophils by Pyridazinone-Scaffold-Based Compounds. Int J Mol Sci 2022; 23:ijms23137226. [PMID: 35806233 PMCID: PMC9266889 DOI: 10.3390/ijms23137226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/07/2022] Open
Abstract
In inflammatory diseases, polymorphonuclear neutrophils (PMNs) are known to produce elevated levels of pro-inflammatory cytokines and proteases. To limit ensuing exacerbated cell responses and tissue damage, novel therapeutic agents are sought. 4aa and 4ba, two pyridazinone-scaffold-based phosphodiesterase-IV inhibitors are compared in vitro to zardaverine for their ability to: (1) modulate production of pro-inflammatory mediators, reactive oxygen species (ROS), and phagocytosis; (2) modulate degranulation by PMNs after transepithelial lung migration. Compound 4ba and zardaverine were tested in vivo for their ability to limit tissue recruitment of PMNs in a murine air pouch model. In vitro treatment of lipopolysaccharide-stimulated PMNs with compounds 4aa and 4ba inhibited the release of interleukin-8, tumor necrosis factor-α, and matrix metalloproteinase-9. PMNs phagocytic ability, but not ROS production, was reduced following treatment. Using a lung inflammation model, we proved that PMNs transmigration led to reduced expression of the CD16 phagocytic receptor, which was significantly blunted after treatment with compound 4ba or zardaverine. Using the murine air pouch model, LPS-induced PMNs recruitment was significantly decreased upon addition of compound 4ba or zardaverine. Our data suggest that new pyridazinone derivatives have therapeutic potential in inflammatory diseases by limiting tissue recruitment and activation of PMNs.
Collapse
Affiliation(s)
- Aurélie Moniot
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Julien Braux
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Renaud Siboni
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Christine Guillaume
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Sandra Audonnet
- Université de Reims Champagne-Ardenne, URCACyt, 51 Rue Cognacq Jay, 51100 Reims, France;
| | - Ingrid Allart-Simon
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Janos Sapi
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, 2015 Uppergate Road, Atlanta, GA 30322, USA
| | - Stéphane Gérard
- Université de Reims Champagne-Ardenne, UMR CNRS 7312 ICMR, 51 Rue Cognacq Jay, 51100 Reims, France; (I.A.-S.); (J.S.); (S.G.)
| | - Sophie C. Gangloff
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
| | - Frédéric Velard
- Université de Reims Champagne-Ardenne, EA 4691 BIOS, 51 Rue Cognacq Jay, 51100 Reims, France; (A.M.); (J.B.); (R.S.); (C.G.); (S.C.G.)
- Correspondence: ; Tel.: +33-3-26-91-80-10
| |
Collapse
|
6
|
Synergistic Antibacterial Effect of Zinc Oxide Nanoparticles and Polymorphonuclear Neutrophils. J Funct Biomater 2022; 13:jfb13020035. [PMID: 35466217 PMCID: PMC9036266 DOI: 10.3390/jfb13020035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Zinc oxide nanoparticles (ZnONPs) are inorganic nano-biomaterials with excellent antimicrobial properties. However, their effects on the anti-infection ability of the innate immune system remains poorly understood. The aim of the present study was to explore the potential immunomodulatory effects of ZnONPs on the innate immune system, represented by polymorphonuclear leukocytes (PMNs), and determine whether they can act synergistically to resist pathogen infections. In vitro experiment showed that ZnONPs not only exhibit obvious antibacterial activity at biocompatible concentrations but also enhance the antibacterial property of PMNs. In vivo experiments demonstrated the antibacterial effect of ZnONPs, accompanied by more infiltration of subcutaneous immune cells. Further ex vivo and in vitro experiments revealed that ZnONPs enhanced the migration of PMNs, promoted their bacterial phagocytosis efficiency, proinflammatory cytokine (TNF-α, IL-1β, and IL-6) expression, and reactive oxygen species (ROS) production. In summary, this study revealed potential synergistic effects of ZnONPs on PMNs to resist pathogen infection and the underlying mechanisms. The findings suggest that attempts should be made to fabricate and apply biomaterials in order to maximize their synergy with the innate immune system, thus promoting the host’s resistance to pathogen invasion.
Collapse
|
7
|
Aoun M, Cai X, Xu B, Lahore GF, Bonner MY, He Y, Bäckdahl L, Holmdahl R. Glycan Activation of Clec4b Induces Reactive Oxygen Species Protecting against Neutrophilia and Arthritis. Antioxidants (Basel) 2021; 11:12. [PMID: 35052516 PMCID: PMC8773064 DOI: 10.3390/antiox11010012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Animal models for complex diseases are needed to position and analyze the function of interacting genes. Previous positional cloning identified Ncf1 and Clec4b to be major regulators of arthritis models in rats. Here, we investigate epistasis between Ncf1 and Clec4b, two major regulators of arthritis in rats. We find that Clec4b and Ncf1 exert an additive effect on arthritis given by their joint ability to regulate neutrophils. Both genes are highly expressed in neutrophils, together regulating neutrophil availability and their capacity to generate reactive oxygen species. Using a glycan array, we identify key ligands of Clec4b and demonstrate that Clec4b-specific stimulation triggers neutrophils into oxidative burst. Our observations highlight Clec4b as an important regulator of neutrophils and demonstrate how epistatic interactions affect the susceptibility to, and severity of, autoimmune arthritis.
Collapse
Affiliation(s)
- Mike Aoun
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
| | - Xiaojie Cai
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Bingze Xu
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
| | - Gonzalo Fernandez Lahore
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
| | - Michael Yi Bonner
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
| | - Yibo He
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
| | - Liselotte Bäckdahl
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
| | - Rikard Holmdahl
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, 171 77 Stockholm, Sweden; (M.A.); (X.C.); (B.X.); (G.F.L.); (M.Y.B.); (Y.H.); (L.B.)
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xibei Hospital, Xi’an 710004, China
| |
Collapse
|
8
|
Kim AR, Bak EJ, Yoo YJ. Distribution of neutrophil and monocyte/macrophage populations induced by the CXCR4 inhibitor AMD3100 in blood and periodontal tissue early after periodontitis induction. J Periodontal Res 2021; 57:332-340. [PMID: 34927238 DOI: 10.1111/jre.12963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/16/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
CXCR4, a CXCL12 receptor, is expressed on epithelial cells, fibroblasts, and inflammatory cells. The CXCR4-CXCL12 interaction is related to the migration of neutrophils and monocytes/macrophages. Periodontitis, an inflammatory disease mainly caused by gram-negative bacteria, is characterized by infiltration of circulating inflammatory cells and alveolar bone (AB) loss. To investigate whether CXCR4 is involved in the distribution of neutrophils and monocytes/macrophages early after periodontitis induction, we examined the effects of AMD3100 (AMD), a CXCR4 antagonist, in ligature-induced periodontitis mice and LPS-injected air pouch mice. The periodontitis study was accomplished in control (C), periodontitis (P), and P + AMD groups. Periodontitis was induced by ligation of the mandibular first molar. AMD was intraperitoneally administered daily beginning the day before ligation until sacrifice on the third day after ligation. The air pouch study was accomplished in C, lipopolysaccharide (LPS), and LPS + AMD groups. Air pouches on mice backs were formed by subcutaneous injection of sterilized air. AMD was administered and then LPS was injected into the air pouch. For the detection of neutrophils and monocytes/macrophages in blood and air pouch exudates, flow cytometry was performed with anti-Ly6G/anti-CD11b antibodies (Abs) and anti-CD115 Ab, respectively. In periodontal tissue, Ly6G+ cells and CD115+ cells were counted by immunohistological analysis. AB loss was estimated by the periodontal ligament area in the furcation. In the periodontitis study, the P group showed higher numbers of Ly6G+ cells and CD115+ cells in blood and periodontal tissue than the C group. The P + AMD group showed a greater number of Ly6G+ cells and CD115+ cells in blood, but not in periodontal tissue compared to the P group. There was no difference in AB loss between the P and P + AMD groups. In the air pouch study, the LPS group had higher levels of Ly6G+ CD11b+ cells and CD115+ cells in both blood and exudates than the C group. The number of these cells in the LPS + AMD group was higher in blood than in the LPS group, but not in the exudates. The CXCR4 antagonist further increased neutrophil and monocyte/macrophage populations in the blood, but did not alter the levels in the periodontal tissue and exudates in mice with periodontitis and LPS-injected air pouches. These results suggest that during inflammatory conditions such as periodontitis, CXCR4 is involved in the distribution of neutrophils and monocytes/macrophages in the blood, but not in inflamed peripheral tissues.
Collapse
Affiliation(s)
- Ae Ri Kim
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea.,Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, South Korea.,BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Eun-Jung Bak
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Yun-Jung Yoo
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| |
Collapse
|
9
|
Dwita LP, Hikmawanti NPE, Yeni, Supandi. Extract, fractions, and ethyl- p-methoxycinnamate isolate from Kaempferia galanga Elicit anti-inflammatory activity by limiting leukotriene B4 (LTB4) production. J Tradit Complement Med 2021; 11:563-569. [PMID: 34765520 PMCID: PMC8572713 DOI: 10.1016/j.jtcme.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 11/29/2022] Open
Abstract
Background and aim Kaempferia galanga, also known as aromatic Ginger (kencur) in Indonesia, has been widely explored and shows potential as an anti-inflammatory agent. However, there has been limited research to show a possible mechanism by which aromatic ginger inhibits lipoxygenase (LOX). Therefore, this study aims to determine the anti-inflammatory activity of aromatic ginger by comparing extract, fractions, and ethyl-p-methoxycinnamate (EPMC) isolate, as well as possible LOX inhibition activity, by reducing the production of leukotriene B4 (LTB4). Experimental procedure Two animal models were used, namely, the carrageenan-induced granuloma air pouch model and the pleurisy model. The test substance was administered 1 h before carrageenan induction, which was performed orally for each animal model. The number of leukocytes and the malondialdehyde (MDA) levels, leukotriene B4 (LTB4) levels, and histology were observed. GC-MS and LC-MS were used for analysis of the chemical compounds in the test samples. Results and conclusion The results of GC-MS analysis showed that aromatic ginger rhizome extract and fractions were dominated by ethyl-trans-p-methoxycinnamate, with the highest level found in the extract. K. galanga showed significant anti-inflammatory activity compared to the control (p < 0.01) in both the granuloma air pouch and pleurisy models. The results of examining the LTB4 concentration showed comparable activity between K. galanga extract, fractions and EMPC isolate, these results were not better than those of zileuton. Overall, this study shows that aromatic ginger extract, fractions and EPMC isolate have anti-inflammatory properties and have the potential to inhibit LOX, thereby reducing LTB4 levels. Comparison of extract, fraction and isolate of Kamperia galanga anti-inflammation activity. Anti-inflammatory activity of Kaempferia galanga in pleurisy model. Extract, fraction and isolate of Kamperia galanga as LOX inhibitor.
Collapse
Key Words
- AA, arachidonic acid
- ARDS, acute respiratory distress syndrome
- Anti-inflammation
- COPD, chronic obstructive pulmonary disease
- COX, cyclooxygenase
- E, ethanol extract
- EAF, ethyl acetate fraction
- EPMC, ethyl-p-methoxycinnamate
- FLAP, 5-lipoxygenase-activating protein
- Granuloma-air pouch
- HF, n-hexane fraction
- IBD, inflammatory bowel disease
- IL, interleukin
- LOX, lipoxygenase
- LTA4H, LTA4 hydrolase
- LTB4, leukotriene B4
- Lipoxygenase
- MDA, malondialdehyde
- PG, prostaglandin
- Pleurisy
- TBA, thiobarbituric acid
- TEP, tetraethoxypropane
- TNF-α, tumor necrosis factor-α
- WF, water fraction
Collapse
Affiliation(s)
- Lusi Putri Dwita
- Faculty of Pharmacy and Science, Universitas Muhammadiyah Prof. DR. HAMKA, Jakarta, Indonesia
| | - Ni Putu Ermi Hikmawanti
- Faculty of Pharmacy and Science, Universitas Muhammadiyah Prof. DR. HAMKA, Jakarta, Indonesia
| | - Yeni
- Faculty of Pharmacy and Science, Universitas Muhammadiyah Prof. DR. HAMKA, Jakarta, Indonesia
| | - Supandi
- Faculty of Pharmacy and Science, Universitas Muhammadiyah Prof. DR. HAMKA, Jakarta, Indonesia
| |
Collapse
|
10
|
Cavalli J, Freitas MA, Gonçalves ECD, Fadanni GP, Santos AA, Raposo NRB, Dutra RC. Chia oil prevents chemical and immune-mediated inflammatory responses in mice: Evidence for the underlying mechanisms. Food Res Int 2021; 149:110703. [PMID: 34600695 DOI: 10.1016/j.foodres.2021.110703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Chia (Salvia hispanica L.) is an herbaceous plant used as omega-3 polyunsaturated fatty acid (ω-3 PUFA) source that presents a range of beneficial effects on human health. Herein, it was used a chia oil containing over than 62% of α-linolenic acid (ALA), a compound widely related to anti-inflammatory actions. Chia oil effect was tested using paw edema and mechanical hyperalgesia induced by carrageenan, and ear edema induced by croton oil, histamine, and capsaicin. Croton oil was used in both preventive and therapeutic treatment schedules of chia oil while histamine and capsaicin were used only in preventive treatment schedule. Chia oil mechanism of action was investigated using nociception and paw edema response induced by intraplantar injection of acidified saline (ASIC activator), PGE2 (prostaglandin pathway), cinnamaldehyde (TRPA1 activator), bradykinin (BK pathway), menthol (TRPM8 activator), and capsaicin (TRPV1 activator). Further, RT-PCR for inflammatory mediators (TRPA1, NF-κB, PPAR-γ, COX-2, IL-6, TNF, FPR2, FAAH, MAGL, and IL-12A) induced by carrageenan, NLRP3 inflammasome activation, and the cell viability were then accessed. Later, chia oil actions were evaluated in the experimental autoimmune encephalomyelitis (EAE), a multiple sclerosis (MS) model. Chia oil showed anti-edematogenic and anti-hyperalgesic effects when administered 1 h before pro-inflammatory stimulus - particularly carrageenan and croton oil. Moreover, chia oil upregulated the mRNA levels of COX-2 and formyl peptide receptor 2 (FPR2) while reduced IL-6 expression in the spinal cord of mice submitted to i.pl. injection of carrageenan. Interestingly, chia oil mediates antinociceptive effects in mice decreasing the nociceptive response induced by acidified saline, PGE2, and cinnamaldehyde, but not by bradykinin, menthol, and capsaicin. On the EAE model, chia oil preventively administered attenuated EAE-induced motor deficits and mechanical hyperalgesia in mice, suggesting a valuable effect of chia oil supplementation in regulating inflammatory responses and some immune functions during immune-mediated inflammatory disorders (IMID). Nonetheless, additional reports will need to assess the effect of chia oil in well-controlled clinical trials performed in MS patients.
Collapse
Affiliation(s)
- Juliana Cavalli
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil
| | - Mariana A Freitas
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil
| | - Elaine C D Gonçalves
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil; Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | - Guilherme P Fadanni
- Center of Innovation and Preclinical Research, 88056-000 Florianópolis, SC, Brazil
| | - Adara A Santos
- Center of Innovation and Preclinical Research, 88056-000 Florianópolis, SC, Brazil
| | - Nádia R B Raposo
- Center for Research and Innovation in Health Sciences (NUPICS), Faculty of Pharmacy, Universidade Federal de Juiz de Fora, 36036-330 Juiz de Fora, MG, Brazil
| | - Rafael C Dutra
- Laboratory of Autoimmunity and Immunopharmacology, Department of Health Sciences, Campus Araranguá, Universidade Federal de Santa Catarina, 88906-072 Araranguá, SC, Brazil; Post-Graduate Program of Neuroscience, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil; Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Center of Biological Sciences, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil.
| |
Collapse
|
11
|
Abstract
The subcutaneous air pouch is an in vivo model that can be used to study the components of acute and chronic inflammation, the resolution of the inflammatory response, the oxidative stress response, and potential therapeutic targets for treating inflammation. Injection of irritants into an air pouch in rats or mice induces an inflammatory response that can be quantified by the volume of exudate produced, the infiltration of cells, and the release of inflammatory mediators. The model presented in this article has been extensively used to identify potential anti-inflammatory drugs. © 2021 Wiley Periodicals LLC. Basic Protocol: Air pouch model in the rat Alternate Protocol: Air pouch model in the mouse.
Collapse
Affiliation(s)
- Jill C Fehrenbacher
- Department of Pharmacology and Toxicology and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kenneth E McCarson
- Kansas Intellectual and Developmental Disabilities Research Center (KIDDRC), Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
12
|
Lenert ME, Avona A, Garner KM, Barron LR, Burton MD. Sensory Neurons, Neuroimmunity, and Pain Modulation by Sex Hormones. Endocrinology 2021; 162:bqab109. [PMID: 34049389 PMCID: PMC8237991 DOI: 10.1210/endocr/bqab109] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Indexed: 12/16/2022]
Abstract
The inclusion of women in preclinical pain studies has become more commonplace in the last decade as the National Institutes of Health (NIH) released its "Sex as a Biological Variable" mandate. Presumably, basic researchers have not had a comprehensive understanding about neuroimmune interactions in half of the population and how hormones play a role in this. To date, we have learned that sex hormones contribute to sexual differentiation of the nervous system and sex differences in behavior throughout the lifespan; however, the cycling of sex hormones does not always explain these differences. Here, we highlight recent advances in our understanding of sex differences and how hormones and immune interactions influence sensory neuron activity to contribute to physiology and pain. Neuroimmune mechanisms may be mediated by different cell types in each sex, as the actions of immune cells are sexually dimorphic. Unfortunately, the majority of studies assessing neuronal contributions to immune function have been limited to males, so it is unclear if the mechanisms are similar in females. Finally, pathways that control cellular metabolism, like nuclear receptors, have been shown to play a regulatory role both in pain and inflammation. Overall, communication between the neuroimmune and endocrine systems modulate pain signaling in a sex-dependent manner, but more research is needed to reveal nuances of these mechanisms.
Collapse
Affiliation(s)
- Melissa E Lenert
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Amanda Avona
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Katherine M Garner
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Luz R Barron
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| |
Collapse
|
13
|
Baliou S, Sofopoulos M, Goulielmaki M, Spandidos DA, Ioannou P, Kyriakopoulos AM, Zoumpourlis V. Bromamine T, a stable active bromine compound, prevents the LPS‑induced inflammatory response. Int J Mol Med 2021; 47:37. [PMID: 33537817 PMCID: PMC7891821 DOI: 10.3892/ijmm.2021.4870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/26/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammation is the most common cause of most acute and chronic debilitating diseases. Towards unveiling novel therapeutic options for patients with such complications, N‑bromotaurine (TauNHBr) has emerged as a potential anti‑inflammatory agent; however, its therapeutic efficacy is hindered due to its relatively poor stability. To address this challenge, the present study focused on examining the effects of a stable active bromine compound, named bromamine T (BAT). The present study examined the protective properties of BAT against lipopolysaccharide (LPS)‑mediated inflammation in vitro, by using LPS‑stimulated murine J774.A1 macrophages (Mφs), as well as in vivo, by using a murine LPS‑mediated air‑pouch model. Additionally, its efficacy was compared with that of taurine, a known potent anti‑inflammatory molecule. In LPS‑stimulated J774A.1 Mφs, BAT and taurine were very effective in reducing the secretion of pro‑inflammatory mediators. The in vitro experiments indicated that LPS‑mediated inflammation was attenuated due to the protective properties of BAT and of taurine, probably through the inhibition of phosphorylated p65 NF‑κB subunit (Ser 536) nuclear translocation. The in vivo experiments also revealed that BAT and taurine inhibited LPS‑mediated inflammation by reducing total cell/polymorphonuclear cell (PMN) infiltration in the air‑pouch and by decreasing pouch wall thickness. The analysis of exudates obtained from pouches highlighted that the inhibitory effects of BAT and taurine on the secretion of pro‑inflammatory cytokines were similar to those observed in vitro. Notably, the effect of BAT at the highest concentration tested was superior to that of taurine at the highest concentration. Taken together, the findings of the present study indicate that BAT prevents the LPS‑induced inflammatory response both in vitro and in vivo.
Collapse
Affiliation(s)
- Stella Baliou
- National Hellenic Research Foundation, 11635 Athens, Greece
| | - Michael Sofopoulos
- Department of Surgical Pathology, Saint Savvas Anticancer Hospital of Athens, 11522 Athens, Greece
| | | | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Petros Ioannou
- Department of Internal Medicine and Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece
| | | | | |
Collapse
|
14
|
Conceição de Souza M, Hubner JT, Gonçalves GM, Ferreira da Costa Fernandes T, Laureano-Melo R, da Silva Côrtes W, Gonçalves L, de Assis da Silva F, Marinho BG. Topical and systemic use of Joannesia princeps vell. LC seed oil in acute pain and inflammation induced by different agents. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113554. [PMID: 33152426 DOI: 10.1016/j.jep.2020.113554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Joannesia princeps (SOJP) has been used in folk medicine as anthelmintic treatment and cutaneous wound healing. AIM OF THE STUDY The purpose of this study is to evaluate the pharmacological activity of seed oil of Joannesia princeps, administered systemically and topically, on acute pain and inflammation. MATERIALS AND METHODS Male swiss mice were treated orally and topically with seed oil of Joannesia princeps in models of acute pain (acetic acid-induced abdominal writhing, formalin-induced licking behaviour and tail flick tests) and acute inflammation (carrageenan- and histamine-induced paw oedema; arachidonic acid-, capsaicin- and croton oil-induced ear oedema and air pouch tests), besides the open field model in the motor performance evaluation. RESULTS Seed oil of Joannesia princeps showed systemic action against acute pain in abdominal writhing test (37% and 56% inhibition in the number of writhes at doses of 30 and 100 mg/kg, respectively) and in the second phase of formalin-induced licking behaviour test (29%, 47 and 52% inhibition in the licking time at doses of 10, 30 and 100 mg/kg, respectively), as well as reducing croton oil-induced ear oedema by 72%, leukocyte recruitment and production of TNF-α and IL-6 in the air pouch tests. In addition, topical administration of SOJP inhibited carrageenan-induced paw oedema by 39% at dose of 500 μg/paw and inhibited histamine-induced oedema by 43 and 52% at doses of 300 and 500 μg/paw, respectively. SOJP also decreased croton oil-induced ear oedema by 67% at dose of 500 μg/paw and arachidonic acid-induced ear oedema by 63% at dose of 500 μg/paw, reducing the production of TNF-α, IL-1β and MIP2 in both. In addition, no adverse effects were observed at doses up to 2000 mg/kg. CONCLUSIONS Seed oil of Joannesia princeps presents antinociceptive and anti-inflammatory actions through its topical and systemic administration, promoted by inhibition of leukocyte recruitment and cytokine production (TNF-α, IL-1β, IL-6 and MIP-2).
Collapse
Affiliation(s)
- Milene Conceição de Souza
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | - Jhones Trindade Hubner
- Department of Chemistry, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | - Gabriela Mastrangelo Gonçalves
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil; Graduate Program in Physiological Sciences, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | | | - Roberto Laureano-Melo
- Behavioral Physiopharmacology Laboratory, Barra Mansa Center University, Barra Mansa, RJ, Brazil.
| | - Wellington da Silva Côrtes
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil; Graduate Program in Physiological Sciences, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | - Lenicio Gonçalves
- Department of Animal Biology, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| | | | - Bruno Guimarães Marinho
- Pharmacology Laboratory, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil; Graduate Program in Physiological Sciences, Institute of Biological and Health Sciences, Rural Federal University of Rio de Janeiro, Seropédica, RJ, Brazil.
| |
Collapse
|
15
|
Huang M, Ye K, Hu T, Liu K, You M, Wang L, Qin H. Silver Nanoparticles Attenuate the Antimicrobial Activity of the Innate Immune System by Inhibiting Neutrophil-Mediated Phagocytosis and Reactive Oxygen Species Production. Int J Nanomedicine 2021; 16:1345-1360. [PMID: 33633450 PMCID: PMC7901559 DOI: 10.2147/ijn.s292482] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/15/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Despite the extensive development of antibacterial biomaterials, there are few reports on the effects of materials on the antibacterial ability of the immune system, and in particular of neutrophils. In this study, we observe differences between the in vivo and in vitro anti-infective efficacies of silver nanoparticles (AgNPs). The present study was designed to further explore the mechanism for this inconsistency using ex vivo models and in vitro experiments. METHODS AgNPs were synthesized using the polyol process and characterized by transmission electron microscopy and X-ray photoelectron spectroscopy. The antibacterial ability of AgNPs and neutrophils was tested by the spread-plate method. The infected air pouch model was prepared to detect the antimicrobial ability of AgNPs in vivo. Furthermore, blood-AgNPs-bacteria co-culture model and reactive oxygen species (ROS) measurement were used to evaluate the effect of AgNPs to neutrophil-mediated phagocytosis and ROS production. RESULTS The antibacterial experiments in vitro showed that AgNPs had superior antibacterial properties in cell compatible concentration. While, AgNPs had no significant antibacterial effect in vivo, and pathological section in AgNPs group indicated less neutrophil infiltration in inflammatory site than S. aureus group. Furthermore, AgNPs were found to reduce the phagocytosis of neutrophils and inhibit their ability to produce ROS and superoxide during ex vivo and in vitro experiments. CONCLUSION This study selects AgNPs as the representative of inorganic nano-biomaterials and reveals the phenomenon and the mechanism underlying the significant AgNPs-induced inhibition of the antibacterial ability of neutrophils, and may have a certain enlightening effect on the development of biomaterials in the future. In the fabrication of antibacterial biomaterials, however, attention should be paid to both cell and immune system safety to make the antibacterial properties of the biomaterials and innate immune system complement each other and jointly promote the host's ability to resist the invasion of pathogenic microorganisms.
Collapse
Affiliation(s)
- Moran Huang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Kai Ye
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Tu Hu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
| | - Kexin Liu
- Department of General Practice, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Mengzhen You
- Department of General Practice, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Lei Wang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Hui Qin
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
16
|
In vitro and in vivo anti-inflammatory activity and chemical composition of Renealmia petasites Gagnep. Inflammopharmacology 2021; 29:451-465. [PMID: 33452968 DOI: 10.1007/s10787-020-00786-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
The study aimed to investigate the chemical composition and the anti-inflammatory activity of the hydroethanolic rhizomes, stems, and leaf extracts of Renealmia petasites using in vitro and in vivo assays. The chemical composition of the extracts was characterized in a linear iron trap mass spectrometer. Total phenolic, flavonoid, and tannin content were determined by spectrophotometry analyses. In vitro anti-inflammatory activity was investigated in lipopolysaccharide-stimulated macrophages evaluating the influence on the production of superoxide anion (O2-), nitric oxide (NO), and the pro-inflammatory cytokines tumor necrosis factor (TNF-α) and interleukin-6 (IL-6). In vivo effects were determined using the air pouch model in which were inoculated carrageenan and thereafter treated with 50 mg/kg of the hydroethanolic extracts of R. petasites. After 4 and 24 h, the cellular influx, protein exudation, cytokines, and nitric oxide were evaluated. Eight compounds were tentatively identified in the R. petasites extracts, suggesting five diarylheptanoids, one flavonoid, and two fatty alcohols. The in vitro results showed that the extracts were capable of blocking free radicals and/or inhibiting their intracellular actions by inhibiting the production of important mediators of the inflammatory process, such as NO, O2-, TNF-α, and IL-6. In vivo, R. petasites significantly decrease the influx of leukocytes, mainly neutrophils, protein exudation, NO, TNF-α, and IL-6 concentration in the air pouch model. The results evidenced that R. petasites can be considered a promising alternative therapy for the treatment and management of osteoarthritis and other inflammatory diseases.
Collapse
|
17
|
Li T, Wang T, Wang L, Liu R, Zhang L, Zhai R, Fu F. Antinociceptive effects of rotigotine-loaded microspheres and its synergistic interactions with analgesics in inflammatory pain in rats. Eur J Pharmacol 2021; 891:173693. [PMID: 33160937 DOI: 10.1016/j.ejphar.2020.173693] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 01/24/2023]
Abstract
Rotigotine-loaded microspheres (RoMS) are sustained-release formulations with prolonged anti-Parkinson's effects. Given that pain is a non-motor symptom of Parkinson's disease, this study investigated the antinociceptive effects of RoMS and their synergistic effects with analgesics on inflammatory pain. A model of inflammatory pain was prepared by intraplantarly injecting male Sprague-Dawley rats with carrageenan. The antinociceptive effects of RoMS, acetaminophen, and tramadol, both alone and in combination, were evaluated using the hind paw withdrawal latency in the hot plate test and Randall-Selitto test. The rotigotine concentrations in serum and tissues were assayed using ultra-performance liquid chromatography-tandem mass spectrometry. Isobolographic analysis was performed to evaluate the nature of the interactions of RoMS with acetaminophen or tramadol. The results showed that hind paw withdrawal latency to thermal and mechanical stimuli was significantly increased on day 3 and 7 after administered RoMS. Rotigotine could be detected in serum and tissues 3 and 7 days after an intramuscular injection of RoMS. However, the rotigotine concentration fell the detection limit of the assay on day 14 after administration. RoMS produced synergistic antinociceptive effects in the inflammatory pain model when RoMS is combined with acetaminophen or tramadol. These findings suggest that RoMS can relieve inflammatory pain in rats. Furthermore, the combination of RoMS with acetaminophen or tramadol produces synergistic antinociception, which may be clinically worthy because combination therapies may reduce the drug doses required for antinociception.
Collapse
Affiliation(s)
- Ting Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, PR China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, PR China
| | - Linlin Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, PR China
| | - Rongxia Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, PR China
| | - Leiming Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, PR China
| | - Rong Zhai
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, PR China
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, 264005, PR China; State Key Laboratory of Long-acting and Targeting Drug Delivery Technologies, Luye Pharma Group Ltd., Yantai, Shandong, 264003, PR China.
| |
Collapse
|
18
|
Meza-Menchaca T, Lizano-Soberón M, Trigos A, Zepeda RC, Medina ME, Galindo-Murillo R. Elucidating Molecular Interactions of Ten Natural Compounds Targeting E6 HPV High Risk Oncoproteins Using Microsecond Molecular Dynamics Simulations. Med Chem 2021; 17:587-600. [PMID: 31995016 DOI: 10.2174/1573406416666200129145733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/03/2019] [Accepted: 12/16/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cervical cancer is a major public health issue worldwide, occurring in the vast majority of cases (85%) in low-income countries. Human papillomavirus (HPV) mainly infects the mucosal epithelium, and a small portion causes over 600,000 cases every year worldwide at various anatomical spots, mainly leading to anogenital and head and neck. INTRODUCTION The E6 oncoprotein encoded by cancer-associated alpha HPV can transform epithelial cells into tumorigenic tissue. Therapy for this infection and blocking of the HPV E6 oncoprotein could be provided with cost-effective and abundant natural products which are an exponentially growing topic in the literature. Finding an active natural compound that readily blocks HPV E6 oncoprotein which could be available for developing countries without expensive extraction processes or costly synthetic pathways is of major interest. METHODS Molecular dynamics simulation was performed using the most up-to-date AMBER protein force field ff14SB and a GPU enabled high performance computing cluster. RESULTS In this research, we present a study of the binding properties between 10 selected natural compounds that are readily available with two variants of the E6 oncoprotein types (HPV-16 and HPV-18) using 10+ microsecond molecular dynamics simulations. CONCLUSION Our results suggest that crocetin, ergosterol peroxide and κ-carrageenan natural products bind strongly to both HPV-16 and HPV-18 and could potentially serve as a scaffolding for further drug development.
Collapse
Affiliation(s)
- Thuluz Meza-Menchaca
- Facultad de Medicina, Laboratorio de Genomica Humana, Universidad Veracruzana. Medicos y Odontologos, Col. Unidad del Bosque, 91010, Xalapa, Veracruz, Mexico
| | - Marcela Lizano-Soberón
- Unidad de Investigacion Biomedica en Cáncer, Instituto Nacional de Cancerologia-Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico, 14080, Ciudad de Mexico, Mexico
| | - Angel Trigos
- Centro de Investigacion en Micologia Aplicada, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | - Rossana C Zepeda
- Centro de Investigaciones Biomedicas, Universidad Veracruzana, Av. Luis Castelazo Ayala, Xalapa-Enriquez, Veracruz 91190, Mexico
| | - Manuel E Medina
- Centro de Investigacion en Micologia Aplicada, Universidad Veracruzana, Xalapa, Veracruz, Mexico
| | - Rodrigo Galindo-Murillo
- Department of Medicinal Chemistry, L.S. Skaggs Pharmacy Institute, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
19
|
Physalin A attenuates inflammation through down-regulating c-Jun NH2 kinase phosphorylation/Activator Protein 1 activation and up-regulating the antioxidant activity. Toxicol Appl Pharmacol 2020; 402:115115. [DOI: 10.1016/j.taap.2020.115115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 11/18/2022]
|
20
|
Kim AR, Kim JH, Choi YH, Jeon YE, Cha JH, Bak EJ, Yoo YJ. The presence of neutrophils causes RANKL expression in periodontal tissue, giving rise to osteoclast formation. J Periodontal Res 2020; 55:868-876. [PMID: 32583887 DOI: 10.1111/jre.12779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/18/2020] [Accepted: 05/30/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUNDS AND OBJECTIVE Increased neutrophil infiltration and osteoclast formation are key characteristics of periodontitis. The effect of these neutrophils on osteoclast formation in periodontitis remains unclear. Therefore, we investigated the effects of neutrophils on osteoclast formation in a neutrophil-deficient mouse model of periodontitis. METHODS Anti-Ly6G antibody (Ab) was used for neutrophil depletion in two mouse models: periodontitis and air pouch. In the periodontitis experiments, mice were divided into PBS-administered control (C), control Ab-administered periodontitis (P), and anti-Ly6G Ab-administered periodontitis (P + Ly6G) groups. Periodontitis was induced by ligature of mandibular first molars. In the air pouch experiments, mice were divided into PBS-administered (C), LPS and control Ab-administered (LPS), and LPS and anti-Ly6G Ab-administered (LPS + Ly6G) groups. Neutrophil migration into air pouches was induced by LPS injection. Flow cytometry was used to examine CD11b+ Ly6G+ neutrophils in the blood of periodontitis mice and CD11b+ Ly6G+ RANKL+ neutrophils in exudates of air pouch mice. In periodontal tissue, Ly6G+ neutrophil and RANKL+ cell numbers in periodontal ligament and alveolar bone areas were estimated using immunohistochemistry, osteoclast numbers were measured using TRAP assay, and alveolar bone loss was determined by H&E staining. RESULTS In blood, CD11b+ Ly6G+ neutrophils were found in greater percentage in the P group than in the C group on days 3 and 7. However, the percentage of neutrophils was lower in the P + Ly6G group than in the C and P groups. In periodontal tissue, the numbers of Ly6G+ neutrophils and RANKL+ cells were lower in the P + Ly6G group than in the P group on day 3. Ly6G+ neutrophil numbers decreased more in the P + Ly6G group than in the P group on day 7, but RANKL+ cell numbers did not decrease in the P + Ly6G group. In exudates, the number of CD11b+ Ly6G+ RANKL+ neutrophils was greater in the LPS group than in the C and LPS + Ly6G groups. On days 3 and 7, the numbers of osteoclasts and alveolar bone loss were greater in periodontal tissue in the P and P + Ly6G groups than in the C group. Interestingly, there were fewer osteoclasts in the P + Ly6G group than in the P group on day 3. CONCLUSION Neutrophil deficiency caused a reduction in numbers of both RANKL+ cells and osteoclasts in periodontitis-induced tissues only on day 3. Furthermore, in the LPS-injected air pouch model, neutrophil deficiency reduced the influx of RANKL+ neutrophils. These findings suggest that the presence of neutrophils induces RANKL expression and could induce osteoclast formation in the early stages of periodontitis.
Collapse
Affiliation(s)
- Ae Ri Kim
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea.,Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, South Korea.,BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Ji-Hye Kim
- Department of Dental Hygiene, Baekseok University, Cheonan, South Korea
| | - Yun Hui Choi
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Yeong-Eui Jeon
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Jeong-Heon Cha
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea.,Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, South Korea.,BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Eun-Jung Bak
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| | - Yun-Jung Yoo
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, South Korea
| |
Collapse
|
21
|
Anti-Inflammatory and Physicochemical Characterization of the Croton Rhamnifolioides Essential Oil Inclusion Complex in β-Cyclodextrin. BIOLOGY 2020; 9:biology9060114. [PMID: 32486128 PMCID: PMC7344496 DOI: 10.3390/biology9060114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/18/2022]
Abstract
Croton rhamnifolioides is used in popular medicine for the treatment of inflammatory diseases. The objective of this study was to characterize and evaluate the anti-inflammatory effect of C. rhamnifolioides essential oil complexed in β-cyclodextrin (COEFC). The physicochemical characterization of the complexes was performed using different physical methods. The anti-inflammatory activity was evaluated in vivo by ear edema, paw edema, cotton pellet-induced granuloma, and vascular permeability by Evans blue extravasation. The mechanism of action was validated by molecular docking of the major constituent into the cyclooxygenase-2 (COX-2 enzyme). All doses of the COEFC reduced acute paw edema induced by carrageenan and dextran, as well as vascular permeability. Our results suggest the lowest effective dose of all samples inhibited the response induced by histamine or arachidonic acid as well as the granuloma formation. The complexation process showed that the pharmacological effects were maintained, however, showing similar results using much lower doses. The results demonstrated an involvement of the inhibition of pathways dependent on eicosanoids and histamine. Complexation of β-cyclodextrin/Essential oil (β-CD/EO) may present an important tool in the study of new compounds for the development of anti-inflammatory drugs.
Collapse
|
22
|
Darlot B, Eaton JRO, Geis-Asteggiante L, Yakala GK, Karuppanan K, Davies G, Robinson CV, Kawamura A, Bhattacharya S. Engineered anti-inflammatory peptides inspired by mapping an evasin-chemokine interaction. J Biol Chem 2020; 295:10926-10939. [PMID: 32471866 PMCID: PMC7415964 DOI: 10.1074/jbc.ra120.014103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/23/2020] [Indexed: 12/27/2022] Open
Abstract
Chemokines mediate leukocyte migration and homeostasis and are key targets in inflammatory diseases including atherosclerosis, cytokine storm, and chronic autoimmune disease. Chemokine redundancy and ensuing network robustness has frustrated therapeutic development. Salivary evasins from ticks bind multiple chemokines to overcome redundancy and are effective in several preclinical disease models. Their clinical development has not progressed because of concerns regarding potential immunogenicity, parenteral delivery, and cost. Peptides mimicking protein activity can overcome the perceived limitations of therapeutic proteins. Here we show that peptides possessing multiple chemokine-binding and anti-inflammatory activities can be developed from the chemokine-binding site of an evasin. We used hydrogen-deuterium exchange MS to map the binding interface of the evasin P672 that physically interacts with C-C motif chemokine ligand (CCL) 8 and synthesized a 16-mer peptide (BK1.1) based on this interface region in evasin P672. Fluorescent polarization and native MS approaches showed that BK1.1 binds CCL8, CCL7, and CCL18 and disrupts CCL8 homodimerization. We show that a BK1.1 derivative, BK1.3, has substantially improved ability to disrupt P672 binding to CCL8, CCL2, and CCL3 in an AlphaScreen assay. Using isothermal titration calorimetry, we show that BK1.3 directly binds CCL8. BK1.3 also has substantially improved ability to inhibit CCL8, CCL7, CCL2, and CCL3 chemotactic function in vitro We show that local as well as systemic administration of BK1.3 potently blocks inflammation in vivo Identification and characterization of the chemokine-binding interface of evasins could thus inspire the development of novel anti-inflammatory peptides that therapeutically target the chemokine network in inflammatory diseases.
Collapse
Affiliation(s)
- Benoit Darlot
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - James R O Eaton
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom.,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lucia Geis-Asteggiante
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Gopala K Yakala
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Kalimuthu Karuppanan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Graham Davies
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Carol V Robinson
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Akane Kawamura
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, Oxford, United Kingdom .,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Shoumo Bhattacharya
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Kim YC, Lee SE, Kim SK, Jang HD, Hwang I, Jin S, Hong EB, Jang KS, Kim HS. Toll-like receptor mediated inflammation requires FASN-dependent MYD88 palmitoylation. Nat Chem Biol 2019; 15:907-916. [PMID: 31427815 DOI: 10.1038/s41589-019-0344-0] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 07/11/2019] [Indexed: 12/24/2022]
Abstract
Toll-like receptor (TLR)/myeloid differentiation primary response protein (MYD88) signaling aggravates sepsis by impairing neutrophil migration to infection sites. However, the role of intracellular fatty acids in TLR/MYD88 signaling is unclear. Here, inhibition of fatty acid synthase by C75 improved neutrophil chemotaxis and increased the survival of mice with sepsis in cecal ligation puncture and lipopolysaccharide-induced septic shock models. C75 specifically blocked TLR/MYD88 signaling in neutrophils. Treatment with GSK2194069 that targets a different domain of fatty acid synthase, did not block TLR signaling or MYD88 palmitoylation. De novo fatty acid synthesis and CD36-mediated exogenous fatty acid incorporation contributed to MYD88 palmitoylation. The binding of IRAK4 to the MYD88 intermediate domain and downstream signal activation required MYD88 palmitoylation at cysteine 113. MYD88 was palmitoylated by ZDHHC6, and ZDHHC6 knockdown decreased MYD88 palmitoylation and TLR/MYD88 activation upon lipopolysaccharide stimulus. Thus, intracellular saturated fatty acid-dependent palmitoylation of MYD88 by ZDHHC6 is a therapeutic target of sepsis.
Collapse
Affiliation(s)
- Young-Chan Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,Korea Research-Driven Hospital, Seoul National University Hospital, Seoul, Korea
| | - Sang Eun Lee
- Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Somi K Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,Korea Research-Driven Hospital, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Duk Jang
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,Korea Research-Driven Hospital, Seoul National University Hospital, Seoul, Korea
| | - Injoo Hwang
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,Korea Research-Driven Hospital, Seoul National University Hospital, Seoul, Korea
| | - Sooryeonhwa Jin
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,Korea Research-Driven Hospital, Seoul National University Hospital, Seoul, Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Eun-Byeol Hong
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea.,Korea Research-Driven Hospital, Seoul National University Hospital, Seoul, Korea
| | - Kyoung-Soon Jang
- Biomedical Omics Center, Korea Basic Science Institute, Cheongju, South Korea
| | - Hyo-Soo Kim
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea. .,Korea Research-Driven Hospital, Seoul National University Hospital, Seoul, Korea. .,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea. .,World Class University Program, Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea.
| |
Collapse
|
24
|
Meng R, Gu L, Lu Y, Zhao K, Wu J, Wang H, Han J, Tang Y, Lu B. High mobility group box 1 enables bacterial lipids to trigger receptor-interacting protein kinase 3 (RIPK3)-mediated necroptosis and apoptosis in mice. J Biol Chem 2019; 294:8872-8884. [PMID: 31000631 DOI: 10.1074/jbc.ra118.007040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/04/2019] [Indexed: 12/14/2022] Open
Abstract
Receptor-interacting protein kinase 3 (RIPK3) is a key regulator of programmed cell death and inflammation during viral infection or sterile tissue injury. Whether and how bacterial infection also activates RIPK3-dependent immune responses remains poorly understood. Here we show that bacterial lipids (lipid IVa or lipid A) form a complex with high mobility group box 1 (HMGB1), released by activated immune cells or damaged tissue during bacterial infection, and that this complex triggers RIPK3- and TIR domain-containing adapter-inducing IFN-β (TRIF)-dependent immune responses. We found that these responses lead to macrophage death, interleukin (IL)-1α release, and IL-1β maturation. In an air-pouch inflammatory infiltration model, genetic deletion of Ripk3, Trif, or IL-1 receptor (Il-1R), or monoclonal antibody-mediated HMGB1 neutralization uniformly attenuated inflammatory responses induced by Gram-negative bacteria that release lipid IVa and lipid A. These findings uncover a previously unrecognized mechanism by which host factors and bacterial components work in concert to orchestrate immune responses.
Collapse
Affiliation(s)
- Ran Meng
- From the Department of Hematology and Key Laboratory of Non-resolving Inflammation and Tumor and.,the Postdoctoral Research Station of Clinical Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province 410000, China
| | - Lan Gu
- From the Department of Hematology and Key Laboratory of Non-resolving Inflammation and Tumor and.,the Key Laboratory of Medical Genetics, School of Biological Science and Technology, Central South University, Changsha, Hunan Province 410000, China
| | - Yanyan Lu
- From the Department of Hematology and Key Laboratory of Non-resolving Inflammation and Tumor and.,the Key Laboratory of Medical Genetics, School of Biological Science and Technology, Central South University, Changsha, Hunan Province 410000, China
| | - Kai Zhao
- From the Department of Hematology and Key Laboratory of Non-resolving Inflammation and Tumor and.,the Key Laboratory of Medical Genetics, School of Biological Science and Technology, Central South University, Changsha, Hunan Province 410000, China
| | - Jianfeng Wu
- the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Haichao Wang
- the Department of Emergency Medicine, North Shore University Hospital, Northwell Health, Manhasset, New York 11030
| | - Jiahuai Han
- the State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Yiting Tang
- From the Department of Hematology and Key Laboratory of Non-resolving Inflammation and Tumor and .,the Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province 410000, China
| | - Ben Lu
- From the Department of Hematology and Key Laboratory of Non-resolving Inflammation and Tumor and .,the Key Laboratory of Medical Genetics, School of Biological Science and Technology, Central South University, Changsha, Hunan Province 410000, China.,the Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan Province 410000, China, and.,the Department of Pathophysiology, School of Basic Medical Science, Jinan University, Guangzhou, Guangdong Province 510632, China
| |
Collapse
|
25
|
Gavel PK, Parmar HS, Tripathi V, Kumar N, Biswas A, Das AK. Investigations of Anti-Inflammatory Activity of a Peptide-Based Hydrogel Using Rat Air Pouch Model. ACS APPLIED MATERIALS & INTERFACES 2019; 11:2849-2859. [PMID: 30589529 DOI: 10.1021/acsami.8b19228] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The growing area of biomaterial sciences has attracted broad attention in recent years in the development of peptide-based biocompatible materials with inherent therapeutic potentials. Here, we developed an Amoc (9-anthracenemethoxycarbonyl)-capped dipeptide-based biocompatible, injectable, thixotropic, and self-healable hydrogel. In vitro cytotoxicity of the hydrogel was investigated with the human embryonic kidney cell (HEK293) line. We observed that the synthesized peptide is noncytotoxic. The hydrogel showed an antibacterial efficacy against Gram-positive and Gram-negative bacteria. In vivo anti-inflammatory activity of the hydrogel was investigated using the rat air pouch model of acute inflammation. The major parameters considered for the anti-inflammatory study were exudate volume, total and differential white blood cell count, tissue histology, and lipid peroxidation assay. These experimental data suggest biocompatibility and potential therapeutic applications of peptide hydrogel in inflammation.
Collapse
Affiliation(s)
- Pramod K Gavel
- Department of Chemistry , Indian Institute of Technology Indore , Indore 453552 , India
| | - Hamendra S Parmar
- School of Biotechnology , Devi Ahilya University , Indore 452001 , India
| | - Versha Tripathi
- School of Biotechnology , Devi Ahilya University , Indore 452001 , India
| | - Narendra Kumar
- School of Biotechnology , Devi Ahilya University , Indore 452001 , India
| | - Ankan Biswas
- Department of Chemistry , Indian Institute of Technology Indore , Indore 453552 , India
| | - Apurba K Das
- Department of Chemistry , Indian Institute of Technology Indore , Indore 453552 , India
| |
Collapse
|
26
|
Non-human papillomaviruses for gene delivery in vitro and in vivo. PLoS One 2018; 13:e0198996. [PMID: 29912929 PMCID: PMC6005490 DOI: 10.1371/journal.pone.0198996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/18/2018] [Indexed: 12/17/2022] Open
Abstract
Papillomavirus capsids are known to have the ability to package DNA plasmids and deliver them both in vitro and in vivo. Of all known papillomavirus types, human papillomaviruses (HPVs) are by far the most intensely studied. Although HPVs work well as gene transfer vectors, their use is limited as most individuals are exposed to this virus either through a HPV vaccination or natural infection. To circumvent these constraints, we produced pseudovirions (PsVs) of ten non-human papillomavirus types and tested their transduction efficiencies in vitro. PsVs based on Macaca fascicularis papillomavirus-11 and Puma concolor papillomavirus-1 were further tested in vivo. Intramuscular transduction by PsVs led to months-long expression of a reporter plasmid, indicating that PsVs have potential as gene delivery vectors.
Collapse
|
27
|
Chelombitko MA, Averina OA, Vasilyeva TV, Pletiushkina OY, Popova EN, Fedorov AV, Chernyak BV, Shishkina VS, Ilinskaya OP. Mitochondria-Targeted Antioxidant SkQ1 (10-(6´-Plastoquinonyl)decyltriphenylphosphonium Bromide) Inhibits Mast Cell Degranulation in vivo and in vitro. BIOCHEMISTRY (MOSCOW) 2018; 82:1493-1503. [PMID: 29486699 DOI: 10.1134/s0006297917120082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The therapeutic effect of mitochondria-targeted antioxidant 10-(6´-plastoquinonyl)decyltriphenylphosphonium bromide (SkQ1) in experimental models of acute inflammation and wound repair has been shown earlier. It was suggested that the antiinflammatory activity of SkQ1 is related to its ability to suppress inflammatory activation of the vascular endothelium and neutrophil migration into tissues. Here, we demonstrated that SkQ1 inhibits activation of mast cells (MCs) followed by their degranulation and histamine release in vivo and in vitro. Intraperitoneal injections of SkQ1 in the mouse air-pouch model reduced the number of leukocytes in the air-pouch cavity and significantly decreased the histamine content in it, as well as suppressing MC degranulation in the air-pouch tissue. The direct effect of SkQ1 on MCs was studied in vitro in the rat basophilic leukemia RBL-2H3 cell line. SkQ1 inhibited induced degranulation of RBL-2H3 cells. These results suggest that mitochondrial reactive oxygen species are involved in the activation of MCs. It is known that MCs play a crucial role in regulation of vascular permeability by secreting histamine. Suppression of MC degranulation by SkQ1 might be a significant factor in the antiinflammatory activity of this mitochondria-targeted antioxidant.
Collapse
Affiliation(s)
- M A Chelombitko
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yuan X, Wang J, Cheng M, Zhang X. Mouse β-defensin-14 for inducing the maturation of dendritic cells. Int Immunopharmacol 2017; 55:133-141. [PMID: 29253819 DOI: 10.1016/j.intimp.2017.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND β-defensins are an excellent antimicrobial peptide against microbial infection in which dendritic cells (DCs) play a crucial role by improving the innate and adaptive immune defense. However, it is unclear whether BDs affect DC maturation. This work aimed to study the effects of mouse β-defensin-14 (MBD-14) on DC maturation. METHODS Via in vitro using mouse bone marrow DCs, the maturation of DCs was evaluated by cell morphological staining, flow cytometry, endocytosis assay, and allogeneic mixed lymphocyte reaction, respectively. And it was also assessed by in vivo establishing a mouse air-pouch model for flow cytometric determination, cytokine analysis, and histological staining. Additionally, CLI-095, an inhibitor of Toll-like receptor-4 (TLR-4), was used to determine whether TLR-4 is possibly involved in DC maturation. RESULTS It was found MBD-14 promoted DCs to form more filopodia and lamellipodia, increased the expression of DC maturation markers (CD40 and MHC-II), decreased their endocytic capacity, and enhanced T-cell proliferation. The analyses of the air-pouch exudates were consistent with the in vitro results of MBD-14 activating DCs. And when CLI-095 was applied, DC maturation was inhibited partly. CONCLUSIONS This work demonstrates that MBD-14 can promote the maturation of DCs in which TLR-4 is possibly involved.
Collapse
Affiliation(s)
- Xiangwei Yuan
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Jiaxing Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Mengqi Cheng
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China.
| |
Collapse
|
29
|
Yuan X, Cao H, Wang J, Tang K, Li B, Zhao Y, Cheng M, Qin H, Liu X, Zhang X. Immunomodulatory Effects of Calcium and Strontium Co-Doped Titanium Oxides on Osteogenesis. Front Immunol 2017; 8:1196. [PMID: 29033930 PMCID: PMC5626827 DOI: 10.3389/fimmu.2017.01196] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/11/2017] [Indexed: 01/07/2023] Open
Abstract
The effects of calcium (Ca) or strontium (Sr) on host osteogenesis and immune responses have been investigated separately. In clinical practice, these two elements may both be present around an orthopedic device, but their potential synergistic effects on osteogenesis and the immune response have not been explored to date. In this work, we investigated the immunomodulatory effects of Ca and Sr co-doped titanium oxides on osteogenesis in vitro using the mouse macrophage cell line RAW 264.7 alone and in co-culture with mouse bone mesenchymal stem cells (BMSCs), and in vivo using a mouse air-pouch model. Coatings containing Ca and Sr at different concentration ratios were fabricated on titanium substrates using micro-arc oxidation and electrochemical treatment. The in vitro and in vivo results demonstrated that the Ca and Sr concentration ratio has a marked influence on macrophage polarization. The coating with a Ca/Sr ratio of 2:1 was superior to those with other Ca and/or Sr concentrations in terms of modulating M2 polarization, which enhanced osteogenic differentiation of mouse BMSCs in co-culture. These findings suggest that the osteoimmunomodulatory effect of a titanium-oxide coating can be enhanced by modulating the concentration ratio of its components.
Collapse
Affiliation(s)
- Xiangwei Yuan
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huiliang Cao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Jiaxing Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kaiwei Tang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Bin Li
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaochao Zhao
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mengqi Cheng
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Qin
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|